++
Dennis Fell, PT, MD; Diane Pitts, PT, DPT; Blair Saale, PT, DPT, NCS; Janet Tankersley PT, DPT PCS, Karen Lunnen PT, EdD
++
*with acknowledgement to the students who assisted with literature searches, summary, and updates of information (listed at the end of this document).
+++
Table of Contents (Alphabetical Listing of Common Neuromuscular Diagnoses – the order in which they are found in this compendium)
++
Acquired Immune Deficiency Syndrome (AIDS) (A) (P)
Amyotrophic Lateral Sclerosis (A)
Angelman Syndrome (P)
Arthrogryposis Multiplex Congenita (P)
Autism Spectrum (Pervasive Developmental Disorder) (P)
Bell's Palsy (A)
Benign Paroxysmal Positional Vertigo (BPPV) (A)
Brachial Plexus Injury (Neonatal Brachial Plexus Palsy) (P)
Cerebellar Disease and Tumor (A) (P)
Cerebral Palsy (P)
Cerebral Tumor (A) (P)
Cerebrovascular Accident (ischemic/hemorrhagic) / Stroke (A) (P)
Cerebrovascular Accident –Arteriovenous Malformation (A) (P)
Charcot-Marie-Tooth (A)
Complex Regional Pain Syndrome (Reflex Sympathetic Dystrophy) (A)
Congenital Limb Deficiency (P)
Conversion Disorder (A) (P)
Delirium (A) (P)
Dementia / Alzheimer's disease (A)
Developmental Coordination Disorder (P)
Down's Syndrome (P)
Dystonia (A)
Encephalitis (A) (P)
Fetal Alcohol Syndrome (P)
Fibromyalgia (A)
Friedreich's Ataxia (A)
Guillain-Barré Syndrome (A)
High Risk Infant / Prematurity (Intraventricular Hemorrhage) (P)
Huntington's Disease (A)
Ischemic Encephalopathy (Near Drowning) (A) (P)
Lyme Disease (A) (P)
Méniere's Disease (A)
Multiple Sclerosis (A)
Muscular Dystrophy (P)
Myasthenia Gravis (A)
Myelominingocele / Spina Bifida (P)
Myotonia (A)
Neurofibromatosis (A) (P)
Normal Pressure Hydrocephalus (A)
Osteogenesis Imperfecta (P)
Parkinson's Disease (A)
Pediatric Infectious Diseases (P)
Peripheral Neuropathy (A)
Poliomyelitis / Post-polio Syndrome (A) (P)
Prader Willi Syndrome (P)
Progressive Bulbar Palsy (A)
Rett's Syndrome (P)
Reyes Syndrome (P)
Seizure Disorder (Epilepsy) (A) (P)
Spinal Cord Tumors (A) (P)
Spinal Muscular Atrophy (P)
Syringomyelia (A)
Transverse Myelitis (A) (P)
Traumatic Brain Injury / Diffuse Axonal Injury (A) (P)
Traumatic Spinal Cord Injury (A) (P)
Trigeminal Neuralgia (A)
Vestibular Hypofunction (Unilateral or Bilateral) (A)
++
(P) denotes primarily Disorder in Pediatrics
++
(A) denotes primarily Adult-onset Disorder
+++
Table of Contents (Listing of Common Neuromuscular Diagnoses Grouped by Categories)
+++
Impaired Oxygen Supply
++
Cerebral Palsy (P)
Cerebrovascular Accident (ischemic/hemorrhagic) / Stroke (A) (P)
Cerebrovascular Accident –Arteriovenous Malformation (A) (P)
Ischemic Encephalopathy (Near Drowning) (A) (P)
++
+++
Spinal Cord Disorders
++
+++
Central Nervous System Neoplasia
++
+++
Degenerative Disorders
++
Amyotrophic Lateral Sclerosis (A)
Dementia / Alzheimer's disease (A)
Friedreich's Ataxia (A)
Huntington's Disease (A)
Multiple Sclerosis (A)
Parkinson's Disease (A)
Progressive Bulbar Palsy (A)
++
+++
Peripheral Nerve Disorders
++
+++
Neuromotor System Disorders
++
Myasthenia Gravis (A)
Myotonia (A)
+++
Infectious/Inflammatory Disorders
++
Acquired Immune Deficiency Syndrome (AIDS) (A) (P)
Encephalitis (A) (P)
Guillain-Barré Syndrome (A)
Lyme Disease (A) (P)
Pediatric Infectious Diseases (P)
Poliomyelitis / Post-polio Syndrome (A) (P)
Reyes Syndrome (P)
+++
Pediatric Genetic Disorders
++
Angelman Syndrome (P)
Down's Syndrome (P)
Muscular Dystrophy (P)
Neurofibromatosis (A) (P)
Osteogenesis Imperfecta (P)
Prader Willi Syndrome (P)
Spinal Muscular Atrophy (P)
+++
Pediatric Developmental Disorders
++
Arthrogryposis Multiplex Congenita (P)
Autism Spectrum (Pervasive Developmental Disorder) (P)
Brachial Plexus Injury (Obstetrical Birth Palsy) (P)
Congenital Limb Deficiency (P)
Developmental Coordination Disorder (P)
Fetal Alcohol Syndrome (P)
High Risk Infant / Prematurity (Intraventricular Hemorrhage) (P)
Myelominingocele / Spina Bifida (P)
++
Cerebellar Disease and Tumor (A) (P)
Complex Regional Pain Syndrome (Reflex Sympathetic Dystrophy) (A)
Conversion Disorder (A) (P)
Delirium (A) (P)
Dystonia (A)
Fibromyalgia (A)
Normal Pressure Hydrocephalus (A)
Rett's Syndrome (P)
Seizure Disorder (Epilepsy) (A) (P)
++
(P) denotes primarily Disorder in Pediatrics
++
(A) denotes primarily Adult-onset Disorder
+++
Organization and Use of this Compendium
++
The full Compendium of Adult and Pediatric Neuromuscular Diagnoses, available in the online resources associated with the book, contains an outline, with reference list, for the common neuromuscular diagnoses – pediatric and adult – that a neurologic therapist will encounter. Each outline summarizes key information for a particular neuromuscular diagnosis, which you likely learned in a previous pathophysiology course. These outlines have a particular emphasis on implications for the patient management process (APTA, 2015), from examination to plan of care / interventions and outcomes.
++
Each outline follows a consistent organization to serve as a resource to help you in making decisions about the examination methods you will chose, the expected degree of recovery, and the patient-centered interventions. For each diagnosis outline, Section I, "Etiology" will summarize known etiologic factors, risk factors, and pathogenesis of the disorder to understand the basis of each disorder. Section II, "Diagnostics" will include a summary of symptoms the patient may report, signs that can be observed or measured during the neurologic examination, and diagnostic tests that may be used to confirm the medical diagnosis. This section will assist in anticipating possible underlying impairments and functional activity limitations that are expected with the diagnosis, therefore focusing the examination and providing guidance for the evaluation. This section will also delineate any diagnosis-specific outcome measures that have been described. Section III, "Prognosis" will specify if the diagnosis is considered progressive or not, describe trends of progression, which should become second nature to the therapist in helping to determine appropriate short-term and long-term goals. This section will also present known prognostic factors, and help to anticipate expected sequelae including secondary body structure impairments and functional limitations. Section IV, "Medical/Surgical Management" will outline the commonly used medical/pharmaceutical interventions, and surgical procedures that may be used to address primary deficits or secondary sequelae such as range-of-motion deficits. Section V, "Key Implications for Therapeutic Management" will specify the potential role of each member of the interprofessional rehabilitation team, general aspects of the most likely aspects of therapy in rehabilitation to be included in the plan of care, and contraindications/precautions for those interventions. This section will also include the most likely "Preferred Practice Pattern(s)" from the "Guide to Physical Therapist Practice"; even though the most recent version of the "Guide" does not include the Preferred Practice Patterns, they are available in an online Guide supplement (APTA, 2017). Section VI, "Consumer and Professional Resources" will list available online resources and specific agencies and associations that provide support to individuals with the diagnosis.
++
While reviewing these diagnosis outlines, the reader can obtain more specific information on examination and intervention by referring to related chapters earlier in the textbook. For example, specific examination tests and measures are described in Section II, Chapter 3,Chapter 4,Chapter 5,Chapter 6,Chapter 7,Chapter 8,Chapter 9,Chapter 10,Chapter 11,Chapter 12,Chapter 13. General principles and therapeutic approaches for intervention are found in Section III, Chapter 14,Chapter 15,Chapter 16,Chapter 17. Specific interventions to address underlying body system impairments are covered in Section IV, Chapter 18,Chapter 19,Chapter 20,Chapter 21,Chapter 22,Chapter 23,Chapter 24,Chapter 25,Chapter 26,Chapter 27,Chapter 28,Chapter 29,Chapter 30,Chapter 31,Chapter 32 and specific interventions to address functional limitations are covered in Section V, Chapter 33,Chapter 34,Chapter 35,Chapter 36,Chapter 37.
+
American Physical Therapy Association (APTA). Guide to Physical Therapist Practice 3.0. 2015: DOI: 10.2522/ptguide3.0_978-1-931369-85-5. Accessed May 20, 2015.
+++
Acquired Immune Deficiency Syndrome (AIDS) – Neurologic Deficits
++
AIDS is the final stage of HIV in which the immune system becomes dysfunctional.
Incidence and Prevalence
Genetic research indicates that the first human contraction occurred in the 1920s in what is now the Democratic Republic of Congo. (Faria, 2014)
Scientists now estimate that the first infection of HIV came from Haiti to the U.S. between 1970-1971 (Worobey, 2016)
Currently, there are 36.9 million people infected with HIV/AIDS worldwide. (Kuznik, 2016)
Approximately 50, 000 new cases are diagnosed each year in the U.S. (Huang, 2016)
Alabama, Florida, Georgia, Louisiana, Mississippi, and North Carolina, South Carolina, Tennessee, and Texas are referred to as "targeted states" for HIV/AIDS due to poverty, population health, and negative health outcomes for HIV-infected individuals. (Reif, 2015)
In the United States, HIV/AIDS infections are highest among males of African American and/or Latin American race. (Center for Disease Control and Prevention, 2015)
Caused by retrovirus human immunodeficiency virus type-1 and type-2. (Hladik, 2008)
HIV-1 and HIV-2 are lentiviruses, which cause chronic infections in mammals (Sharp, 2011)
HIV-1 and HIV-2 are spread through contact with infected bodily fluids, mainly sexual contact, IV drug use particularly when sharing needles, contaminated blood, and from mother to child during birth. (Greene, 2007)
30-40% of women infected with H IV acquire it through sexual intercourse. (Hladik, 2008)
Immunoactivation, the activation of the immune system below the level of chronic inflammation, encourages pathogenesis of the etiological agent HIV-1. (Margolis, 2015)
CD4+ T-cells are the clear target of HIV-1. HIV-1 infects the CD4+ T-cells and then uses these cells to replicate. The natural immune response activates other lymphocytes in the area, bringing in additional uninfected CD4+ T cells as targets for HIV-1 infection (Margolis, 2015), with ultimate reduction of the number of CD4+ T-cells, the bodies main infection-fighting mechanisms.
++
Symptoms
Initial Infection
A small number of individuals will experience flu-like symptoms upon initial infection (fever, chills, rash, and sore throat).
Later Stages
After initial flu-like symptoms resolve, individuals can be asymptomatic for up to 10 years after exposure to HIV.
Signs
Opportunistic Diseases
The manifestation of opportunistic diseases that normally do not infect individuals with normal immune systems, particularly infections and cancer. (Greene, 2007)
The presence of viral induced cancers including Kaposi's sarcoma, Burkitt's lymphoma, primary central nervous system lymphoma, and cervical cancer. (Vogel, 2010)
Wasting syndrome
Compromised Cognition and Dementia
HIV infected individuals experience premature aging in the neocortex, the thalamus, and all regions that are involved in higher-cognition and integration, with risk for compromised cognition and dementia (Pfefferbaum, 2014)
If the patient with AIDS has AIDS dementia they will present with cognitive, motor, and/or behavioral problems. (Valcour, 2004)
Distal sensory polyneuropathy
The most common neurological complication associate with AIDS. (Gonzalez-Duarte, 2008)
May occur once AIDS is confirmed (Gonzalez-Duarte, 2008)
Tests
A recent study found the "test window period" range for HIV infection to be 5.9 to 24.8 days after introduction – meaning this is the amount of time it takes for the doctor and patient to be 100% sure of infection. (Delaney, 2017)
Hemotologic testing
Used to determine CD4 cell levels. (Williams, 2000)
An individual is considered to have AIDS if CD4 levels are below 200 cells/mm3 and are infected with the HIV virus. (Greene, 2007)
In developing countries, testing for CD4 cell count is expensive and sometimes impossible to access. In those cases, serum albumin levels directly correlate with CD4 cell counts in immunosuppressed individuals. (Rajasekhar, 2016)
Nucleic acid testing
Used if a person has had a recent exposure to the virus and needs a definite positive or negative diagnosis.
It can detect the virus early in the first stages of infection, but it is not used as a screening tool because of the high cost.
Viral load testing
Rapid assays are now available that allow detection of HIV infection using home testing kits.
These provide a simple yes-no result format and use saliva to detect specific antibodies in less than 30 minutes. These tests are available in most developed countries; however, there are limitations to these assays. Development of assays as a simple packaged test that only needs a cell phone to provide test results may enable developing countries with little infrastructure to accurately test for HIV. (Ocwieja, 2015).
Subjective memory complaints (SMC) have been found to be a reliable marker in HIV-positive adults for actual cognitive deficits. (Kamkwalala, 2016)
++
There is currently no effective vaccine for HIV/AIDS. (Greene, 2007)
AIDS is always fatal (Vogel, 2010)
Antiretroviral Pre-Exposure Prophylaxis (PrEP) prior to HIV exposure has been shown to prevent HIV/AIDS contraction in high-risk populations. This relatively new treatment requires patients to take daily, oral doses of one or two drugs continually while their lifestyle involves risky behaviors. (Okwundu, 2012)
AIDS-related deaths have declined by 42% since peaking in 2004. (UNAIDS, 2016)
Effects of Antiretroviral Therapy
With highly active antiretroviral therapy treatment (HAART) life expectancy is greatly increased. A healthy 25-year-old infected with HIV and on HAART therapy can expect to live to approximately 77. (Vogel, 2010)
Mortality has increased by 50% among adolescents with HIV. This increase occurred from 2005 to 2012 and is associated with lack of adherence to antiretroviral therapy (ART). The importance of adhering to ART cannot be overstated. (Denison, 2015)
Those with HIV/AIDS not on a HAART treatment cycle show a five-time great incidence of AIDS related complications than those on a HAART treatment cycle. (Cain, 2009)
HIV-infected individuals who are homeless or have unstable housing are least likely to adhere to antiretroviral therapy and in result have poorer prognosis. (Aidala, 2016) E
Individuals with AIDS show a higher incidence of dementia, CVA, and decreased neuroglial function. (Valcour, 2004)
Individuals with neurological problems caused by cerebral toxoplasmosis and a CD4 level <50 cells/mm3 have an average life span of 10 months after diagnosis of cerebral toxoplasmosis. (Laing, 1996)
+++
IV. Medical management
++
+++
V. Implications for therapeutic management
++
Preferred practice patterns (APTA, 2017):
4C: Impaired muscle performance
5A: Primary prevention/risk reduction for loss of balance and falling
5E: Impaired motor function and sensory integrity associated with progressive disorders of the CNS
5G: Impaired motor function and sensory integrity associated with acute or chronic polyneuropathies
6A: Primary prevention/risk reduction for cardiovascular/pulmonary disorders (decreased functional work capacity and maximum aerobic capacity)
6B: Impaired aerobic capacity/endurance associated with deconditioning
6C: Impaired ventilation, respiration/gas exchange and aerobic capacity/endurance associated with deconditioning
6D: Impaired aerobic capacity/endurance associated with cardiovascular pump dysfunction or failure.
Treatment for weakness and decreased cardiopulmonary function
Resistance training, and resistance training and aerobic training are considered to have a beneficial effect on cardiopulmonary function, body weight, and strength without affecting immune status. (O'Brien, 2009)
In one case study, a 12-week manual therapy and exercise program demonstrated total elimination of pain during exertion as well as a total elimination of shortness of breath. Patient also had an increase in CD4+ cell count and showed significant improvements in nearly all categories of quality of life. (Pullen, 2014)
Treatment for peripheral neuropathy
Night splints are shown to decrease pain scores in AIDS patients with peripheral neuropathy. (Sandoval, 2010)
Consistent progressive resistive exercise has been shown to improve quality of life for patients with distal symmetrical polyneuropathy associated with HIV/AIDS antiretroviral therapy. (Mkandla, 2016)
Prevention of the spread of HIV
Physical therapists are at low risk for infection when working with a patient with AIDS, unless performing wound care or debridement, in which case they should don PPE. (U.S. Public Health Service, 2001)
Education:
Education should be centered on reducing sexual risk factors: especially not having sex, being monogamous, and each person knowing self and partner's status. (AIDS.gov)
Other members of the health care team need to provide information about medication and possible side effects, test results, any procedures being performed, medical treatments, and symptom management. (AIDS.gov)
Further education on the importance of adhering to ART should be emphasized to adolescents living with HIV. Several factors have been shown to help improve ART adherence among this population: using a cell phone's alarm clock, attending clinic-sponsored youth groups, and a desire to maintain a healthy lifestyle. (Denison, 2015)
Five important factors for exercise with HIV/AIDS (Li, 2017):
Group exercise for social interaction
Exercise, but also consider the social/self-management aspect and offer support sessions
Integrate with other chronic illnesses to decrease stigma
Flexible structure due to unpredictability of HIV/AIDS
Make sure the program is affordable.
+++
VI. Consumer and Professional Resources
++
For information on the HIV virus:
AIDS.gov
U.S. Centers for Disease Control and Prevention
AIDS Info – Facts, Education, and Next Steps after Infection at
For information about AIDS support groups:
For Information on Camps/Retreats:
For information about AIDS organizations:
A website with a list of AIDS organizations
+
Aidala
A, Wilson
M, Shubert
V, Gogolishvili
D, Globerman
J, Rueda
S, Bozack
A, Caban
M, Rourke
S. Housing status, medical care, and health outcomes among people living with HIV/AIDS: A systematic review. Am J Public Health. 2016;106(1):1–23.
+
Cain
L, Cole
S, Greenland
S, Brown
T, Chmeil
J, Kingsley
L, Detels
R. Effect of highly active antiretroviral therapy on incident AIDS using calendar period as an instrumental variable. Am J Epidemiol. 2009;169(9):1124–1132.
+
Caskey
M, Schoofs
T, Gruell
H, Settler
A, Karagounis
T, Kreider
EF, Murrell
B, Pfeifer
N, Nogueira
L, Oliveira
TY, Learn
GH, Cohen
YZ, Lehmann
C, Gillor
D, Shimeliovich
I, Unson-O'Brien
C, Weiland
D, Robles
A, Kummerle
T, Wyen
C, Levin
R, Witmer-Pack
M, Eren
K, Ignacio
C, Kiss
S, West
AP, Mouquet
H, Zingman
BS, Gulick
RM, Keler
T, Bjorkman
PJ, Seaman
MS, Hahn
BH, Fatkenheuer
G, Schlesinger
SJ, Nussenzweig
MC, Klein
F. Antibody 10-1074 suppresses viremia in HIV-1-infected individuals. Nat Med. 2017. DOI: 10.1038/nm.4268.
+
Cohen
M, Chen
Y, McCauley
M,
et al. Prevention of HIV-1 infection with early antiretroviral therapy. N Engl J Med. 2011; 365:493–505.
+
Delaney
KP, Hanson
DL, Masciotra
S, Ethridge
SF, Wesolowski
L, Owen
SM. Time until emergence of HIV test reactivity following infection with HIV-1:Implications for interpreting test results and retesting after exposure. Clin Infect Dis. 2017;64(1):53–59.
+
Denison
JA, Branda
H, Dennis
A, Packer
C, Nyambe
N, Stalter
R, Mwansa
J, Katayamoyo
P, McCarraher
D. "The sky is the limit": adhering to antiretroviral therapy and HIV self-management from perspectives of adolescents living with HIV and their adult caregivers. J Int AIDS Soc. 2015; 18:19358. doi: 10.7448/IAS.18.1.19358.
+
Dubé
K, Henderson
GE, Margolis
DM. Framing expectations in early HIV cure research. Trends Microbiol. 2014; 22(10):547–549.
+
Faria
NR, Rambaut
A, Suchard
MA, Baele
G, Bedford
T, Ward
MJ, Tatem
AJ, Sousa
JD, Arinaminpathy
N, Pepin
J, Posada
D, Peeters
M, Pybus
OG, Lemey
P. The early spread and epidemic ignition of HIV-1 in human populations. Science. 2014;346(6205):56–61.
+
Gonzalez-Duarte
A, Robinson-Papp
J, Simpson
DM. Diagnosis and management of HIV-associated neuropathy. Neurol Clin. 2008; 26(3):821–832.
+
Green
W. A history of AIDS: Looking back to see ahead. Eur J Immunol. 2007; 37: 94–102.
+
Hladik
F, McElrath
J. Setting the stage: Host invasion by HIV. Nature Review Immunology. 2008; 8: 447–457.
+
Huang
MB, Ye
L, Liang,
BY, Ning
CY, Roth
WW, Jiang
JJ, Huang
JG, Zhou
B, Zang
N, Powell
MD, Liang
H, Bond
VC. Characterizing the HIV/AIDS epidemic in the united states and china. Int J Environ Res Public Health. 2015;13(1):30.
+
Kamkwalala
A, Hulgan
T, Newhouse
P. Subjective memory complaints are associated with poorer cognitive performance in adults with HIV. AIDS Care. 2016. DOI: 10.1080/09540121.2016.1248348
+
Kuznik
A, Iliyasu
G, Habib
A, Musa
BM, Kambugu
A, Lamorde
M. Initiation of antiretroviral therapy based on the 2015 WHO guidelines. AIDS. 2016.30(18):2865–2873.
+
Laing
RB, Flegg
PJ, Brettle
RP, Leen
CL, Burns
SM. Clinical features, outcome and survival from cerebral toxoplasmosis in Edinburgh AIDS patients. Int J STD AIDS. 1996;7(4):258–264.
+
Li
A, McCabe
T, Silverstein
E, Dragan
S, Salbach
NM, Zobeiry
M, Beldick
S, Godi
C, O'Brien
KK. Community-based exercise in the context of HIV. J Int Assoc Provid AIDS Care. 2017. DOI: 10.1177/2325957416686836
+
Lundgren,
JD, Babiker,
AG, Gordin,
F. Initiation of antiretroviral therapy in early asymptomatic HIV infection. N Engl J Med. 2015;373(9):795–807.
+
Margolis
L. Immunoactivation at the crossroads of human diseases. Am J Med. 2015. doi: 10.1016/j.amjmed.2014.12.026.
+
Mkandla,
K, Myezwa,
H, Musenge,
E. The effects of progressive-resisted exercises on muscle strength and health-related quality of life in persons with HIV-related poly-neuropathy in Zimbabwe. AIDS Care. 2016:1–5. Epub ahead of print. doi: 10.1080/09540121.2015.1125418
+
O'Brien
K, Nixon
S, Glazier
R, Tynan
A. Progressive resistive exercise interventions for adults living with HIV/AIDS. The Cochrane Collaboration. 2009; 1:1–27.
+
Ocwieja
KE, Sherrill-Mix
S, Liu
C, Song
J, Bau
H, Bushman
FD. A reverse transcription loop-mediated isothermal amplification assay optimized to detect multiple HIV subtypes. PLoS ONE. 2015;10(2):e0117852. doi:10.1371/journal.pone.0117852
+
Okwundu
CI, Uthman
OA, Okoromah
CAN. Antiretroviral pre-exposure prophylaxis (PrEP) for preventing HIV in high-risk individuals. Cochrane Database of Systematic Reviews 2012, Issue 7. Art. No.: CD007189. DOI: 10.1002/14651858.CD007189.pub3.
+
Pfefferbaum
A, Rogosa
DA, Rosenbloom
MJ, Chu
W, Sassoon
SA, Kemper
CA, Deresinski
S, Rohlfing
T, Zahr
NM, Sullivan
EV. Accelerated aging of selective brain structures in HIV infection: A controlled, longitudinal MRI study. Neurobiol Aging. 2014;35(7):1755–1768.
+
Pullen
SD, Chigbo
NN, Nwigwe
EC, Chukwuka
CJ, Amah
CC, Idu
SC. Physiotherapy intervention as a complementary treatment for people with HIV/AIDS. HIV/AIDS-Research and Palliative Care. 2014;6:99–107.
+
Rajasekhar
PV, Kumar
Akhilesh. Role of serum albumin level compared to CD4+ cell count as a marker of immunosuppression in HIV/AIDS patients. Indian Journal of Applied Research. 2016;6(11)244–246.
+
Reif
S, Pence
BW, Hall
I, Hu
X, Whetten
K, Wilson
E. HIV diagnoses, prevalence and outcomes in nine southern states. J Community Health. 2015; 40(4): 642–651.
+
Sandoval
R, Runft
B, Roddey
T. Pilot study: does lower extremity night splinting assist in the management of painful peripheral neuropathy in the HIV/AIDS population? J Int Assoc Physicians AIDS Care. 2010;9(6):368–381.
+
Sharp
P, Hahn
B. Origins of HIV and the AIDS pandemic. Cold Spring Harbor Perspectives in Medicine. 2011; 1: 1–22.
+
U.S. Public Health Service: Updated guidelines for the prevention of HIV and hepatitis virus transmission, and the management of occupational exposures to HIV, BHV, and HCV. 2001;1–67
+
Valcour
V, Shikuma
C, Watters
M, Sacktor
N. Cognitive impairment in older HIV-1-seropositive individuals: prevalence and potential mechanisms. AIDS. 2004;1(18): S79–S86.
+
Vogel
M, Schwarze-Zander
C, Wasmuth
J
et al.. The treatment of patients with HIV. Journal of Medicine. 2010; 107(28-29)507–516.
+
Worobey
M, Watts
TD, McKay
RA, Suchard
MA, Granade
T, Teuwen
DE, Koblin
BA, Heneine
W, Lemey
P, Jaffe
HW. 1970s and 'patient 0' HIV-1 genomes illuminate early HIV/AIDS history in north America. Nature. 2016;539(7627):98–101.
+++
Amyotrophic Lateral Sclerosis (Lou Gehrig's Disease)
++
Incidence and Prevalence
2-3/100,000 per year in developed countries, worldwide rates unavailable due to lack of data in developing regions (Hersi, 2016; Wang, 2016))
Cost to society in the United States is estimated to be $256 million to $433 million (Chen, 2016)
More common in men than women (ratio 1.5:1) (Wang, 2016)
There are some sex and age-related factors: "respiratory onset disease tends to be more prevalent in men, and a predilection for isolated corticobulbar disease has been noted in elderly women"(Turner, 2013).
Ages between 55-65; distinctly uncommon before 20 (Ferguson, 2007; NINDS 2011)
Mortality lowest in population of mixed ancestry (Zaldivar, 2009)
Established risk factors include: male sex, older age, genetics, family history, occupational and environmental factors (exposure to chemicals, heavy metals, and pesticides) and head trauma" (Hersi, 2016)
Other proposed factors include: Mechanical and electrical traumas; smoking; heavy physical activity; some professions such as welding, farming and animal breeding; and previous viral infections (Mitchell, 2000; Armon, 2003; Ingre, 2015; Wang, 2016); professional sports involvement, decreased body mass index, lower educational attainment, and repetitive/strenuous work/occupations (Wang, 2016)
ALS is now thought to represent a heterogeneous group of diseases that share clinical features, rather than a single disease (Marangi, 2015).
ALS is characterized by progressive degeneration of upper motor neurons in the brain with UMN symptoms, and lower motor neurons in the brainstem (cranial nerve nuclei) and spinal cord (alpha motor neurons) with LMN symptoms (Gordon, 2013; Wang, 2016).
More than 25 genes linked to ALS have been identified (Marangi, 2015).
"70% of all monogenic cases of ALS have been reported to result from mutations in three genes: C9orf72, SOD1, and TARDBP." (Hersi, 2016)
Familial ALS (fALS): genetic mutation of SOD1 (Brown, 2005) on chromosome 21, which produces free radicals; 10-20% of all ALS cases (Benatar, 2009).
Requires only one parent to carry the gene (NINDS, 2011)
20% of fALS caused by mutation of SOD1 gene (Turner, 2013), while 50% of fALS is thought to be linked to a mutation to the recently discovered C9orf72 gene (Robberecht , 2013)
fALS is inherited autosomal dominant; X-linked/recessive is rare for fALS (Robberecht, 2013)
Sporadic ALS (SALS): No known cause, accounts for 90% of all ALS cases (Brown, 1997).
In addition to genetic factors; age, tobacco use, and athleticism may contribute to sporadic ALS, but important etiologies are unidentified for most patients (Gordon, 2013).
Theories of ALS causes:
Increased glutamate in CSF causing excitotoxicity, autoimmune disorder, or environmental causes (Brown, 2005) such as cigarette smoking (Gallo, 2009)
Mutations and pathology associated with the TDP-43 gene and protein now thought to be more common than SOD1 mutations in familial and sporadic ALS (Rothstein, 2009)
Recently linked to an expansion of an intronic hexanucleotide repeat gene, C9orf72, providing evidence that ALS and frontotemporal dementia are manifestations of a clinicopathological spectrum (Turner, 2013)
"The apparent RNA binding function of the C9orf72 expansion adds to known manifestations in other key RNA processing and proteosomal genes linked to ALS: TARDP, FUS, and UBQLN2; these reinforce the concept that ALS pathogenesis involves multiple pathways (Turner, 2013).
"SOD1 A4V variant produces a rapidly progressive LMN syndrome" (Turner, 2013).
"FUS mutations are linked to a juvenile onset with mostly LMN syndrome" (Turner, 2013).
"C9orf72 hexanucleotide repeat expansion gene is associated with a younger age of onset, shortened survival, increased rates of cognitive and behavioral impairment, and a strong family history of neurodegenerative disease" (Turner, 2013).
Pathologic mechanisms involve: "mitochondrial dysfunction, protein aggregation, excitotoxicity, and oxidative stress, with consequent loss NMJ integrity, retrograde axonal degeneration, and motor neuronal cell death" (Bucchia, 2015); "loss of neurotrophic factors, inflammatory signaling, endoplasmic reticulum dysfunction, and protein misfolding" (Chen, 2016)
++
"ALS manifests as an insidious, inexorable decline in motor function, with progressively compromised strength, coordination, gait, and respiratory function, leading to death within an average of 3 to 5 years from diagnosis." (Chen, 2016)
Signs and Symptoms
Vary with each patient
Muscle weakness, without sensory abnormalities, in one or more limbs (most common initial symptom) (Zoccolella, 2006)
Loss of functional motor units is rapidly progressive, especially in the early stages of the disease and may begin as early as 18 months before clinical weakness is apparent. (Carvalho, 2016)
Lower motor neuron (LMN) involvement, often the first evidence of the disease, includes insidiously developing asymmetrical weakness, usually in the distal aspect of one limb and progressing to weakness of contiguous muscles.
Focal muscle weakness symptoms may start either distally or proximally in the upper limbs and lower limbs. (Wijesekera, 2009)
ALS begins in the limbs, usually in the arms, in about two-thirds of patients. The first symptoms are most often unilateral and focal (Gordon, 2013).
Upper Motor Neuron (UMN) symptoms can include spasticity (loss of inhibition) with impaired dexterity, poor motor control and weakness.
Gradually, spasticity may develop in the weakened atrophic limbs, affecting manual dexterity and gait. During late stages of the disease patients may develop 'flexor spasms', which are involuntary spasms occurring due to excess activation of the flexor arc in a spastic limb. (Wijesekera, 2009)
Difficulty with speech, projection, and breathing (ALSA.ORG, 2007)
On examining the cranial nerves, the jaw jerk may be brisk, especially in bulbar-onset disease. An upper motor neuron type facial weakness affects the lower half of the face causing difficulty with lip seal and blowing cheeks, but often varying degrees of UMN and LMN facial weakness co-exist. (Wijesekera, 2009)
Twitching or muscle cramps, especially in hands and feet
Sensation including temperature, pain, taste, hearing, vision, and olfaction is usually spared in ALS. Also spared are muscles of sphincters including both smooth and striated and muscles of ocular movement. (Wang, 2016)
"Cognition, eye movement, autonomic, bowel, bladder, and sexual function usually remain intact" (Bello-Haas, 1998).
Cognitive impairments may be present in 50% of patients with ALS (Ringholz, 2005), including dementia and impaired executive dysfunction (Wang, 2016)
Diagnosis: (ALSA.ORG, 2009)
No specific test currently available to diagnose ALS (Wang, 2016), and particularly because of rapid disease progression and lack of study participants in early stage of ALS. (Huynh, 2016)
History, physical, and appropriate neurological examinations to ascertain clinical finding that may suggest suspected, possible, probable or definite ALS.
Electrophysiological examinations to ascertain findings which confirm LMN degeneration in clinically involved regions, identify LMN degeneration in clinically uninvolved regions and exclude other disorders
Muscle ultrasound can detect loss of muscle volume and can show changes in muscle echo which could be used in early diagnosis of ALS. (Carvalho, 2016)
Combining threshold tracking TMS and FDG-PET imaging have the highest sensitivity and specificity for identifying UMN abnormalities. (Huynh, 2016)
Neuroimaging examinations to ascertain findings which may exclude other disease processes (ALSA 2010)
Clinical laboratory examinations, determined by clinical judgment, to ascertain possible ALS related syndromes
Diagnosis typically made clinically when unusual clinical signs are seen and using differential diagnoses. Findings such as both UMN and LMN signs in spinal and bulbar voluntary muscles (Wang, 2016)
Neuropathologic examinations, where appropriate, to ascertain findings which may confirm or exclude sporadic ALS, coexistent sporadic ALS, ALS-related syndromes, or ALS variants
Repetition of clinical and electrophysiological examinations at least six months apart to ascertain evidence of progression.
"The current clinical standard for diagnosis of ALS is the Revised El Escorial World Federation of Neurology Criteria." (Ferguson, 2007)
"Clinical evaluation and Awaji criteria, which are based on innervation and denervation, have been established as a useful diagnostic aid" (Turner, 2013)
The Awajii criteria allow earlier diagnosis and are clinically significant
Awaji criteria Diagnostic Classification (Costa, 2012):
"Clinically definite ALS is defined by clinical or electrophysiological evidence by the presence of LMN as well as UMN signs in the bulbar region and at least 2 spinal regions or the presence of LMN and UMN signs in 3 spinal regions."
"Clinically probable ALS is defined on clinical or electrophysiological evidence by LMN and UMN signs in at least 2 regions with some UMN signs necessarily rostral to the LMN signs."
"Clinically possible ALS is defined when clinical or electrophysiological signs of UMN and LMN dysfunction are found in only 1 region or UMN signs are found alone in >2 regions or LMN signs are found rostral to UMN signs."
Clinical diagnosis, defined by progressive signs and symptoms of both upper and lower motor neuron dysfunction, is confirmed by electromyography (Gordon, 2013)
Requirements to be diagnosed with ALS:
LMN signs by clinical, electrophysiological, or neuropathological examination
UMN signs by clinical observation (+ Babinski found in 30-50% of patients) (Ferguson, 2007)
Progression of the disease within a body region or to another body region
Absence of electrophysiological and pathological evidence of other diseases affecting UMN and LMN
With the exception of a genetic test for FALS, there is no one specific diagnostic test used for ALS (ALSA 2010; Pradham, 2006)
Presymptomatic individuals have been shown to have changes in LMN as well as the "development of cortical hyperexcitability within a year of symptom onset" (Turner, 2013).
++
+++
IV. Medical/Surgical Treatment
++
There is no cure for ALS
Research for prospective pharmacological treatment limited because mouse studies do not correlate well with human studies because onset of the symptoms occurs "long before an ALS diagnosis is confirmed so pathogenesis has progressed too far to have any therapeutic effect." (Browne, 2016; Kumar, 2016)
Pharmaceutical Treatment
Vitamin D
Shown to influence multiple aspects of Amyotrophic Lateral Sclerosis and can improve compromised human muscular ability, increase muscle size, and ATP production. Vitamin D can also reduce the expression of biomarkers associated with oxidative stress and inflammation in patients with MS, RA, CHF, PD, and Alzheimer's, all of which share common pathophysiology's with ALS (Gianforcaro, 2014).
Anticonvulsant medication, benzodiazepams, lorazepam, and narcotics for muscle cramps, spasticity, fasciculations, & pain treatment (respectively)
Antidepressants and anti-anxiety medication
Palliative Care (Hardiman, 2010):
Dyspnea: treated with opiates or benzodiazepines
Morphine:2.5mg: administered 4-6/day followed by morphine if necessary
Nocturnal Symptom Control and Anxiety: Midazolam(2.5-5mg) or diazepam
Pain management: opiates
Terminal restlessness and confusion due to hypercapnia: Neuroleptics
Cell-based therapies aimed at modulating the neuroinflammatory response in ALS may help arrest the progressive and devastating nature of this disease (Hooten, 2015).
Dysphagia and Dysarthria treatment:
Respiratory weakness treatment:
Yearly influenza vaccine, supplemental O2, noninvasive positive pressure ventilation oral/pulmonary secretion management, tracheostomy
Non-invasive ventilation (NIV) has been shown to increase quality of life and prolong survival in ALS patients with respiratory failure. (Radunovic, 2013)
Surgical Techniques:
The decision ultimately faced by all patients (with extensively progressed ALS) is whether to elect to undergo a tracheostomy for long-term mechanical ventilation. That choice can be postponed by the use of noninvasive positive-pressure ventilation, which relieves symptoms and prolongs life. (Rowland, 2001)
Current strategies include: "directing treatment against inflammation factors, kinases, and key genes. Stem cell transplantation is promising due to its ability to reduce inflammation and replace micro- environmental cells, such as astrocytes and NSC's" (Bucchia, 2015)
New studies are showing in treating ALS by injecting mesenchymal stem cells into the spinal cord and slowing the linear decline of forced vital capacity and of the ALS-FRS score. (Mazzini, 2008)
New stem cell research shows potential for induced pluripotent stem (iPS) cells for motor neuron survival by dedifferentiating adult fibroblasts. The iPS cells have many possibilities but currently show the need for immunosuppression in ALS patients, which would raise costs. (Chen, 2016)
Ongoing research for treatment of ALS include: gene therapy, immunotherapy, and vaccines (Kumar, 2016)
+++
V. Therapeutic Management
++
Practice Pattern
Team Approach to treatment includes:
Speech Therapist, Physical Therapist, Nutritionist, Occupational Therapist (Mayo Clinic, 2012)
The primary goal of all health care professionals involved with treating patients with ALS is maintaining patients' quality of life and their independence in functional mobility and ADLs. (Lewis, 2007). There is no current cure for ALS. (John Hopkins University, 2011)
Restorative therapy is used to decrease impairments and improve functional status.
Compensatory therapy is used to modify activities, tasks, and environment.
Preventative therapy is used to increase ROM, strength, and aerobic capacity.
Re-evaluations are crucial with ALS patients to continually update the patient's status and adjust the plan of care accordingly.
Muscle cramps and spasticity treatment:
Massage and stretching for cramps; moderate exercise for spasticity (Ashworth, 2004), PROM, and botulinum toxin injections (Lewis, 2007)
Intrathecal Baclofen via ITB pump is an effective and safe treatment modality for relief of spasticity-related pain, with no postoperative neurologic morbidity/mortality (McClelland, 2007).
Respiratory weakness treatment:
Education, positioning and strategies, breathing exercises, airway clearance techniques
Inspiratory muscle training (IMT) is shown to have short term effects in ALS patients with respiratory insufficiency. (Macpherson, 2016)
Lung volume recruitment training (LVRT) has been found to increase survival and quality of life in ALS due to its long term effects on pulmonary function. (Macpherson, 2016)
Mechanical Ventilation: Invasive procedure that can be undertaken willingly and in emergency situations. Patients should be made aware of emergency situations and a plan should be made to avoid those situations at all costs (Hardiman, 2010).
Non-invasive ventilation is an important development and can prolong life up to 9 months (Hardiman, 2010)
Muscle weakness treatment:
Exercise to maintain strength, appropriate use of splints, slings, orthoses, etc.
Exercises to promote strength, range of motion, balance and endurance may dominate the early therapy program. (Walling, 1999)
Carefully evaluate each individual with ALS every few months to gather adequate information to safely adjust strengthening and conditioning exercise. Approach exercise programs with caution for individuals in later stage ALS who have low respiratory capacity and/or poor functional scores (Lui, 2009)
Decreased mobility and ADL function:
Education for transfers and energy saving techniques, appropriate adaptive equipment, proper wheelchair prescription, possible home modifications
In addition to carrying out the therapy program, physical therapists can help patients and families select and use equipment ranging from simple splints and neck supports to complex, multiple supportive devices and wheelchairs. (Walling, 1999)
Involvement of Physical Therapists along with orthotists is important to identify the optimal brace; brace use will need to be adjusted as the disease progresses. (Majmudar, 2014)
It is important to use equipment/technology that can prolong independence as long as possible; unfortunately, most insurance carriers do not cover these devices. (Majmudar, 2014)
"Because of the uncertainty of its effects, exercise has not been a standard treatment for ALS." (Lewis, 2007), but a Cochrane review found that more research is needed in this area. They did note "exercise produced a greater average improvement in function than usual care, and there were no reported adverse events due to exercise" (Dal Bello-Haas, 2013).
Psychological treatment:
Discerning the signs of depression and making an appropriate referral, caregiver education in the cognitive deficits associated with fronto-temporal dementia (Lewis, 2007)
Ongoing change and adaptation model framework for the decision-making processes (King, 2009)
One RCT indicated that a group of patients that disclosed feelings about ALS had higher well-being (at three months post intervention, but not six months) than a group of patients that did not disclose feelings. (Averill, 2013)
"Expressive disclosure (being able to disclose all thought and feelings about the diagnosis of ALS), may be helpful for people with ALS, but only those who have difficulty expressing emotions." (Averill, 2013)
Nutritional Support
"Early in the disease course, without dysphagia, monitor food and fluid intake monthly to assure that nutrition and weight stability is sufficient, (Atsuta, 2009).
After onset of dysphagia, diet modification to soft semisolid textured foods, with thicker liquids is important to help prevent aspiration (Langmore, 1999; Perry, 2002)
Continued weight loss despite intervention (BMI <20 kg/m2 or >5–10 % body weight loss) requires artificial nutrition (Piquet, 2006; Cameron, 2002; Katzberg, 2011)
+++
IV. Community and Professional Resources
++
Amyotrophic Lateral Sclerosis Association
National Institute of Neurological Disorders and Stroke: Amytrophic Lateral Sclerosis (ALS) Fact Sheet
National ALS Registry
Les Turner ALS Foundation
Project A.L.S. – Finding and Funding a Cure for ALS
ALS Caregiver's Guide
ALS Therapy Development Institute
Robert Packard Center for ALS Research at John Hopkins
Team Gleason
+
ALS Association. The ALS Association page. Available at
www.alsa.org. Accessed February 23, 2007.
+
APTA, Guide to Physical Therapy Practice. 2003
+
Armon
C. An evidence-based medicine approach to the evaluation of the role of exogenous risk factors in sporadic amyotrophic lateral sclerosis. Neuroepidemiology. 2003; 22: 217–28.
+
Atsuta
N, Watanabe
H, Ito
M, Tanaka
F, Tamikoshi
A, Nakano
I, Aoki
M, Tsuji
S, Yuasa
T, Takano
H, Hayashi
H, Kuzuhara
S, Sobue
G. Age at onset influences on wide-ranged clinical features of sporadic amyotrophic lateral sclerosis. J Neurol Sci. 2009;276(1–2):163–9.
+
Ashworth
NL, Satkunam
LE, Deforge
D. Treatment for spasticity in amyotrophic lateral sclerosis/motor neuron disease (Cochrane Review) The Cochrane Library. 2004; 4.
+
Averill
AJ, Kasarskis
EJ, Segerstrom
SC. Expressive disclosure to improve well-being in patients with amyotrophic lateral sclerosis: a randomized, controlled trial. Psychol Health. 2013;28(6):701–13.
+
Bello-Haas
V, Kloos
A, Mitsumoto
H. Physical therapy for a patient through 6 stages of amyotrophic lateral sclerosis. Phys Ther. 1998; 78:1312–1324.
+
Benatar
M, Kurent
J, Moore
DH. Treatment for familial amyotrophic lateral sclerosis/motor neuron disease. Cochrane Database Syst Rev. 2009
+
Brooks
BR. Managing amyotrophic lateral sclerosis: slowing disease progression and improving patient quality of life. Ann Neurol. 2009; 65: Suppl 1: S17–23
+
Brown
R, Lockwood
A, Sonawane
B. Neurodegenerative Diseases: an overview of environmental risk factors. Environmental Health Perspectives. 2005; 113: 1250–1256.
+
Browne
E, Abbott
B. Recent progress towards an effective treatment of amyotrophic lateral sclerosis using the SOD1 mouse model in a preclinical setting. Eur J Med Chem. 2016;121:918–925.
+
Bucchia
M, Ramirez
A, Parente
V, Simone
C, Nizzardo
M, Magri
F, Dametti
S, Corti
S. Therapeutic Development in Amyotrophic Lateral Sclerosis. Clin Ther. doi:10.1016/j.clinthera.2014.12.020.
+
Cameron
A, Rosenfeld
J. Nutritional issues and supplements in amyotrophic lateral sclerosis and other neurodegenerative disorders. Curr Opin Clin Nutr Metab Care. 2002;5(6):631–43.
+
Capozzo
R, Quaranta
VN, Pellegrini
F, Fontana
A, Copetti
M, Carratu
P, Panza
F, Cassano
A, Falcone
VA, Tortelli
R, Cortese
R, Simone
IL, Resta
O, Logroscino
G. Sniff nasal inspiratory pressure as a prognostic factor of tracheostomy or death in amyotrophic lateral sclerosis. J Neurol. 2015;262(3):593–603.
+
Carvalho
M, Swash
M. Lower motor neuron dysfunction in ALS. Clinl Neurophysiol. 2016;127(7):2670–2681.
+
Chen
K, Sakowski
S, Feldman
E. Intraspinal stem cell transplantation for amyotrophic lateral sclerosis. Ann Neurol. 2016;79(3):342–353.
+
Costa
J, Swash
M, de Carvalho
M. Awaji Criteria for the Diagnosis of Amyotrophic Lateral Sclerosis: A Systematic Review.
Arch Neurol.[Archives of Neurology Full Text] 2012;69(11):1410–1416.
+
Ferguson
T, Elman
L. Clinical Presentation and Diagnosis of Amyotrophic Lateral Sclerosis. NeuroRehabilitation. 2007; 22:409–416.
+
Floyd
Ag
et al.. Transcranial magnetic stimulation in ALS: utility of central motor conduction tests. Neurology. 2009 Feb 10; 72(6): 498–504
+
Gallo
V,
et al. Smoking and risk for amyotrophic lateral sclerosis: analysis of the EPIC cohort. Ann Neurol. 2009 Apr; 65(4): 378–85
+
Gianforcaro
A, Hamadeh
MJ. Vitamin D as a potential therapy in amyotrophic lateral sclerosis. CNS Neurosci Ther. 2014;20(2):101–11.
+
Gil,
J, Funalot,
B. Causes of death amongst French patients with amyotrophic lateral sclerosis: a prospective study. European Journal Neurology. 2008 Nov Vol 15 (11):1245–51.
+
Gordon
PH. Amyotrophic Lateral Sclerosis: An update for 2013 Clinical Features, Pathophysiology, Management and Therapeutic Trials. Aging Dis. 2013;4(5):295–310.
+
Hardiman
O. Management of respiratory symptoms in ALS. J Neurol. 2010;258(3):359–365.
+
Hersi
M, Quach
P, Wang
M, Gomes
J, Gaskin
J, Krewski
D. Systematic reviews of factors associated with the onset and progression of neurological conditions in humans: A methodological overview.
Neurotoxicology. 2016;
http://dx.doi.org/10.1016/j.neuro.2016.06.017.
+
Hooten
KG, Beers
DR, Zhao
W, Appel
SH. Protective and Toxic Neuroinflammation in Amyotrophic Lateral Sclerosis. Neurotherapeutics. 2015;12(2):364–75.
+
Huynh
W, Simon
N, Grosskreutz
J, Turner
M, Vucic
S, Kiernan
M. Assessment of the upper motor neuron in amyotrophic lateral sclerosis. Clin Neurophysiol. 2016; 127: 2643–2660.
+
Ingre
C, Roos
PM, Piehl
F, Kamel
F, Fang
F. Risk factors for amyotrophic lateral sclerosis. Clin Epidemiol. 2015;7:181–93.
+
Katzberg
H, Benatar
M. Enteral tube feeding for amyotrophic lateral sclerosis/motor neuron disease. Cochrane Database Syst Rev. 2011;1:CD004030.
+
King
SJ, Duke
MM, O'Connor
BA. Living with amyotrophic lateral sclerosis/motor neurone disease (ALS/MND): decision-making about 'ongoing change and adaptation'. J Clin Nurs 2009 Mar; 18(5): 745–54
+
Kumar
V, Islam
A, Hassan
M, Ahmad
F. Therapeutic progress in amyotrophic lateral sclerosis-beginning to learning. Euro J Med Chem. 2016; 121: 903–917.
+
Lacomblez
L, Bensimon
G, Leigh
PN, Guillet
P, Meininger
V. Dose-ranging study of riluzole in amyotrophic lateral sclerosis. Amyotrophic Lateral Sclerosis/Riluzole Study Group II. Lancet. 1996;347:1425–1431.
+
Langmore
S. Issues in the management of dysphagia. Folia Phoniatr Logop. 1999;51(4–5):220–30.
+
Logroscino
G, Traynor
BJ, Hardiman
O, Chio'
A, Couratier
P, Mitchell
JD, Swingler
RJ, Beghi
E. Descriptive epidemiology of amyotrophic lateral sclerosis: new evidence and unsolved issues. J Neurol Neurosurg Psychiatry. 2008;79:6–11.
+
Lee
J, Annegers
J, Appel
S. Prognosis of amyotrophic lateral sclerosis and the effect of referral selection. Journal of the Neurological Sciences. 1995; 132:207–215.
+
Lewis
M, Rushanan
S. The Role of Physical Therapy and Occupational Therapy in the Treatment of Amyotrophic Lateral Sclerosis. NeuroRehabilitation. 2007; 22:451–461.
+
Macpherson
C, Bassile
C. Pulmonary Physical Therapy Techniques to Enhance Survival in Amyotrophic Lateral Sclerosis. J Neurol Phys Ther. 2016;40(3):165–175.
+
Majmudar
S, Wu
J, Paganoni
S. Rehabilitation in ALS: Why it matters. Muscle Nerve. 2014; doi: 10.1002/mus.24202.
+
Marangi
G, Traynor
BJ. Genetic causes of amyotrophic lateral sclerosis: new genetic analysis methodologies entailing new opportunities and challenges. Brain Res. 2015;1607:75–93.
+
Mazzini
L, Mareschi
K, Ferrero
I, Vassallo
E, Oliveri
G, Nasuelli
N, Oggioni
G, Testa
L, Fagioli
F. Stem Cell Treatment in Amyotrophic Lateral Sclerosis. Journal of the Neurological Sciences. 2008; 265:78–83.
+
McClelland
S, Bethoux
FA, Boulis
NM, Sutliff
MH, Stough
DK, Schwetz
KM, Gogol
DM, Harrison
M, Pioro
EP. Intrathecal baclofen for spasticity-related pain in amyotrophic lateral sclerosis: efficacy and factors associated with pain relief. Muscle Nerve. 2008;37(3):396–8.
+
Miller
RG
et al. Outcomes research in amyotrophic lateral sclerosis: lessons learned from the amyotrophic lateral sclerosis clinical assessment, research, and education database. Ann Neurol. 2009; 65: Suppl 1: S24–8
+
Mitchell
JD. Amyotrophic lateral sclerosis: toxins and environment. Amyotroph Lateral Scler Other Motor Neuron Disord. 2000; 1: 235–50.
+
National Institute of Neurologic Disorders and Stroke. ALS (Amyotrophic Lateral Sclerosis) Fact Sheet. Bethesda, Maryland. National Institute of Health. 2011
+
Perry
A, Anderson
K, Lean
R, Cotton
S. Elevation of the soft palate in speech and swallowing in normal female participants and females with motor neuron disease: an innovative procedure for measuring palatal elevation. Int J Lang Commun Disord. 2002;37(2):197–214.
+
Piquet
MA. [Nutritional approach for patients with amyotrophic lateral sclerosis]. Rev Neurol (Paris). 2006;162(Spec No 2):4S177–4S187.
+
Pradhan
S, Yadav
R, Mishra
V,
et al. Amyotrophic lateral sclerosis with predominant pyramidal signs-early diagnosis by magnetic resonance imaging. Magnetic Resonance Imaging. 2006; 24:173–179.
+
Radunovic
A, Annane
D, Rafiq
MK, Mustfa
N. Mechanical ventilation for amyotrophic lateral sclerosis/motor neuron disease. Cochrane Database Syst Rev. 2013, Issue 3. Art. No.: CD004427.
+
Rothstein
JD. Current hypotheses for the underlying biology of amyotrophic lateral sclerosis. Ann Neurolog. 2009; 65: Suppl 1: S3–9
+
Rowland,
L, Shneider,
N. Amytrophic Lateral Sclerosis. NEJM. 2001;344(22): pg 1688–1700.
+
Saeed
et al., Age and founder effect of SOD1 A4V mutation causing ALS. Neurology, 2009; 72(19): 1634–9.
+
Turner
MR, Hardiman
O, Benatar
M, Brooks
BR, Chio
A, de Carvalho
M, Ince
PG, Lin
C, Miller
RG, Mitsumoto
H, Nicholson
G, Ravits
J, Shaw
PJ, Swash
M, Talbot
K, Traynor
BJ, Van den Berg
LH, Veldink
JH, Vucic
S, Kiernan
MC. Controversies and priorities in amyotrophic lateral sclerosis. The Lancet Neurology 2013;12(3):310–322.
+
Walling,
A. Amyotrophic Lateral Sclerosis: Lou Gehrig's Disease. American Academy of Family Physicians. 1999 Mar 15; Volume 59(6):1489–1496.
+
Wang
M, Little
J, Gomes
J, Cashman
N, Krewski
D. Identification of risk factors associated with onset and progression of amyotrophic lateral sclerosis using systematic review and meta-analysis. Neurotoxicology. 2016. doi:10.1016/j.neuro.2016.06.015.
+
Wijesekera
LC
et al.. Natural history and clinical features of the flail arm and flail leg ALS variants. Neurology. 2009 Mar 24; 72(12): 1087–94.
+
Robberecht
W, Philips
T. The changing scene of amyotrophic lateral sclerosis. Nat rev neurosci. 2013;14(4):248–264.
+
Wisjesekera,
L, Leight,
P. Amytrophic Lateral Sclerosis. Orphanet Journal of Rare Diseases. 2009;4:3. BioMed Central. DOI: 10.1186/1750-1172-4-3.
+
Zaldivar
T
et al.. Reduced frequency of ALS in an ethnically mixed population: A population-based mortality study. Neurology. 2009; 72(19): 1640–5.
+
Zoccolella
S, Beghi
E, Palagano
G,
et al. Signs and symptoms at diagnosis of amyotrophic lateral sclerosis: a population-based study in southern Italy. European Journal of Neurology. 2006; 13:789–792.
++
A disorder of genomic imprinting that causes severe developmental and neurological deficits (Lalande, 2007)
Estimated frequency of 1:10,000 to 1:40,000 births (Paprocka, 2007)
Etiology: Genetic mechanisms:
Chromosome 15q11-q13 abnormalities: deletion in 70-75% of cases (most severely affected) and paternal uniparental disomy (UPD) in 2-5% of cases (Pelc, 2008; Clayton-Smith, 2003; Lalande, 2007; Didden, 2006)
Mutated UBE3A gene in 20-25% of cases (Jiang, 1999; Clayton-Smith, 2003; Didden, 2006)
Imprinting defects 2-5% of cases (Jiang, 1999; Clayton-Smith, 2003; Didden 2006)
Increased risk in child when mother's time trying to become pregnant is greater than 2 years (Ludwig, 2005).
Increased risk in child conceived by intracytoplasmic sperm injection (Cox, 2002)
Pathogenesis: UBE3A encodes for ubiquitin protein ligase- responsible for eliminating damaged/unnecessary proteins (Matsuura 1997; Jiang, 1999; Guerrini, 2003, Dagli, 2012).
++
++
Non-progressive, but with variable degrees of symptoms secondary to acquired genetic etiology; a typical lifespan is expected.
Developmental history and neurological sequelae
6-12 months: developmental delay, decreased head circumference; 3 years of age.: seizures and abnormal EEG
5-9 years of age: wide based gait, structurally characterized by ankle valgus
Adulthood: ↓hyperactivity, ↓sleep disturbances, ↓seizure frequency, exaggerated facial features; unable to live independently, but can perform simple home tasks (Guerrini, 2003; Peters, 2004; Didden, 2004)
+++
IV. Medical and Surgical Management
++
Surgical techniques (for symptoms)-adenoidectomy and tonsillectomy (↓ sleep apnea); surgical tightening of the esophageal sphincter (manage GERD); corrective eye surgery (↓ nystagmus); heel cord surgery (improve structure of foot and ankle); thoraco-lumbar rod stabilization (corrects severe scoliosis) (Guerrini, 2003)
Pharmaceuticals-Depakote, Klonopin, Lamotrigene therapy, corticosteroids for seizures; Melatonin and diphenylhydramines for sleep disorders; laxatives for constipation, motility drugs for GERD, Methylphenidate for hyperactivity (Guerrini, 2003; Dion, 2007; Forrest, 2009)
Minocycline has shown promising results in improving communication, fine motor ability, and auditory comprehension. (Grieco, 2014)
+++
V. Implications of Therapeutic Management
++
Interventions should be individualized to address musculoskeletal impairments, disorders of gait and movement, speech and language impairments, and behavioral issues that have led to activity limitations and participation restrictions. (Bonanni, 2009)
Use of aquatic therapy to address range of motion deficits, decreased cardiovascular endurance, gait deficits, and poor motor control and coordination due to an affinity for water (Didden, 2009)
Hippotherapy- may improve postural control, strength, coordination, self-esteem, and quality of life (Thompson, 2014)
Physical therapy to monitor musculoskeletal development for signs of deformity and evaluate the use of adaptive equipment for prevention of joint deformity and to assist with functional activities i.e. helmet, gait devices, adaptive car seat, adaptive chairs and positioners, and orthotic bracing for foot deformity or scoliosis (Guerrini, 2003)
Speech therapy to promote functional communication using augmentative communication and sign language (i.e. Digivox, Cheap-Talk4, Macaw) (Guerrini, 2003; Didden, 2004)
Occupational therapy for environmental adaptations; special educators for educational adaptations and psychologists for behavior management (Guerrini, 2003)
Dietician or nutritionist for consultation for children with increased body mass index (Brennan, 2015)
All members of the rehabilitation team, along with parents and teachers provide input for individualized education programs to assist with success at school. Discrete Trial Instruction (DTI) approaches are appropriate for building functional skills in children with AS (Guerrini, 2003; Summers, 2009)
+
Beckung
E, Steffenburg
S, Kyllerman
M. Motor impairments, neurological signs, and developmental level in individuals with Angelman syndrome. Dev Med Child Neurol. 2004; 46:239–243.
+
Bonanni
P, Gobbo
A, Nappi
S,
et al. Functioning and disability in patients with Angelman syndrome: utility of the International Classification of Functioning Disability and Health, children, and youth adaptation framework. Disabil Rehabil. 2009;31(sup1):S121–S127.
+
Brennan
M-L, Adam
MP, Seaver
LH,
et al. Increased body mass in infancy and early toddlerhood in Angelman syndrome patients with uniparental disomy and imprinting center defects. Am J Med Genet. Part A. 2015;167(1):142–146.
+
Bruni
O, Ferri
R, D'Agostino
G, Miano
S, Roccella
M, Elia
M. Sleep disturbances in Angelman syndrome: a questionnaire study. Brain Dev. 2004;26:233–240.
+
Clayton-Smith
J, Laan
L. Angelman syndrome: a review of the clinical and genetic aspects. J Med Genet. 2003;40:87–95.
+
Coppola
G, Verotti
A, Mainolfi
C, Auricchio
G, Fortunato
D, Operto
F, Pascotto
A. Bone mineral density in Angelman syndrome. Pediatr Neurol. 2007;37(6):411–416.
+
Cox
GF, Bürger
J, Lip
V,
et al. Intracytoplasmic sperm injection may increase the risk of imprinting defects. Am J Hum Genet. 2002;71:162–164.
+
Dagli
A, Buiting
K, Williams
CA. Molecular and clinical aspects of Angelman syndrome. Mol Syndromol. 2012;2:100–12.
+
Didden
R, Korzilius
H, Duker
P, Curfs
L. Communicative functioning in individuals with Angelman syndrome: a comparative study. Disabil Rehabil. 2004;26(21/22):1263–1267.
+
Didden
R, Korzilius
H, Kamphuis
A, Sturmey
P, Lancioni
G, Curfs
L. Preferences in individuals with Angelman syndrome assessed by a modified Choice Assessment Scale. J Intellect Disabil Res. 2006;50(1):54–60.
+
Didden
R, Korzilius
H, Sturmey
P, Lancioni
GE, Curfs
LMG. Preference for water-related items in Angelman syndrome, Down syndrome and non-specific intellectual disability. J Intellect Dev Disabil. 2008;33(1):59–64.
+
Didden
R, Sigafoos
J, Korzilius
H,
et al. Form and function of communicative behaviors in individuals with Angelman syndrome. J Appl Res Intellect. 2009;22:526–537.
+
Dion
M, Novotny
E, Carmant
L, Cosette
P, Nguyen
D. Lamotrigene therapy of epilepsy with Angelman syndrome. Epilepsia. 2007;48:593–596.
+
Forrest
KML, Young
H, Dale
RC, Gill
DS. Benefit of corticosteroid therapy in Angelman syndrome. J Child Neurol. 2009; 24: 952–958.
+
Grieco
JC, Ciarlone
SL, Gieron-Korthals
M,
et al. An open-label pilot trial of minocycline in children as a treatment for Angelman syndrome. BMC Neurol. 2014;14(1):232.
+
Guerrini
R, Carrozzo
R, Rinaldi
R, Bonanni
P. Angelman syndrome: etiology, clinical features, diagnosis and management of symptoms. Pediatr Drugs. 2003;5(10):647–661.
+
Jiang
Y, Lev-Lehman
E, Bressler
J, Tsai
T, Beaudet
A. Genetics of Angelman syndrome. Am J Hum Genet. 1999;65:1–6.
+
Korff
C, Kelley
K, Nordli
D. Notched delta, phenotype, and Angelman syndrome. J Clin Neurophysiol. 2005;22:238–243.
+
Lalande
M, Calciano
M. Molecular epigenetics of Angelman syndrome. Cell Mol Life Sci. 2007;64:947–960.
+
Ludwig
M, Katalinic
A, Grob
S,
et al. Increased prevalence of imprinting defects in patients with Angelman syndrome born to subfertile couples. J Med Genet. 2005;42:289–291.
+
Matsuura
T, Sutcliffe
JS, Fang
P,
et al. De novo truncating mutations in E6-AP ubiquitin-protein ligase gene (UBE3A) in Angelman syndrome. Nat Genet. 1997;15:74–7.
+
Oliver
C, Horsler
K, Berg
K, Bellamy
G, Dick
K, Griffiths
E. Genomic imprinting and the expression of affect in Angelman syndrome: what's in the smile? J Child Psychol Psychiatry. 2007;48(6):571–579.
+
Paprocka
J, Jamroz
E, Szwed-Bialozyt
B, Jezela-Stanek,
A, Kopyta
I, Marszal
E. Angelman syndrome revisited. The Neurologist. 2007;13(5):305–312.
+
Pelc
K, Cheron
G, Dan
B. Behavior and neuropsychiatric manifestations in Angelman Syndrome. Neuropsychiatr Dis Treat. 2008;4(3):577–84.
+
Peter
S, Goddard-Finegold
J, Beaudet
A, Madduri
N, Turchich
M, Bacino
C. Cognitive and adaptive behavior profiles of children with Angelman syndrome. Am J of Med Genet. 2004;128:110–113.
+
Summers
J, Szatmari
P. Using Discrete Trial Instruction to teach children with Angelman Syndrome. Focus Autism Other Dev Disabil. 2009;24:216–226.
+
Thompson
F, Ketcham
CJ, Hall
EE. Hippotherapy in children with developmental delays: physical function and psychological benefits. Adv Phys Educ. 2014;4(2):10.
+
Williams
C. Neurological aspects of Angelman syndrome. Brain Dev. 2005;27:88–94.
+
Yis
U, Giray
O, Kurul
S,
et al. Long-standing fever and Angelman syndrome: report of two cases. J Paediatr Child H. 2008;44:308–310.
+++
Arthrogryposis Multiplex Congenita
++
Arthrogryposis multiplex congenita (AMC) is a term used to describe the presence of multiple congenital joint contractures in than one area of the body; not a disease entity but a non-progressive clinical presentation of joint contractures (Kalampokas, 2012).
The single term arthrogryposis denotes congenital contractures either in an isolated joint or multiple joints (Bamshad, 2009)
Causes of arthrogrypotic syndromes are largely unknown, but most often multifactorial (Bevan, 2007).
Subgroups of AMC (Bamshad, 2009, Kalampokasc, 2012)
More than 35 specific genetic disorders associated with arthrogryposis have been identified (Bevan, 2007).
AMC has been described in all races and equally in both genders with an incidence of 1 in 5-10,000 (Mennen 2005).
Normal intelligence is typical (Bamshad, 2009).
Cases that present with decreased fetal movement during the first semester present are associated with the most long-term functional deficits (Donohoe, 2012).
++
Signs and Symptoms
Common signs and symptoms include: dimpling of skin over joints, clubfeet, muscle malformation, hemangiomas, absent or decreased finger creases, congenital heart disease, facial abnormalities (Donohoe, 2012).
All types have decreased fetal movement in common with limited range of motion and weakness is associated joint (Mennen, 2005).
Affected body areas: foot (78-95%), hip (60 -82%), wrist (43-81%), knee (41-79%), elbow (35-92%) and shoulder (20%-92%) (Donohoe, 2012)
Type I ("jackknifed"): flexed and dislocated hips, extended knees, clubfeet, internally rotated shoulders, flexed elbows, flexed and ulnarly deviated wrists (Donohoe, 2012).
Type II ("froglike"): abducted and externally rotated hips, flexed knees, clubfeet, internally rotated shoulders, extended elbows, flexed and ulnarly deviated wrists (Donohoe, 2012).
Comorbidities include decreased muscle mass at times inhibiting distribution of some medications, difficult venous access, post-op more prone to respiratory problems (atelectasis, stridor, increased risk of aspiration, difficulty with intubation, and vertebral instability/scoliosis, facial/jaw abnormalities, congenital heart defects, abdominal hernias, tracheoesophageal fistulas, ophthalmologic abnormalities, gastrointestinal difficulties, and speech-language delays) (Bevan, 2007, Kalampokas, 2012)
Diagnostic testing
Abnormal ultrasound and maternal perception of decreased movement in 2nd and 3rd trimesters of pregnancy (Bevan, 2007)
Radiographs, CT's, MRI's, EEG's, EMG's, muscle biopsy, and NCV studies are useful tools (Donohoe, 2012).
++
Non-progressive
In most cases, the outlook for those with AMC is positive with most children with amyoplasia functionally ambulating by 5 years old with varying degrees of functional ambulation in all types (50-78%) (Bevan, 2007)
Long term prognosis depends on the extent of involvement: mortality rate of 1%, 7%, and 50% for infants with limb, limb and other organ, and CNS involvement respectively.
+++
IV. Medical/surgical Management
++
Surgery may be utilized to align the angle of alkalosis, but range of joint motion is rarely improved (Milanese, 2007).
Various operative procedures aim at reducing the deformities associated with AMC and are effective in improving mobility and ambulatory gains (Harold et al., 2007).
Osteotomies, carpectomies, elbow flexorplasty, various muscle transfers (Mennen, 2005).
+++
V. Key Aspects of Therapeutic Management
++
Congenital malformations and deformations of the musculoskeletal system: 2015 ICD-10-CM Diagnosis Code Q74.3
Role of the physical therapist: to maximize functional skills by using knowledge of biomechanics and typical development (Donohoe, 2012).
Health promotion and prevention measures (education)
Parental involvement is a key factor in the success of physical therapy for infants (O'Flaherty, 2001).
Problem-solving with the family, child when appropriate, and healthcare team to promote opportunities for health and fitness
Interdisciplinary health care team contributions include:
Pediatrician, geneticist, physical therapist, occupational therapist, and a pediatric psychiatrist (Bevan, 2007)
Addressing family centered goals to promote positive self-image and to maximize lifelong potential for independent living
Serving as a member of the educational team to promote success in the educational environment.
+++
VI. Consumer and Professional Resources
++
+
Bamshad
M, VanHeest
AE, Pleasure
D. Arthrogryposis: a review and update. J Bone Joint Surg Am. 2009;91 Suppl 4:40–6.
+
Bevan
WP, Hall
JG, Bamshad
M, Staheli
LT, Jaffe
KM, Song
K. Arthrogryposis multiplex congenita (amyoplasia): an orthopaedic perspective. J Pediatr Orthop. 2007;27(5):594–98.
+
Donohoe
M. Arthrogryposis multiplex congenita. In: Campbell
SK, Palisano
RJ, Orlin
MN. Physical Therapy for Children. 4th ed. St. Louis, MO: Saunders Elsevier; 2012: 313–332.
+
Fassier
A, Wicart
P, Dubousset. Arthrogryposis multiplex congenital. Long-term follow-up from birth until skeletal maturity. J Child Orthop. 2009:3:383–390.
+
Hall
JG. Arthrogryposis multiplex congenita: etiology, genetics, classification, diagnostic approach, and general aspects. J Pediatr Orthop. 1997;6(3):159–66.
+
Van Bosse
HJ, Feldman
DS, Anavian
J, Sala
DA. Treatment of knee flexion contractures in patients with arthrogryposis. J Pediatr Orthop. 2007; 27(8);930–937.
+
Kalampokas
E, Kalampokas
T, Sofoudi
C, Deligeoroglou
E, Botsis
D. Diagnosing arthrogryposis multiplex congenita: a review. ISRN Obstet Gynecol. 2012;2012:264918.
+
Mennen
U, van Heest
A, Ezaki
MB, Tonkin
M, Gericke
Gd. Arthrogryposis multiplex congenita. J Hand Surg Br. 2005;30(5):468–74.
+
Milanes
GM, Napolitano
R, Quaglia
F,
et al. Prenatal diagnosis of arthrogryposis. Minerva Ginecologica. 2007;59(2):203–204.
+
O'Flaherty
P. Arthrogryposis multiplex congenita. Neonatal Network. 2001;20(4):13–20.
+++
Autism Spectrum (Pervasive Developmental Disorder)
++
Consistent with Diagnostic and Statistical Manual of Mental Disorders, 5th edition (DSM-5) published by the American Psychiatric Association (APA), the preferred terminology is "autism spectrum disorder" (ASD) which encompasses autism disorder, Asperger's disorder and pervasive developmental disorder but not Rett syndrome (APA, 2013).
According to the Centers for Disease Control and Prevention (CDC), Autism and Developmental Disabilities Monitoring Network data from 2012, the combined estimated prevalence of autism among 8 year old children is 14.6 per 1000 (1 in 68 children); 4.5 times more prevalent in boys (1 in 42 boys); and highest among non-Hispanic white population (CDC, 2017).
Likely multiple causes for different types of ASD including environmental, biologic and genetic risk factors (CDC, 2017):
Specific genes or gene mutations
Siblings of child with ASD (Hallmayer, 2011)
Children born to older parents
Maternal use of valproic acid or thalidomides during pregnancy
In association with certain other genetic disorders (e.g., fragile X, tuberous sclerosis, Down syndrome, neurofibromatosis)
Neuropathophysiology: poorly understood, but findings include:
Under connectivity (Just, 2012)
Altered function of mirror neuron system (Enticott, 2012)
Variable abnormal findings with neuro-imaging
Co-occurring diagnoses among 2,568 8-year old children with ASD: developmental (83%), psychiatric (10%), neurologic (16%) and possibly causative genetic or neurologic (4%) (Levy, 2010).
++
DSM-5 criteria for the diagnosis of autism spectrum disorder (APA, 2013):
Deficits in social communication and social interaction across multiple contexts:
Deficits may be in social-emotional reciprocity
Deficits in nonverbal communicative behaviors
Deficits in developing, maintaining, and understanding relationships
Restricted repetitive patterns of behavior, interest, or activities: the individual must demonstrate two of the following four criteria:
Stereotyped or repetitive motor movement, use of objects, or speech (line up toys, echolalia, etc.)
Insistence on sameness, inflexible adherence to routines, or ritualized patterns of verbal or nonverbal behavior (difficulties with transitions, greeting rituals, etc.)
Highly restricted, fixated interests that are abnormal in intensity or focus (perseverative interests, strong attachment to or preoccupation with unusual objects, etc.)
Hyper-or hypo-reactivity to sensory input or unusual interest in sensory aspects of the environment (adverse responses to sounds or textures, apparent indifference to pain/temperature, etc.)
Symptoms must be present in the early developmental period.
Symptoms cause clinically significant impairment in social, occupational, or other important areas of current functioning.
Disturbances are not better explained by intellectual disability or global developmental delay.
American Academy of Pediatrics recommends that primary care providers screen for ASD with valid and reliable tool during 18 and 24 month well-child visits because early detection and referral for early intervention services can improve outcomes (Zwaigenbaum, 2015; Daniels, 2014).
Screening tools are available, including:
Modified Checklist for Autism Spectrum Disorder in Toddlers (Dumont-Mathieu, 2005)
Pervasive Developmental Disorders Screening Test-II (Siegel, 2004)
Screening Tool for Autism in Two Year-Olds (Stone, 2000)
Social Communication Questionnaire (SCQ) (Rutter, 2003a; Rutter, 2003b; Bolte, 2008)
Comprehensive interdisciplinary evaluation should be based on DSM-5 diagnostic criteria. A number of evaluative tools are available (Academy of Pediatric Physical Therapy, 2015).
Diagnosis can sometimes be made before 18 months of age and should be possible by an experienced professional by 2 years of age but may be delayed until children enter school environment (CDC, 2017).
New genetic testing technology can identify a genetic etiology for approximately 15.8% of children with ASD (Tammimies, 2015).
Common behaviors/symptoms in children or adults with ASD (CDC, 2017):
Not point at objects to show interest (e.g., not point at an airplane flying over)
Not look at objects when another person points at them
Have trouble relating to others or not have an interest in other people at all
Avoid eye contact and want to be alone
Have trouble understanding other people's feelings or talking about their own feelings
Prefer not to be held or cuddled, or might cuddle only when they want to
Appear to be unaware when people talk to them, but respond to other sounds
Be very interested in people, but not know how to talk, play, or relate to them
Repeat or echo words or phrases said to them, or repeat words or phrases in place of normal language
Have trouble expressing their needs using typical words or motions
Not play "pretend" games (for example, not pretend to "feed" a doll)
Repeat actions over and over again
Have trouble adapting when a routine changes
Have unusual reactions to the way things smell, taste, look, feel, or sound
Lose skills they once had (for example, stop saying words they were using)
Almost half of children with ASD have average or above average intelligence, according to CDC-funded 2017 Study to Explore Early Development (SEED) (CDC, 2017)
Possibly altered sleeping and eating patterns (CDC, 2017)
Gross and fine motor symptoms common but not part of DSM-5 criteria (Gkotzia, 2017; Downey, 2012).
Motor coordination deficits a "cardinal feature" of ASD (Fournier, 2010)
Motor delays on standardized testing
Impaired imitation skills, clumsiness; difficulties with motor planning
Possible asymmetries in early motor development
Decreased postural stability – tendency to rely on reactive rather than more typical anticipatory postural control – in standing, increased sway, abnormal weight distribution and absence of typical ankle strategies – rely more heavily on visual input for balance
Sensory processing symptoms common but not part of DSM-5 criteria
Tomchek et al (2014) examined sensory processing patterns in 400 children with ASD and identified 6 parsimonious factors: (1) low energy/weak, (2) tactile and movement sensitivity, (3) taste/smell sensitivity, (4) auditory and visual sensitivity, (5) sensory seeking/distractibility, and (6) hypo-responsivity.
Sensory integration and praxis patterns: relative strengths in visual praxis; difficulties in imitation praxis, vestibular bilateral integration, somatosensory perception and sensory reactivity (Roley, 2015)
Hypo or hyper-reactivity to sensory input
+++
III. Prognosis and Sequelae
++
Autism is non-progressive and has no known cure
Twenty-year outcomes for adults with cognitive functioning in near-average and average ranges (Farley, 2009):
Rating of "Very Good" or "Good" on global outcome measure for more than half of participants
Considerable variability in measured cognitive and adaptive abilities over time (consistent with other studies)
+++
IV. Medical/Surgical Management
++
Medications may be used to help manage behaviors that interfere with function (e.g., decrease anxiety, alleviate depression, improve attention, balance energy levels or decrease self-injurious behaviors).
Nutrition/Diet – a variety of diets have been recommended that provide vitamin/nutritional supplements or restrict certain foods, but scientific evidence does not support any particular diet for individuals with autism (CDC, 2017).
Obesity is a concern for individuals with ASD and should be managed with appropriate diet and physical activity (Curtin, 2010; Srinivasan, 2014).
No surgical management applicable to primary symptoms of ASD
+++
V. Key Aspects of Therapeutic Management
++
Behavioral and education approaches focus on providing consistent organization, structure and direction, often breaking tasks down into manageable discrete steps. It is important that team members and family members follow basic constructs of these approaches. A valuable resource is an Evidence-Based Practices Comparison Chart developed by the Centers for Medicare and Medicaid Services, the National Autism Center and the National Professional Development Center on ASD available at: https://www.autism-society.org/wp-content/uploads/2014/04/ebp-comparison-chart-o.pdf. Some of the more common programs:
Applied Behavior Analysis (ABA): uses positive reinforcement to teach specific tasks/skills; several different defined approaches
Developmental, Individual Differences, Relationship-Based Approach (DIR) or "Floortime": focuses on emotional and relational development and how the child deals with sights, sounds, and smells.
Treatment and Education of Autistic and related Communication-handicapped Children (TEACCH): uses visual cues to teach skills. For example, picture cards can help teach a child how to get dressed by breaking information down into small steps.
Picture Exchange Communication System (PECS): teaches communication skills using picture symbols
Intervention for motor delays or movement disorders
Assessment utilizing standardized instruments
A comprehensive resource on assessment tools for evaluation of children with ASD in school-based practice was published as a "Fact Sheet" by the Academy of Pediatric Physical Therapy (2015).
Three commonly used tests of motor proficiency (Gkotzia, 2017):
Bruininks-Oseretsky Test of Motor Proficiency, 2nd edition (Bruininks, 2005)
Movement Assessment Battery for Children-2 (Henderson, 2007)
Test of Gross Motor Development, 2nd edition (Ulrich, 2000)
Utilize general approaches to evaluate and treat gross and fine motor delays or movement disorders; consider likelihood of coordination and balance problems
Incorporate instructional strategies that will maximize potential for success (Srinivasan, 2014; Gkotzia, 2017)
Support development of structured exercise programs for general health, improved motor performance, weight control, academic engagement, and behavioral management
Meta-analysis examined effects of structured exercise programs for children and adults with autism provided in individual and group contexts; combined results provide support for improvements in motor performance and social skills with the largest benefit achieved with individual vs. group intervention (Sowa, 2012).
Systematic review of literature by Lang et al (2010) on effects of participation in a physical exercise program for, across the corpus of studies, 64 individuals with ASD (ages 3-41): decreases in stereotypy, aggression, off-task behavior and elopement; and increases in on-task behavior, academic responding, and appropriate motor behavior.
Physical activity may help prevent obesity among individuals with ASD (Curtin, 2010; Srinivasan, 2014)
Aerobic exercise prior to class may improve academic engagement (Oriel, 2011)
Aquatic therapy – water exercise swimming program can improve swimming ability and has potential for improving social skills (Pan, 2010).
Toe walking is common in children with ASD; cause unclear; likely at least partially a response to abnormal sensory processing. If toe walking becomes habitual and persistent, it is important not to let the gastro-soleus tendon shorten. Stretching exercises, serial casting and splinting may all be required to prevent or alleviate structural deformity (Barkocy, 2017)
Intervention for sensory processing disorder – focus self-regulation (Baranek, 2002)
Evaluation: Sensory Profile 2 (Dunn, 2014) based on teacher or caregiver interview; appropriate for infants/children from birth to 14 years 11 months (separate profiles for three different age spans)
Sensory integration therapy – "clinic-based interventions that use sensory-rich, child-directed activities to improve a child's adaptive responses to sensory experiences" (Case-Smith, 2015)
Sensory-based interventions – "classroom-based interventions that use single-sensory strategies, for example, weighted vests or therapy balls, to influence a child's state or arousal" (Case-Smith, 2015)
Music therapy involving both active, improvisational methods and receptive music therapy approaches; evidence to support improved motivation, communication skills, social interaction and attention (Wigram, 2006).
+
American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. 5th ed. Arlington, VA: American Psychiatric Association; 2013.
+
Baranek
GT. Efficacy of sensory and motor interventions for children with autism. J Autism Dev Dis. 2002;32(5):397–422.
+
Barkocy
M, Dexter
J, Petranovich
C. Kinematic gait changes following serial casting and bracing to treat toe walking in a child with autism. Peds Phys Ther. 2017;29(3):270–274.
+
Bolte
S. The social communication questionnaire (SCQ) as a screener for autism spectrum disorders: additional evidence and cross-cultural validity. J Am Acad Child Adolesc Psych. 2008;47:719–720.
+
Bruininks
RH, Bruininks
BD. Bruininks-Oseretsky Test of Motor Proficiency. 2nd ed. Minneapolis, MN: London, UK: Pearson Assessment; 2005.
+
Case-Smith
J, Weaver
LL, Fristad
MA. A systematic review of sensory processing interventions for children with autism spectrum disorders. Autism. 2015;19(2):133–148.
+
Centers for Disease Control and Prevention, Division of Birth Defects, National Center on Birth Defects and Developmental Disabilities.
Autism Spectrum Disorder (ASD).
https://www.cdc.gov/ncbddd/autism/index.html. Updated May 9, 2017. Accessed October 12, 2017.
+
Centers for Disease Control and Prevention, Division of Birth Defects, National Center on Birth Defects and Developmental Disabilities, Autism Spectrum Disorder, Study to Explore Early Development (SEED).
https://www.cdc.gov/ncbddd/autism/seed.html. Updated September 6, 2017. Accessed November 1, 2017.
+
Curtin
C, Anderson
SE, Must
A, Bandini
L. The prevalence of obesity in children with autism: a secondary data analysis using nationally representative data from the National Survey of Children's Health. BMC Pediatr. 2010;10:11.
+
Daniels
AM, Halladay
AK, Shih
A, Elder
LM, Dawson
G. Approaches to enhancing the early detection of autism spectrum disorders: a systematic review of the literature. J Am Acad Child Adolesc Psychiatry. 2014;53(2):141–152.
+
Downey
R, Rapport
MK. Motor activity in children with autism: a review of current literature. Peds Phys Ther. 2012;24(1):2–20.
+
Dumont-Mathieu
T, Fein
D. Screening for autism in young children: the Modified Checklist for Autism in Toddlers (M-CHAT) and other measures. Ment Retard Dev Disabil Res Rev. 2005;11(3):253–262.
+
Dunn
W. Sensory Profile 2. San Antonio, TX: Pearson Clinical; 2014.
+
Enticott
PG, Kennedy
HA, Rinehart
NJ,
et al. Mirror neuron activity associated with social impairments but not age in autism spectrum disorder. Biol Psychiatry. 2012;71(5):427–433.
+
Farley
MA, McMahon
WM, Fombonne
E,
et al. Twenty-year outcome for individuals with autism and average or near-average cognitive abilities. Autism Res. 2009;2(2):109–118.
+
Fournier
KA, Hass
CJ, Naik
SK, Lodha
N, Cauraugh
JH. Motor coordination in autism spectrum disorders: a synthesis and meta-analysis. J Autism Dev Disord. 2010;40(10):1227–1240.
+
Gkotzia
E, Venetsanou
F, Kambas
A. Motor proficiency of children with autism spectrum disorders and intellectual disabilities: a review. Eur Psychomotricity J. 2017;9(1):46–69.
+
Henderson
SE, Sugden
DA, Barnett
AL. Movement Assessment Battery for Children- 2: Examiner's Manual. London, UK: Pearson Assessment; 2007.
+
Johnson
CP, Meyers
SM. Identification and evaluation of children with autism spectrum disorders. Pediatrics. 2007;120:1183–1215.
+
Just
MA, Keller
TA, Malave
VL, Kana
RK, Varma
S. Autism as a neural systems disorder: a theory of frontal-posterior underconnectivity. Neurosci Biobehav Rev. 2012;36(4):1292–1313.
+
Lang
R, Koegel
LK, Ashbaugh
K, Regester
A, Ence
W, Smith
W. Physical exercise and individuals with autism spectrum disorders: a systematic review. Res Autism Spectr Disord. 2010;4(4):565–576.
+
Levy
SE, Giarelli
E, Lee
LC,
et al. Autism spectrum disorder and co-occurring developmental, psychiatric, and medical conditions among children in multiple populations of the United states. J Dev Behav Pediatr. 2010;31(4):267–275.
+
Oriel
KN, George
CL, Peckus
R, Semon
AL. The effects of aerobic exercise on academic engagement in young children with autism spectrum disorder. Peds Phys Ther. 2011;23(2):187–193.
+
Pan
CY. Effects of water exercise swimming program on aquatic skills and social behavior in children with autism spectrum disorders. Autism. 2010;14:9–28.
+
Roley
SS, Mailloux
Z, Parham
LD, Schaaf
RC, Lane
CJ, Cermak
S. Sensory integration and praxis patterns in children with autism. Am J Occup Ther. 2015;69(1):6901220010 [online only: doi: 10.5014/ajot.2015.012476]
+
Rutter
M, Bailey
A, Lord
C. The Social Communication Questionnaire. Los Angeles: Western Psychological Services; 2003a.
+
Rutter
M, LeCouteur
A, Lord
C. Autism Diagnostic Interview, Revised. Los Angeles, Western Psychological Services; 2003b.
+
Sowa
M, Meulenbroek
R. Effects of physical exercise on autism spectrum disorders: a meta-analysis. Res Autism Spectr Disord. 2012;6:46–57.
+
Srinivasan
SM, Pescatello
LS, Bhat
AN. Current perspectives on physical activity and exercise recommendations for children and adolescents with autism spectrum disorders. Phys Ther. 2014;94(6):875–889.
+
Stone
WL, Coonrod
EE, Ousley
OY. Brief report: Screening Tool for Autism in Two-year-olds (STAT): development and preliminary data. J Autism Dev Disord. 2000;30(6):607–612.
+
Tammimies
K, Marshall
CR, Walker
S,
et al. Molecular diagnostic yield of chromosomal microarray analysis and whole-exome sequencing in children with autism spectrum disorder. J Am Med Assoc. 2015;314(9):895–903.
+
Tomchek
SD, Huebner
RA, Dunn
W. Patterns of sensory processing in children with an autism spectrum disorder. Res Autism Spectr Disord. 2014;8:1214–1224.
+
Ulrich
DA. Test of Gross Motor Development. 2nd ed. Austin, TX: PRO-ED; 2000.
+
Wigram
T, Gold
C. Music therapy in the assessment and treatment of autistic spectrum disorder: clinical application and research evidence. Child Care Health Dev. 2006;32(5):535–542.
+
Zwaigenbaum
L, Bauman
ML, Stone
W,
et al. Early identification of autism spectrum disorders: Recommendations for practice and research. Pediatrics. 2015;136(suppl 1)
++
Acute idiopathic, acute peripheral-nerve palsy involving the facial nerve (CNVII)
Caused by inflammation of CN VII at geniculate ganglion, leading to nerve compression and possible ischemia and demyelination of CN VII (Tiemstra, 2007).
Bell's Palsy has a strong association with infectious agents. Infectious agent prompts the initial step of a cascade of facial nerve inflammation, degeneration, and paralysis (Morgan, 1992).
Results in unilateral facial paralysis or weakening (Gilden, 2008).
Etiology is unknown
Possibly dormant viral infections of Herpes Simplex/Herpes Zoster (Morgan, 1992; Holland, 2004; Stjernquist-Desatnik, 2006).
The reactivation of a latent herpes simplex virus is the most likely cause of Bell's palsy, but it often diagnosed by ruling out other diagnoses (Zandian, 2014).
Herpes simplex virus (HSV-1) and varicella-zoster virus (VZV) were thought to provoke an autoimmune response leading to demyelination of cranial nerves, particularly the facial nerve. However, the role of HSV-1 and VZV in causing Bell's palsy is now unclear. Researchers are now considering HHV-6 (high levels in the saliva of patients with Bell's palsy) in the pathogenesis of Bell's palsy. (Turriziani, 2014).
Rapid onset Bell's palsy can be due to Lyme's disease, autoimmune disease, neoplasms, or can be viral. (Hohman, 2013)
Structural lesions of ear or parotid gland, cholesteatoma, salivary tumors, Lyme disease, otitis media, Ramsay Hunt Syndrome, sarcoidosis, and some influenza vaccines may present as isolated facial nerve palsies; Diabetes Mellitus is common and may be contributing factor (Tiemstra, 2007).
Peripheral facial palsy can be caused by neoplastic meningitis, with symptoms and MRI findings mimicking those of benign facial palsy (Hiraumi, 2014).
May present as one form of a neural manifestation in individuals with Human Immunodeficiency Virus infection (Morgan, 1992; Alakram, 2008).
May be related to mumps virus, rubella virus, HIV, and B. burgdorferi (Morgan, 1992)
Bilateral facial paralysis is very rare and can occur during pregnancy. Preeclampsia is usually reported in the third trimester and can cause peripheral edema, which can cause compression of the facial nerve, and subsequent paralysis. (Vogell, 2014).
Previous use of Statin drugs has been suggested to be associated with the development of Bell's Palsy. (Hung, 2014).
Glycated hemoglobin levels (HbAlc) correlate with the severity of Bell's Palsy, although it does not seem to affect prognosis (Riga, 2012).
Prevalence and Incidence
Equal occurrence in either gender and at any age but most common between ages 15-45 with peak incidence in 40's (Holland, 2004; Gilden, 2004).
Incidence of 20-30 cases per 100,000 people per year (Holland, 2004; Gilden, 2004). 1 in 5000 people worldwide (Alakram, 2008)
Patients with DM and pregnancy have increased incidence (Cohen, 2000).
While seasonal and monthly distributions of Bell's palsy do not differ significantly, it has been suggested that stronger wind speeds on the preceding day may be related to the occurrence of Bell's palsy (Jeon, 2013).
++
Acute unilateral facial paralysis/weakness with onset of 1-3 day resulting in incomplete eyelid closure with dry eye, facial drooping, diminished tearing, drooling, and thicker saliva (Holland, 2004; Tiemstra, 2007).
Postauricular pain, dry eye, dysgeusia (disordered taste), dry mouth, facial pain, hyperlacrimation, aural fullness, and hyperacusis (on the affected side) are some of the symptoms that can precede and accompany facial paresis. (De Sata, 2014)
Unable to smile, fill cheeks with air, or wrinkle forehead on affected side.
Hyperacusis (hypersensitivity to sound) and/or altered taste (Holland, 2004).
Bell's Phenomenon (upward diversion of eye with attempted eyelid closure) (Holland, 2004, Tiemstra, 2007).
A high Neutrophil-to-Lymphocyte ratio (NLR) was shown to be a reliable predictor of Bell's palsy (Bucak, 2014).
Red flag symptoms that indicate other diagnoses include diplopia, dysphagia, numbness of the face, or dizziness (Phan, 2016).
++
Clinical diagnosis of exclusion (Gilden, 2008; Hohman, 2013). The objective of the history and examination is to rule out an infectious, neurologic, otologic, or neoplastic cause (Phan, 2016).
History of onset, progression, and detailed PMH (Tiemstra, 2007).
The first step in the diagnosis is to determine whether facial weakness has a central or peripheral etiology. (Murthy, 2011)
Currently the trigeminal blink reflex is the only test to measure intracranial pathway of the facial nerve and also useful test to study various postparalysis sequelae such as synkinesis and hemifacial spasms. (Murthy, 2011)
Physical examination and inspection of the ear canal, tympanic membrane, and oropharynx (Tiemstra, 2007).
Peripheral nerve testing of extremities, evaluation of cranial nerves and facial muscles (Tiemstra, 2007).
Imaging:
CT, MRI (indicated for insidious onset, central weakness, trauma) (Tiemstra 2007, Gilden 2004).
Laboratory tests:
Electrodiagnostic Test:
House-Brackmann Facial Nerve Grading Scale Facial Disability Inventory (Alakram, 2008)
"Has poor evaluation quality for measuring severe facial nerve palsy, and is suitable for patients with mild facial nerve injury. There are six classifications for grading facial nerve injury: normal, mild, moderate, moderate-severe, severe dysfunction, and complete paralysis" (Huang, 2014).
Electrical response grading is a newer method for assessing the degree of facial nerve injury that is superior to the House-Brackmann scale for measuring the degree of facial nerve palsy and is superior in efficacy and reliability. "The electrical response grading system is divided into 4 grades based on a muscle-specific response to electroacupuncture. The grades and output intensities include: excellent (4 mA with facial muscle twitching and motion of facial muscle reaches or exceeds normal motion), moderate (4-6 mA with muscle twitching and slight muscle contraction, but does not reach normal range of motion), poor (>6 mA with slight twitch, and no contraction), and no response (>6 mA with no muscle activity)" (Huang, 2014).
A complete blood count with high Neutrophil to Lymphocyte Ratio (NLR) is supported as a good indicator of Bell's Palsy in pediatrics; However, this value cannot determine the severity of the disease. (Eryilmaz, 2015).
+++
Differential Diagnosis: (Tiemstra,2007)
++
Nuclear (peripheral)
Lyme disease: Tick exposure, rash, arthralgias, exposure to endemic Lyme disease
Otitis media: Bacterial pathogens, gradual onset; ear pain, fever, conductive hearing loss
Ramsay Hunt syndrome: Herpes zoster, pronounced pain; vesicular eruption in ear canal or pharynx
Sarcoidosis or Guillain-Barré syndrome: Autoimmune response, more often bilateral
Tumor: Cholesteatoma or parotid gland, gradual onset
Supranuclear (central) forehead motor function less affected because of bilateral cortical control of forehead muscles
Multiple sclerosis: Demyelination, neurological symptoms
Stroke, Ischemic or hemorrhagic:, extremities on affected side are often involved, while the frontalis muscle on the affected side is not involved (Mooney, 2013). Stroke often includes changes in level of consciousness, dizziness, loss of coordination, or changes in vision. (Mayhew, 2015).
Tumor: Metastases or primary brain, gradual onset; mental status changes; history of cancer
People are often misdiagnosed with Bell's palsy, versus actual diagnosis of stroke, herpes zoster, Guillain-Barre and otitis media. Emergency rooms have a very low rate of misdiagnosis. This low rate is, in part, due to CT and MRI use. "Increasing age and diabetes are modest risk factors for misdiagnosis" (Fahimi, 2014).
If facial nerve palsy continues without improvement over 3 months, it is recommended the patient have a high-resolution MRI, perilesional biopsies, or direct nerve biopsy to determine the correct diagnosis and rule out microcystic adnexal carcinoma-like squamous cell carcinoma or other neoplastic etiology (Mueller, 2017).
++
Non-Progressive and Self-Limiting
Spontaneous recovery in 85% of patients within 3 weeks; most recover completely in 3 months (Tiemstra, 2007).
Small population of patients may never completely recover. Paralysis considered permanent after 6 months (Holland, 2004).
For complete involvement: 75% recover motor function (Holland, 2004).
Age and degree of facial paralysis are prognostic factors, with recovery of symptoms in 21 days predicting a favorable prognosis (Zandian, 2014).
Severity of paralysis, partial or complete, may be predictive:
94% of patients who show incomplete paralysis of the face will restore normal function without treatment, and only 30% of patients with complete paralysis will show no recovery. However, at the end of 1 month if paralysis is still complete, chance of recovery is 10-40%, and after 2 months, the predicted chance of full recovery falls to 0%. (Li Y, 2015).
Poor outcomes are associated with:
Age >60, systemic comorbidities, and a lesion with autonomic involvement (Cedarwall, 2006).
Decreased blink reflex and electroneurography are the best predictors of residual palsy and no facial function recovery. These tests, along with clinical findings, can be used as assessment tools in the early post-palsy period (Mancini, 2014).
A high Neutrophil-to-Lymphocyte ratio (NLR) is associated with nonsatisfactory recovery in patients with Bell's palsy (Bucak, 2014).
Bilateral paralysis has a poor prognosis, with increased risk of permanent damage, and may be the first sign of preeclampsia in pregnant women (Vogell, 2014).
Dry mouth at onset was correlated with severe grade palsy and poor prognosis (De Sata, 2014).
Crocodile tears (facial mm contract resulting in tear formation) can occur during recovery for up to 1 year
If untreated, the prognosis of pregnancy related Bell's Palsy may have a better prognosis than for patients with non-pregnancy related palsy (Sax, 2006).
The Yanagihara system can be used to diagnose the severity of Bell's palsy. Using this system to diagnose patients can lead to prompt treatment and a better overall prognosis (Hato, 2014).
"Electroneugrophy and House Brackmann grading during the first to fourth weeks of Bell's Palsy are useful prognostic indicators.
Serial electroneurography (ENoG) examinations are recommended to predict the status of neural degeneration and the prognosis of the palsy" It is best to use ENoG in early periods of paralysis in order to determine the most accurate state of the paralysis, maximum denervation, and any residual nerve function. "The correlation between facial function and ENoG value begins to decrease in late periods of paralysis" (Arslan, 2014).
+++
IV. Medical/Surgical Management
+++
Pharmacologic Options:
++
Bell's palsy is the leading cause of facial nerve palsy, with corticosteroids being the drug of choice when medical therapy is needed (Zandian, 2014).
Combined use of Prednisone and Acyclovir very effective if given within 72 hours of onset (Holland, 2004; Sullivan 2007; Gilden 2008).
High-dose corticosteroids for 10 days; prevents permanent damage, begin right away
Corticosteroids show clinical and statistical significant recovery of motor function of facial nerve (Ramsey, 2000).
Steroids are likely to be effective and should be offered to increase the probability of recovery of facial nerve function (Gronseth, 2012), and reduce the duration of symptoms (Albers, 2014)
Corticosteroids are the drug of choice for treating Bell's palsy. However, this drug class can be problematic for those patients with diabetes mellitus. Stellate ganglion blocks have been a successful method for treating patients who have diabetes mellitus and Bell's palsy (Liu, 2014).
The clinical guideline for Bell's Palsy by the American Academy of Otolaryngology– Head and Neck Surgery recommends a 10-day regimen of oral steroids with at least 5 days at a high dose. Two options are given: 1) 10 days of Prednisolone at 50 mg or 2) 5 days of Prednisone at 60 mg with a following 5-day taper. This is best done within 72 hours of onset (Baugh, 2013).
Antibiotics/anti-virals fight bacteria/viruses, analgesics for pain
Steroids and antiviral agents have been used to treat Bell's palsy. A combination of the two has shown higher recovery rates in those with initially severe Bell's palsy and those who do not have diabetes mellitus or hypertension (Kang, 2014), and is more effective than steroids alone (Lee, 2013).
In one systematic review, moderate quality evidence supported no significant difference between the use of corticosteroid with antivirals and corticosteroids only, or corticosteroids and a placebo, or the use of only antivirals compared to placebo in treating Bell's Palsy. (Gagyor, 2015).
Studies in children do not support the use of steroids or antivirals (Sanders, 2004).
"Intratympanic steroid injection might be a safe and useful adjuvant treatment modality for Bell's palsy" (Chung, 2014).
++
Decompression surgery of facial nerve has high risk of complications and is generally not recommended (Holland 2004; Tiemstra, 2007) Permanent cases of facial symmetry and muscle contracture may require cosmetic surgery (Tiemstra, 2007)
Some surgeons agree that surgical decompression of the facial nerve will help to release the entrapment of the nerve through the facial canal. This surgery should occur early after the onset of full paralysis (2-3 weeks) in the patients who have poor prognosis for full recovery. If the surgery occurs more than 2 months after onset of full paralysis, it is possible that frequency of adverse effects matches the benefits. Therefore, decompression surgery after two months of onset would not benefit the patient. (Li , 2015).
In one study: only half of otologist and neurologist think surgery decompression should be the standard for patients with an electroneuronography < 10% normal (Smouha, 2010).
Plastic surgery using facial slings may be used to replace active muscle contraction
To protect cornea, wear eye patch or glasses, use of artificial tears, and heat or massage can be used
Botox injections have been shown to improve facial symmetry at rest and during movement, by reducing facial synkinesis (Toffola, 2010).
+++
V. Implications for Therapeutic Management
++
Practice pattern (APTA, 2017)
5D: Impaired Motor Function and Sensory Integrity Associated With Nonprogressive Disorders of the Central Nervous System-Acquired in Adolescence or Adulthood; 10-60 visits per episode of care
4E: Impaired joint mobility, motor function, muscle performance, and range of motion associated with localized inflammation
5F: impaired peripheral nerve integrity and muscle performance associated with peripheral nerve injury
Physical Therapy
No evidence of significant benefit or harm from any physical therapy for idiopathic facial paralysis. (Teixeira, 2008; 2011)
For patients with idiopathic facial palsy who remain symptomatic for over a year, facial physiotherapy assists in improving facial grading scores. (Watson, 2015)
Physical therapy has only been shown to be effective in those with severe Bell's palsy, while those with less severe palsy usually recover on their own (Nicastri, 2014).
Physical therapies, such as exercise, biofeedback, laser, electrotherapy, massage and thermotherapy are used to hasten recovery. However, evidence supporting the effectiveness of the previously mentioned therapies is lacking (Murthy, 2011)
There is low quality evidence that tailored facial exercises can help to improve facial function, mainly for people with moderate paralysis and chronic cases. (Teixeira, 2011)
There is low quality evidence that facial exercise reduces sequelae in acute cases. (Teixeira, 2011)
A significant and long lasting effect of facial function can be achieved through using education, neuromuscular training, massage, meditation-relaxation, and an individualized home program. (Lindsay, 2009)
Individualized facial neuromuscular re-education is more effective in improving facial symmetry in patients with Bell's palsy than conventional therapeutic measures (including electrical stimulation). (Manikandan, 2007)
To promote facial muscle control and function HEP should consist of a twice a day exercise program emphasizing facial movement
Local superficial moist heat for pain, tissue extensibility, circulation (Shafshak, 2006); ultrasound (Shafshak, 2006).
Mime therapy and facial exercises with biofeedback (mirror or EMG) to promote muscle reeducation (Shafshak,2006; Beurskens, 2003)
If the patient cannot fully close the affected eye, the physical therapist should implement eye protection and educate the patient (Baugh, 2013).
Electrical Stimulation
Electrical stimulation in patients with Bell's palsy is safe but may not be any better than spontaneous recovery (Alakram, 2010)
The use of electrical stimulation, shortly after palsy onset (4 weeks), has been shown to be beneficial when performed three times a week. Using this modality has improved functional facial movements and electrophysiologic outcome measures in patients with Bell's palsy (Tuncay, 2014).
Some research indicates that E-stim causes damage and delayed healing while other research suggests a positive effect in preventing muscle atrophy and encouraging neuromuscular reeducation (Shafshak, 2006)
Electrical stimulation to the facial muscles during the early phase of Bell's palsy was no more effective than a control group who did not receive electrical stimulation. (Alakram, 2010)
High intensity laser therapy and low level laser therapy can both be used to treat patients with Bell's palsy, with high intensity showing greater improvements (Alayat, 2014).
Gentle massage—circulation, prevention of contractures (Shafshak, 2006)
Acupuncture is superior to massage and methylcobalamin, but the benefits of acupuncture have not been fully determined. (Wang, 2012)
+++
VI. Consumer and Professional Resources
++
National Institute of Health
Bell's palsy Information Site
Bell's palsy Support Group
Bell's palsy Support Group
Bell's Palsy Information Site
National Institute of Neurological Disorders and Stroke
Facial Paralysis Institute
+
Alakram
P, Puckree
T. Assessment, diagnosis and prognosis of Bells Palsy: a literature review. S AfrJ Physiother. 2008; 64(2): 18–21.
+
Alakram
P, Puckree
T. Effects of electrical stimulation on House-Brackmann scores in early Bell's palsy. Physiother Theory Pract. 2010; 26(3): 160–166.
+
Alayat
MSM, Alsodany
AM, El Fiky
AAR. Efficacy of High and Low Level Laser Therapy in the Treatment of Bell's Palsy: A Randomized Double Blind Placebo-Controlled Trial. Lasers in Medical Science. 2014;29(1):335–42.
+
Albers
JR, Tamang
S. Common Questions About Bell's Palsy. Am Fam Physician. (3):209–212.
+
Arslan
HH, Satar
B, Yildizoglu
U, Edizer
DT, Akgun
H. Validity of Late-Term Electroneurography in Bell's Palsy. Otology & Neurotology. 2014;35(4):656–61.
+
Baugh
R, Basura
G, Ishii
L
et al.. Clinical Practice Guideline: Bell's Palsy. Otolaryngol Head Neck Surg. 2013;149(3S):S1–S27.
+
Beurskens
CH, Heymans
PG. Positive effects of mime therapy on sequelae facial paralysis: stiffness, lip mobility, and social and physical aspects of facial disability. Otol Neurol. 2003; 24:677–81.
+
Bucak
A, Ulu
S, Yucedag
F, Tekin
MS, Karakaya
F. Neutrophil-to-Lymphocyte Ratio as a Novel-Potential Marker for Predicting Prognosis of Bell's Palsy. Laryngoscope, 2013: 123(12).
+
Bucak
A, Ulu
S, Oruc
S, Yucedag
F, Tekin
MS, Karakaya
F, Aycicek
A. Neutrophil-to-Lymphocyte Ratio as a Novel-Potential Marker for Predicting Prognosis of Bell's Palsy. Laryngoscope. 2014;124(7):1678–81.
+
Cederwall
E, Olsen
M, Hanner
P,
et al: Evaluation of a physiotherapeutic treatment intervenion in "Bell's" facial palsy, Physio Theory Pract 22:43–52, 2006.
+
Chung
JH, Park
CW, Lee
SH, Kim
BS, Cho
SH, Kim
HY, Kim
SH. Intratympanic Steroid Injection for Bell's Palsy: Preliminary Randomized Controlled Study. Otology & Neurotology. 2014;35(9):1673–78.
+
Cohen
Y, Lavie
O, Granovsky-Grisaru
S,
et al: Bell palsy complicating pregnancy. A review, Obstet Gynecol Surv 55:184–188, 2000.
+
De Seta
D, Mancini
P, Minni
A, Prosperini
L, De Seta
E, Attanasio
G, Covelli
E, De Carlo
A, Filipo
R. Bell's Palsy: Symptoms Preceding and Accompanying the Facial Paresis. The Scientific World Journal. 2014; 2014. doi:10.1155/2014/801971
+
Eryilmaz
A, Basal
Y, Tosun
A, Omurlu
IK, Basak
S. The neutrophil to lymphocyte ratios of our pediatric patients with Bell's palsy. Int J Pediatr Otorhinolaryngol. 2015;79(12):2374–2377.
+
Fahimi
J, Navi
BB, Kamel
H. Potential Misdiagnoses of Bell's Palsy in the Emergency Department. Annals of Emergency Medicine. 2014;63(4):428–34.
+
Fujiwara
T, Hato
N, Gyo
K, Yanagihara
N. Prognostic Factors of Bell's Palsy: Prospective Patient Collected Observational Study. European Archives of Oto-Rhino-Laryngology. 2014;271(7):1891–95.
+
Gagyor
I, Madhok
VB, Daly
F,
et al. Antiviral treatment for Bell's palsy (idiopathic facial paralysis). Cochrane Database Syst Rev. 2015;11:CD001869.
+
Gilden
DH. Bell's palsy. New England Journal of Medicine. 2004; 351(13).
+
Gilden
D. Treatment of Bell's palsy –the pendulum has swung back to steroids alone. The Lancet Neurology. 2008: 7(11): 976–77.
+
Gronseth
GS, Paduga
R. Evidence-based guideline update: steroids and antivirals for Bell palsy: report of the Guideline Development Subcommittee of the American Academy of Neurology. Neurology. 2012 Nov 27;79(22):2209–13.
+
Hato
N, Fujiwara
T, Gyo
K, Yanagihara
N. Yanagihara Facial Nerve Grading System as a Prognostic Tool in Bell's Palsy. Otology & Neurotology. 2014;35(9):1669–72.
+
Hiraumi
H, Yamamoto
N, Sakamoto
T, Ito
J. Peripheral Facial Palsy Caused by Neoplastic Meningitis. The Laryngoscope. 2014;124(9):2139–43.
+
Hohman
MH, Hadlock
TA. Etiology, Diagnosis and Management of Facial Palsy: 2000 Patients at a Facial Nerve Center. Laryngoscope. 2014: 124(1).
+
Holland
NJ, Weiner
GM. Recent developments in bell's palsy. British Medical Journal. 2004; 329(7465):553–7.
+
Huang
B, Zhou
ZL, Wang
LL, Zou
C, Lu
Y, Chen
Y. Electrical response grading versus House-Brackmann scale for evaluation of facial nerve injury after Bell's palsy: a comparative study. Journal of Integrative Medicine. 2014;12(4):367–71.
+
Hung
SH, Wang
LH, Lin
HC, Chung
SD. Association between statin use and Bell's palsy: a population-based study. Drug Saf. 2014;37(9):735–42.
+
Jeon
EJ, Park
YS, Kim
DH, Nam
IC, Park
SY, Noh
H, Yeo
SW. Effects of Meteorological Factors on the Onset of Bell's Palsy. Auris, Nasus, Larynx. 2013;40(4):361–65.
+
Kang
HM, Jung
SY, Byun
JY, Park
MS, Yeo
SG. Steroid plus Antiviral Treatment for Bell's Palsy. Journal of Internal Medicine. 2014. doi:10.1111/joim.12288
+
Lee
HY, Byun
JY, Park
MS, Yeo
SG. Steroid-Antiviral Treatment Improves the Recovery Rate in Patients with Severe Bell's Palsy. The American Journal of Medicine. 2013;126(4):336–41.
+
Li
Y, Sheng
Y, Feng
GD, Wu
HY, Gao
ZQ. Delayed surgical management is not effective for severe Bell's palsy after two months of onset. Int J Neurosci. 2015; 0(0): 1–7. Doi: 10.3109/00207454.2015.1092144
+
Lindsay
RW, Robinson
M, Hadlock
TA. Comprehensive Facial Rehabilitation Improves Function in People With Facial Paralysis: A 5-Year Experience at the Massachusetts Eye and Ear Infirmary. Phys Ther. 2010;90(3):391–397.
+
Liu
GD, He
CJ. Stellate Ganglion Block Promotes Recovery of Bell's Palsy in Patients with Diabetes Mellitus. Acta Oto-Laryngologica. 2014;134(6):652–55.
+
Mancini
P, De Seta
D, Prosperini
L, Nicastri
M, Gabriele
M, Ceccanti
M, Sementilli
G,
et al. Prognostic Factors of Bell's Palsy: Multivariate Analysis of Electrophysiological Findings. The Laryngoscope. 2014; 124(11):2598–2605.
+
Manikandan
N. Effect of facial neuromuscular re-education on facial symmetry in patients with Bell's palsy: a randomized controlled trial. Clin Rehabil 2007; 21(4): 338–343.
+
Mayhew
G, Carhart
E. Differential Diagnosis: Bell's Palsy vs. Stroke. They can look similar, but it's important to tell them apart. EMS World. 2015;44(9):47–53.
+
Mooney
T. Diagnosis and management of patients with Bell's palsy. Nursing Standard. December 4, 2013;28(14):44–49.
+
Murthy
J, Saxena
AB. Bell's palsy: Treatment guidelines. Ann Indian Acad Neurol 2011;14:70–2.
+
Morgan
M, Nathwani
D. Facial Palsy and Infection: The Unfolding Story. Clinical Infectious Diseases. 1992;14(1):263–271.
+
Mueller
S, Iro
H, Lell
M
et al.. Microcystic adnexal carcinoma (MAC)-like squamous cell carcinoma as a differential diagnosis to Bell's palsy: review of guidelines for refractory facial nerve palsy. J Otolaryngol Head Neck Surg. 2017;46(1):1–5.
+
Nicastri
M, Mancini
P, De Seta
D, Bertoli
G, Prosperini
L, Toni
D, Inghilleri
M, Filipo
R. Efficacy of Early Physical Therapy in Severe Bell's Palsy: A Randomized Controlled Trial. Neurorehabilitation and Neural Repair. 2013;27(6):542–51.
+
Phan
N, Panizza
B, Wallwork
B. A general practice approach to Bell's palsy. Australian Family Physician. 2017;45(11):794–797.
+
Pourmomeny
AA, Asadi
S. Management of Synkinesis and Asymmetry in Facial Nerve Palsy: A Review Article. Iranian Journal of Otorhinolaryngology. 2014;26(77):251–56.
+
Ramsey
M, DeSimonian
R, Holtel
M,
et al: Corticosteroid treatment for idiopathic facial nerve paralysis. A meta-analysis, Laryngoscope 110:335–341, 2000.
+
Riga
M, Kefalidis
G, Danielides
V. The Role of Diabetes Mellitus in the Clinical Presentation and Prognosis of Bell Palsy. J Am Board Fam Med. 2012;25(6):819–826.
+
Sanders
RJ, Hammond
SL: Supraclavicular first rib resection and total scalenectomy. Techniques and results, Hand Clin 20:61–70, 2004.
+
Sax
T, Rosenbaum
R. Neuromuscular Disorders in Pregnancy. Muscle Nerve. 2006;34:559–571.
+
Shafshak
TS. The treatment of facial palsy from the point of view of physical and rehabilitation medicine. Europa Medicophysica. 2006; 42(1):41–7.
+
Smouha
E, Toh
E, Schaitkin
B. Otology/Neurotology: Surgical Treatment of Bell's Palsy: Current Attitudes. Otolaryngology -- Head and Neck Surgery. 2010;143(5):720.
+
Stjernquist-Desatnik
A, Skoog
E, Aurelius
E. Detection of herpes simplex and varicella-zoster viruses in patients with bell's palsy by the polymerase chain reaction technique. Annals of Otology, Rhinology and Laryngology. 2006;115(4):306–11.
+
Sullivan
FM, Swan
IRC, Donnan
PT,
et al. Early treatment with prednisolone or acyclovir in bell's palsy. New England Journal of Medicine. 2007;357(16):1598–607.
+
Teixeira
LJ, Soares
BGO, Vieira
VP, Prado
GF. Physical therapy for Bells palsy (idiopathic facial paralysis). Cochrane Database Syst Rev), 2008(3)
+
Teixeira
LJ, Valbuza
JS, Prado
GF. Physical therapy for Bell's palsy (idiopathic facial paralysis). Cochrane Database Syst Rev. 2011 Dec 7;(12):CD006283.
+
Tiemstra
JD, Khatkhate
N. Bell's palsy: diagnosis and management. American Family Physician. 2007; 76(7).
+
Toffola
ED, Furini
F, Redaelli
C, Prestifilippo
E, Bejor
M. Evaluation and treatment of synkinesis with botulinum toxin following facial nerve palsy. Disability & Rehabilitation. 2010;32(17):1414–1418.
+
Tuncay
F, Borman
P, Taşer
B, Unlü
I, Samim
E. Role of Electrical Stimulation Added to Conventional Therapy in Patients with Idiopathic Facial (Bell) Palsy. American Journal of Physical Medicine & Rehabilitation / Association of Academic Physiatrists. 2014. doi:10.1097/PHM.0000000000000171
+
Turgeon
RD, Wilby
KJ, Ensom
MH. Antiviral Treatment of Bell's Palsy Based on Baseline Severity: A Systematic Review and Meta-Analysis. The American Journal of Medicine. 2014. doi:10.1016/j.amjmed.2014.11.033.
+
Turriziani
O, Falasca
F, Maida
P, Gaeta
A, De Vito
C, Mancini
P, De Seta
D, Covelli
E, Attanasio
G, Antonelli
G. Early Collection of Saliva Specimens from Bell's Palsy Patients: Quantitative Analysis of HHV-6, HSV-1, and VZV. Journal of Medical Virology. 2014;86(10):1752–58.
+
Vogell
A, Boelig
RC, Skora
J, Baxter
JK. Bilateral Bell Palsy as a Presenting Sign of Preeclampsia. Obstetrics and Gynecology. 2014;124(2 Pt 2 Suppl 1):459–61.
+
Wang
Q, Bai
M, Lei
D. Effectiveness of acupuncture in treatment of facial spasm: a meta-analysis. Alternative Therapies In Health & Medicine. May 2012;18(3):45–52.
+
Watson
GJ, Glover
S, Allen
S, Irving
RM. Outcome of facial physiotherapy in patients with prolonged idiopathic facial palsy. J Laryngol Otol. 2015;129(4):348–52.
+
Zandian
A, Osiro
S, Hudson
R,
et al. The neurologist's dilemma: A comprehensive clinical review of Bell's palsy, with emphasis on current management trends. Medical Science Monitor. 2014;20:83–90.
+++
Benign Paroxysmal Positional Vertigo (BPPV)
++
The most common peripheral vestibular pathology, representing ~ 50% of all cases of vertigo
Otoconia in the utricle are dislodged, fragment, and migrate into semi-circular canal(s). Calcium carbonate particles free-float in endolymph (canalithiasis) or adhere to cupula (cupulolithiasis), causing disruption of endolymph flow and cupular deflection (Furman, 1999; Strupp, 2013) Creates abnormal action potentials and confusing vestibular signals (Furman, 1999; Strupp, 2013). Results in episodes of vertigo characterized by brief spinning sensations, often triggered by specific changes in head position (Oh, 2014; Kim, 2014)
Canalithiasis of the posterior semicircular is the most frequent cause of BPPV (Lee, 2010)
Posterior semicircular canal (the most gravity-dependent canal) BPPV is most common (60-90% of cases), while lateral or horizontal (10-30% of cases) and anterior canal (3% of cases) are less common (Baloh, 1993; von Brevern, 2013; Anagnostou, 2015)
The right ear is affected more often than the left, often due to sleeping position of individuals (De Sousa Oliveira, 2013; Babac, 2014)
Incidence
Approximately 1/10,000 people (Mizukoshi, 1988), increases with age; mean age of onset at 54 y.o. (von Brevern, 2007; Bhattacharyya, 2008)
BPPV occurs more often in females, with a 2:1-3:1 ratio of female to males, possibly suggesting that hormones may play a role (Pisani, 2015; Ogun 2014)
BPPV can occur at any age, but occurrence does increase with age (Strupp, 2013; von Brevern, 2007; Baloh, 1987)
Actual cause of BPPV unknown.
Risk factors:
Head injury, ear injury, ear infection, ear surgery, degeneration of inner ear structures, vestibular neuritis, Meniere's disease, nasal allergies, menopause, decreased Vitamin D intake, low bone mineral density, and any jolt or strenuous action that could cause the otoconia to break free (Mandala, 2012; Strupp, 2013; Talaat, 2014; Ogun, 2014).
BPPV following both major and minor head trauma is most often bilateral, involves multiple canals, and in younger individuals (Pisani 2015)
Post traumatic BPPV accounts for about 15%-20% of cases (Pisani, 2015)
BPPV has also been found in individuals after long term bed rest (Strupp, 2013)
Can be a complication following closed sinus lift procedure using a mallet, as opposed to a screwable (Sammartino, 2011)
There is a possible link between patients with migraines developing BPPV. Patients with migraine have a 2.03 fold greater risk of developing BPPV compared to the age- and sex- matched control cohort. The relationship can partially be explained due to the vasospasm and downstream of vascular events that take place intracranially and in the inner ear during a migraine (Chu, 2015).
++
Signs/Symptoms:
Occur with head movements specific to each patient and observed with provocative testing: vertigo, lightheadedness, disequilibrium, dizziness, nausea/vomiting, difficulty concentrating, blurred vision, and nystagmus (Furman, 1999; Mayo Clinic Staff, 2010; Parnes, 2003)
Patients who report vertigo provoked by head movements should first undergo Hallpike-Dix maneuver (Kim, 2014)
Radiographic imaging not recommended unless signs unrelated to BPPV are present (Bhattacharyya, 2008)
Use of an anamnesis questionnaire based on the presence/absence of six typical features of BPPV could reduce the risk of a misdiagnosis of BPPV (Lapenna, 2015).
Provocative tests
Positional tests: place migrated otoconia in a gravity-dependent position and provoke symptoms
Hallpike-Dix test: tests for anterior or posterior semi-circular canal BPPV
Vertical and torsional nystagmus occurs with anterior/posterior canal involvement. The posterior canal most commonly affected (Korres, 2002; Huebner, 2013)
Upbeating torsional nystagmus towards uppermost ear = PSCC involvement; downbeating torsional nystagmus towards downside ear = ASCC involvement
Side-lying test: tests for posterior canal involvement
Used less often than the Hallpike-Dix test, but has been found to be equally valid and can be used if the patient does not tolerate the Hallpike-Dix maneuver (Helminski, 2014; Hilton, 2014)
Roll test (aka Paganini-McLure maneuver): tests for horizontal semi-circular canal (HC) BPPV (Parnes, 2003; Vannucchi, 1997; Kinne, 2012)
Vestibular Evoked Myogenic Potentials (VEMP)
Observation of patient's eye movements enhanced by:
Frenzel lenses: goggles magnify eye for observer, eliminates patient's ability to fixate eyes on stable target
Infrared goggle system (video nystagmography): blocks visual fixation, infrared light illuminates the eye for videography but without fixation since using non-visual spectrum (an additional visible light can be activated in the goggle to test with visual fixation), includes capability for video and replay on a large video monition and with slow motion replay.
Positive test if nystagmus observed, nausea experienced, and vertigo/dizziness occurs (Norre, 1995)
Nystagmus in 30 seconds of provocative positioning
Direction of nystagmus indicates the canal(s) affected. The nystagmus is named according to the direction of the fast component of nystagmus. So right-beating nystagmus indicates that the right side is affected. However the direction of the nystagmus does not predict the likelihood of lateralization of those with horizontal BPPV (Oh, 2014).
Duration of nystagmus indicates type of BPPV: Cupulolithiasis with nystagmus lasting >60s,canalithiasis with nystagmus lasting < 60s
Diagnosed into subgroups (posterior, horizontal or lateral, and anterior) also based on characteristics of nystagmus (torsional, vertical, horizontal, latency, crescendo, decrescendo, transience, reversibility, and fatiguability). (Korres, 2004; Balatsourus, 2012)
++
Non-progressive, usually self-limiting
Presents weeks or years with unpredictable remissions/recurrences
Patients avoid head movements that provoke symptoms (Furman, 1999)
Intractable BPPV persists despite conservative treatment
Patients with BPPV exhibit a significantly higher risk for fractures than those without BPPV secondary to falls and high ratio of osteoporosis (Wen-Ling, 2015)
Factors that suggest a poorer prognosis:
Older age, history of head trauma, long duration of symptoms, osteoporosis, and anterior canal involvement (Babac, 2014)
Patient prognosis improved with proper therapeutic/surgical management
BPPV is the most successfully treatable cause of vertigo (Von Brevern, 2007).
Treatment designed for cupulolithiasis: 70% remission of vertigo and nystagmus; another 20% had improvement of symptoms (Herdman, 1993)
Treatment designed for canalithiasis: 57% remission of vertigo and nystagmus; another 33% improvement of symptoms (Herdman, 1993)
97% of patients have resolution within three treatments of CRPs and less than 16% reoccurrence. (Balatsouras, 2012)
50-80% of patients have complete resolution with one session of either Epley's or Semont's maneuver (Herdman, 1993; Serfini, 1996). However, patient must be informed that BPPV may recur and require treatment (Kim, 2014)
For patients with posterior canal BPPV, the repeated Epley maneuver is effective in 95% of cases (Strupp, 2013)
With no intervention, BPPV has a high natural recovery rate of 60% (Strupp, 2013)
With multiple canal involvement, BPPV often takes more treatment sessions over a longer time for effective treatment (Shim, 2014; Pisani, 2015).
Positional restrictions are often given to decrease the rate of recurrence, however patients often fail to heed the sleeping restrictions, which leads to increased recurrence of their symptoms (Li, 2013).
Potential complications/negative side effects: few were reported during treatment, however nausea, imbalance, neck/back injury or debris moving into another canal can occur (De Sousa Oliveira, 2013; Hilton, 2014)
BPPV that is brought on by trauma seems to be more difficult to treat than idiopathic BPPV (Pisani, 2015).
+++
IV. Medical/Surgical Management:
++
Drug therapy: no drugs are recommended for long-term use due to high risk of vestibular damage. Drugs are used with caution to relieve symptoms of acute attacks (Hain, 2003)
Vestibular suppressants decrease symptom intensity but not attack frequency. They can be given following provocative testing to reduce symptoms: antihistamines (Antivert), anticholinergics (Scopolamine), benzodiazepines (Diazepam) (Strupp, 2013), and phenothiazines (Promethazine)
Vestibular suppressants (dimenhydrinate) treatment after canalith repositioning procedures may help reduce residual symptoms in patients who have BPPV (Kim 2014).
Surgical Management- reserved for the intractable cases of BPPV due to complications and decreased vestibular functioning
Singular Neurectomy: Sectioning posterior ampullary nerve, eliminating neural input from canal; effective, but high risk hearing loss (Gacek, 2005)
Transmastoid posterior semi-circular canal occlusion: bony plug placed in PSCC, preventing endolymph flow from stimulating the cupula. Effective, with less risk of hearing loss (Agrawal, 2005; Parnes, 1990; Beyea, 2012; Ahmed, 2012)
CO2 laser-assisted posterior semicircular canal ablation (LAPSCCA): bony plug placed in both ends of the PSCC, the CO2 laser is then used to completely section the labyrinth.
+++
V. Implications for Therapeutic Management:
++
+++
VI. Consumer and Professional Resources
++
American Physical Therapy Association, Section on Neurology
Vestibular Disorders Association
Mayo Clinic
Johns Hopkins Medicine
National Institute on Deafness and Other Communication Disorders
American Hearing Research Foundation
+
Agrawal
SK, Parnes
LS. Surgical treatment of benign paroxysmal positional vertigo. Audiol Med. 2005; 3 63–68.
+
Amor-Dorado
J, Barreira-Fernández
M, Aran-Gonzalez
I, Casariego-Vales
E, Llorca
J, González-Gay
M. Particle repositioning maneuver versus Brandt-Daroff exercise for treatment of unilateral idiopathic BPPV of the posterior semicircular canal: a randomized prospective clinical trial with short- and long-term outcome. Otol Neurotol. 2012;33(8):1401–1407.
+
Anagnostou
E, Kouzi
I, Spengos
K. Diagnosis and Treatment of Anterior-Canal Benign Paroxysmal Positional Vertigo: A Systematic Review. J Clin Neurol. 2015;11(3):262–7.
+
Appiani
G, Catania
G, Gagliardi
M. A liberatory maneuver for the treatment of horizontal canal paroxysmal positional vertigo. Otol Neurotol. 2001;22(1):66–69.
+
Asawavichianginda
S, Isipradit
P, Snidvons
K, Supiyaphun
P. Canalith repositioning for benign paroxysmal positional vertigo: A randomized controlled trial. Ear Nose Throat J. 2000; 79:732–37.
+
Balatsouras
DG. Benign paroxysmal positional vertigo with multiple canal involvement. Am J Otolaryngol Head Neck Med Surg. 2012;33:250–258.
+
Baloh
RW, Honrubia
V, Jacobson
K. Benign positional vertigo: clinical and oculographic features in 240 cases. Neurology. 1987; 37:371–8.
+
Baloh
RW, Jacobson
K, Honrubia
V. Horizontal semicircular canal variant of benign positional vertigo. Neurology 1993;43:2542–9.
+
Bhattacharyya
N, Baugh
RF, Orvidas
L, Barrs
D, Bronston
LJ, Cass
S, Chalian
AA, Desmond
AL, Earll
JM, Fife
TD, Fuller
DC, Judge
JO, Mann
NR, Rosenfeld
RM, Schuring
LT, Steiner
RW, Whitney
SL, Haidari
J. Clinical practice guideline: benign paroxysmal positional vertigo. Otolaryngol Head And Neck Surg. November 2008;139(5 Suppl 4):S47–S81.
+
Chu
C-H, Liu
C-J, Lin
L-Y, Chen
T-J, Wang
S-J. Migraine is associated with an increased risk for benign paroxysmal positional vertigo: a nationwide population-based study. J Headache Pain. 2015;16:62. doi:10.1186/s10194-015-0547-z.
+
de Sousa
Oliveira AK, Suzuki
FA, Boari
L. Is it important to repeat the positioning maneuver after the treatment for benign paroxysmal positional vertigo? Braz J Otorhinolaryngol. 2015;81(2):197–201.
+
Durga
G, Kumari
V, Madhavi
K. A Comparative Study on the effectiveness of Epleys Maneuver Over Semont Maneuver in Improving the Quality of Life in Subjects with Benign Paroxysmal Positional Vertigo. Indian J Physiother Occup Ther. January 2014;8(1):99–104.
+
Francesco
R, Francesco
D, Salvatore
G, Gautham
K, Rosalia
G, Riccardo
S. Management of benign paroxysmal positional vertigo of lateral semicircular canal by Gufoni's manoevre. Am J Otolaryngol Head Neck Med Surg. 2009;30:106–111.
+
Furman
JM, Cass
SP. Benign paroxysmal positional vertigo. N Engl J Med. 1999;341:1590–96.
+
Gacek
RR. Technique and results of singular neurectomy for the management of benign paroxysmal positional vertigo. Acta Otolaryngol. 1995; 115:154–7.
+
Hain
TC, Uddin
M. Pharmacological treatment of vertigo: therapy in practice. CNS Drugs. 2003; 17:85–100.
+
Helminski
JO. Effectiveness of the Canalith Repositioning Procedure in the Treatment of Benign Paroxysmal Positional Vertigo. Phys Ther. 2014;94(10):1373–1382.
+
Helminski
J, Zee
D, Janssen
I, Hain
T. Effectiveness of particle repositioning maneuvers in the treatment of benign paroxysmal positional vertigo: a systematic review. Phys Ther. May 2010;90(5):663–678.
+
Herdman
SJ, Tusa
RJ, Zee
DS, Proctor
LR, Mattox
DE. Single treatment approaches to benign paroxysmal positional vertigo.
Arch Otolaryngol Head Neck Surg.[Archives of Otolaryngology Full Text] 1993;119(4):450–4.
+
Hilton
MP, Pinder
DK. The Epley (canalith repositioning) manoeuvre for benign paroxysmal positional vertigo. Cochrane Database Syst Rev. 2014;12:CD003162. doi:10.1002/14651858.CD003162.pub3.
+
Huebner
A, Lytle
S, Doettl
S, Plyler
P, Thelin
J. Treatment of Objective and Subjective Benign Paroxysmal Positional Vertigo. J Am Acad Audiol. 2013;24(7):600–606.
+
Kim
JS, Zee
DS. Clinical practice. Benign paroxysmal positional vertigo. N Engl J Med. 2014;370:1138–1147.
+
Kim
M, Lee
H, Ban
J. Vestibular suppressants after canalith repositioning in benign paroxysmal positional vertigo. Laryngoscope. October 2014;124(10):2400–2403 4p.
+
Kinne
B, Strace
C, Crouch
N. The effectiveness of current interventions in the management of lateral canal benign paroxysmal positional vertigo. Phys Ther Rev. 2012;17(5):292–300.
+
Knox
GW. CO2 laser-assisted posterior semicircular canal ablation for benign paroxysmal positional vertigo. Laryngoscope. 2010;120 Suppl 4:S209.
+
Korres
S, Balatsouras, Dimitrios
G. Occurrence of semicircular canal involvement in benign paroxysmal positional vertigo. Otology & Neurology. 2002; 23:926–32.
+
Korres
SG, Balatsouras
DG. Diagnostic, pathophysiologic, and therapeutic aspects of benign paroxysmal positional vertigo. Otolaryngol Head Neck Surg. 2004;131:438–444.
+
Lapenna
R, Faralli
M, Del Zompo
M, Cipriani
L, Mobaraki
P, Ricci
G. Reliability of an anamnestic questionnaire for the diagnosis of benign paroxysmal positional vertigo in the elderly. Aging Clin Exp Res.. November 4, 2015:1–8. PMID: 26537236.
+
Lee
S-H, Kim
JS. Benign Paroxysmal Positional Vertigo. J Clin Neurol (Seoul, Korea). 2010;6(2):51–63. doi:10.3988/jcn.2010.6.2.51.
+
Li
S, Tian
L, Han
Z, Wang
J. Impact of postmaneuver sleep position on recurrence of benign paroxysmal positional vertigo. PLoS ONE. 2013;8(12):e83566. doi:10.1371/journal.pone.0083566.
+
Liao
WL, Chang
TP, Chen
HJ, Kao
CH. Benign Paroxysmal Positional Vertigo Is Associated With an Increased Risk of Fracture: A Population-Based Cohort Study. J Orthop Sports Phys Ther. May 2015;45(5):406–412 7p. Available from: CINAHL Complete, Ipswich, MA. Accessed February 2, 2016.
+
Liu
Y, Wang
W, Zhang
A, Bai
X, Zhang
S. Epley and Semont maneuvers for posterior canal benign paroxysmal positional vertigo: A network meta-analysis. Laryngoscope. 2015;126(4):951–955.
+
Longo
G, Onofri
M, Pellicciari
T, Quaranta
N. Benign paroxysmal positional vertigo: Is vestibular evoked myogenic potential testing useful? Acta Otolaryngol. 2012 Jan; 132(1):39–43.
+
Mandalà
M, Pepponi
E, Santoro
GP, Cambi
J, Casani
A, Faralli
M, Giannoni
B, Gufoni
M, Marcelli
V, Trabalzini
F, Vannucchi
P, Nuti
D. Double-blind randomized trial on the efficacy of the Gufoni maneuver for treatment of lateral canal BPPV. Laryngoscope. July 2013;123(7):1782–1786.
+
Mandala
M, Santoro
GP, Libonati
GA, Casani
AP, Faralli
M, Giannoni
B, Gufoni
M, Marcelli
V, Marchetti
P, Pepponi
E, Vannucchi
P, Nuti
D. Double-blind randomized trial of short-term efficacy of the Semont maneuver for the treatment of posterior canal benign paroxysmal positional vertigo. J Neurol. 2012;259:882–885.
+
Mizukoshi
K, Watanabe
Y, Shojaku
H, Okubo
J, Wantanbe
I. Epidemiological studies on benign paroxysmal positional vertigo in Japan. Acta Otolaryngol Suppl. 1988; 447: 67–72.
+
Norre
ME. Reliability of examination data in the diagnosis of benign paroxysmal positional vertigo. American J Otology. 1995; 16:806–10.
+
Ogun
OA, Janky
KL, Cohn
ES, Büki
B, Lundberg
YW. Gender-based comorbidity in benign paroxysmal positional vertigo. PLoS ONE. 2014;9(9):e105546. doi:10.1371/journal.pone.0105546.
+
Oh
J-H, Song
S-K, Lee
JS, Choi
JC, Kang
S-Y, Kang
J-H. Lying-down nystagmus and head-bending nystagmus in horizontal semicircular canal benign paroxysmal positional vertigo: are they useful for lateralization? BMC Ophthalmol. 2014;14:136.
+
Organ
B, Liu
H, Bromwich
M. An iPhone-Assisted Particle Repositioning Maneuver for Benign Paroxysmal Positional Vertigo (BPPV): A Prospective Randomized Study. J Am Board Fam Med. 2015;28(1):118–120.
+
Parnes
LS, Agrawal
SK, Atlas
J. Diagnosis and management of benign paroxysmal positional vertigo (BPPV). CMAJ. 2003; 169:681–93.
+
Parnes
LS, McClure
JA. Posterior semicircular canal occlusion for intractable benign paroxysmal positional vertigo. Ann Otol Rhinol Laryngol. 1990; 99:330–4.
+
Pisani
V, Mazzone
S, Di Mauro
R, Giacomini
P, Di Girolamo
S. A survey of the nature of trauma of post-traumatic benign paroxysmal positional vertigo. Int J Audiol. May 2015;54(5):329–333.
+
Ribeiro
K, Freitas
R, Ferreira
L, Deshpande
N, Guerra
R. Effects of balance Vestibular Rehabilitation Therapy in elderly with Benign Paroxysmal Positional Vertigo: a randomized controlled trial. Disabil Rehab. 2016; 1–9.
+
Saberi
A, Nemati
S, Sabnan
S, Mollahoseini
F, Kazemnejad
E. A safe-repositioning maneuver for the management of benign paroxysmal positional vertigo: Gans vs. Epley maneuver; a randomized comparative clinical trial. Eur Arch Otorhinolaryngol. 2016. doi:10.1007/s00405-016-4235-7.
+
Sammartino
G, Mariniello
M, Scaravilli
MS. Benign paroxysmal positional vertigo following closed sinus floor elevation procedure: mallet osteotomes vs. screwable osteotomes. A triple blind randomized controlled trial. Clin Oral Impl Res. 2011;22:669–672.
+
Serafini
G, Palmieri
AMR, Simincelli
C. Benign paroxysmal positional vertigo of posterior semicircular canal: results in 160 cases treated with Semont's maneuver. Ann Otol Rhinol Laryngol. 1996;105:770–5.
+
Shim
DB, Song
CE, Jung
EJ, Ko
KM, Park
JW, Song
MH. Benign Paroxysmal Positional Vertigo with Simultaneous Involvement of Multiple Semicircular Canals. Korean J Audiol. 2014;18(3):126–130.
+
Smith,
M. The Peripheral System. In: Goodman
K. Pathology Implications for the Physical Therapist. 3rd edition. St Louis, MO: Saunders Elsevier. 2009:1634–1636.
+
Strupp
M, Dieterich
M, Brandt
T. The Treatment and Natural Course of Peripheral and Central Vertigo. Deutsches Aerzteblatt International. 2013;110(29/30):505–516.
+
Talaat
HS, Abuhadied
G, Talaat
AS, Abdelaal
MSS. Low bone mineral density and vitamin D deficiency in patients with benign positional paroxysmal vertigo. Eur Arch Otorhinolaryngol. 2014. doi:10.1007/s00405-014-3175-3.
+
van den Broek
E, van der Zaag-Loonen
H, Bruintjes
T. Systematic Review: Efficacy of Gufoni Maneuver for Treatment of Lateral Canal Benign Paroxysmal Positional Vertigo with Geotropic Nystagmus. Otolaryngol -- Head Neck Surg 2014;150(6):933–938.
+
Vannucchi
P, Giannoni
B, Pagnini
P. Treatment of horizontal semicircular canal benign paroxysmal positional vertigo. J Vest Res. 1997;7:1–6.
+
Von Brevern
M, Radtke
A, Lezius
F, Feldman
M, Ziese
T, Lempert
T, Neuhauser
H. Epidemiology of benign baroxysmal positional vertigo: a population based study. J Neurol Neurosurg Psychiatry. 2007;78:710–715.
+
Von Brevern
M. Benign paroxysmal positional vertigo. Semin Neurol 2013;33:204–211.
+++
Brachial Plexus Injury (Neonatal Brachial Plexus Palsy)
++
Brachial plexus injury (BPI) in both adults and children is often caused by extreme lateral traction applied to the head and neck with depression or hyperextension of the shoulder (Eser, 2009; Hudic, 2006; Wellington, 2009).
Adult BPI
Common mechanism of injury: motor vehicle accident, industrial accidents, falls, lacerations, sports related, gunshot, and iatrogenic factors, including tumors/masses and radiotherapy (Eser, 2009; Wellington 2009). BPI is often caused by fracture and/or dislocation (Eser, 2009).
Neonatal brachial plexus palsy (NBPP)
Etiology involves stretching of the brachial plexus during delivery with multiple risk factors (Benjamin, 2005; Heise 2015).
Risk Factors/Possible Causes:
Maternal: uterine abnormalities, diabetes, stature, obesity or excessive weight gain during pregnancy, age > 35 years, primiparity, pelvic anatomy, uterine anomalies, previous child with NBPP (Benjamin, 2005; Doumouchtsis, 2009; Kay, 1998)
Infant: large birth weight (over 4000 grams) most important fetal factor, gestational age (Hudic, 2006; Kay, 1998; Heise 2015)
Delivery: shoulder dystocia observed in at least half of the cases (Heise, 2015); breech delivery, prolonged labor, vacuum-assisted labor, induction of labor, propulsive forces of labor (Doumouchtsis, 2009; Hudic, 2006; Kay, 1998; Mackinnon, 2004)
Other: atraumatically in-utero, intrauterine maladaptation, failure of the shoulder to rotate (Doumouchtsis, 2009)
Although risk factors are known, it is difficult to predict NBPP, which can also occur in the absence of any risk factors. However, the presence of multiple risk factors does increase risk, particularly maternal diabetes and macrosomia (Doumouchtsis, 2009)
Injury is typically unilateral, may be to the roots or trunk of the brachial plexus, and is classically described as being a neurapraxia, axonotmesis, neurotmesis, or avulsion (Malessy, 2009; Waters, 2005).
Brachial plexus injury most commonly occurs in the anterior shoulder and is attributed to a stretch injury with tearing of the cervical nerves or avulsion of the nerve roots from the spinal cord (Hankins, 1995)
Avulsions will not recover motor function spontaneously (Jellicoe, 2008)
Mechanically, lesions can be described as a stretch, varying degrees of rupture, and avulsion (Waters, 2005). In many BPIs, the damaged and stretched nerve forms a neuroma, which can block growth and impede axonal continuity (Malessy, 2009).
++
++
++
++
Signs of NBPP are usually present immediately after birth, though a more precise diagnosis can be made after 48 hours (Gilbert, 1995).
If untreated, the internal rotators and adductors will eventually lead to posterior subluxation and progress to dislocation of the glenohumeral joint (Jellicoe, 2008).
Physical Examination
The reference standard for evaluating severity in NBPP is the clinical examination repeated over several months (Curtis, 2002).
Observation of the child's spontaneous use of the affected upper extremity during activities (Frykman, 1976).
Observation of posture, preferred pattern of prehension, muscle atrophy, sudomotor function (Ho, 2015)
Provocative tests: Tinel's sign (Ho, 2015); scarf sign
Strength: Manual muscle testing (Ho, 2015), Modified Mallet Shoulder Scale (Abzug, 2014), Pinch/grip dynamometery (Ho, 2015)
Sensory testing: Weinstein Enhanced Sensory Test (WEST)
Tactile gnosis: Two-point discrimination (2PD) (Bell, 1993); shape/texture identification (STI-test) (Rosen, 2000)
Froment's test if suspected or known ulnar nerve injury (McRae, 1990)
++
++
Diagnosis begins with observation and is characterized by asymmetrical or lack of active movement in an upper extremity although passive movement is equal on both sides.
Imaging:
Three-dimensional proton density MRI: Does not require radiation, contrast agents or sedation to evaluate the spinal nerve root leading to earlier detection of injury severity (Bauer, 2017).
CT: myelographies show nerve root avulsions, such as those occurring with OBPI more clearly (Dunham, 2003).
Electrodiagnostic testing (EDX) is a noninvasive method commonly used to assess lesion site, type of lesion, and extent of neural dysfunction (Spires, 2017)
Imaging studies should be used in adjunct with physical examination and may be used to guide preoperative planning (Waters, 2005). Imaging is also used to assess the shoulder joint to see if there is any secondary deformity (Waters, 2005).
Classification of perinatal brachial plexus injuries (Al-Qattan, 2009; Hearse, 2015):
Grade 1: Upper brachial plexus injury or Erb's Palsy (C5-6, sometimes C7): the shoulder is internally rotated and adducted, elbow extended, forearm pronated, and hand and wrist flexed (waiter's tip positioning) (Jellicoe, 2008). This is the most common form with the best prognosis (Huffman, 2005); 70-80% of cases resolve within one year (Doumouchtsis, 2009).
Grade II: Extended Erb's Palsy involving upper and middle trunks (C5-C7): seen in about one third of cases; involves motor deficits seen with Erb's palsy, plus impaired elbow and wrist extension and weak finger flexion (Hearse, 2015).
Grade III: Global Palsy (C5-C8, T1): Complete flaccid paralysis of the arm, the hand is clawed, and no movement or sensation is present in the entire arm. This is the most devastating type of OBPI and has the worst prognosis of the three types (Jellicoe, 2008; Kay, 1998). The infant may ignore the arm (Jellicoe, 2008). 30-50% experience spontaneous recovery of the shoulder and elbow but have residual hand deficits.
Grade IV: Global Palsy with Horner's syndrome (C5-C8, T1): Complete flaccid paralysis of the arm with Horner syndrome. Severe arm deficits without surgical intervention (Al-Qattan; et al, 2009).
Lower brachial plexus injury or Klumpkes paralysis (C7/8-T1): Resting position of forearm is supination, paralysis of wrist flexor and extensors and intrinsic muscles of hand (claw hand). This type is very rare (Dodds, 2000; Jellicoe, 2008). Usually persists (Doumouchtsis, 2009).
In NBPP: the right side is effected in two thirds of the cases because of the most common fetal presentation (Heise, 2015).
+++
III. Prognosis & Sequelae
++
++
Non-progressive; 12 -25% will experience permanent impairment and injury (Andersen, 2006).
Prognosis depends on the severity of the lesion (stretch, rupture, or avulsion), extent of the injury (upper, middle, lower plexus), associated fractures, associated Horner's sign, phrenic nerve involvement and if the lesion is pre- or postganglionic (Jellicoe, 2008; Waters 2005). Upper root lesions generally have a better prognosis (Jackson, 1988).
Recovery begins first in the hand, often within 8-12 weeks. Prognosis is good if any signs of recovery occur during the first month, while no recovery of function by 3 months indicates a poorer prognosis. This is considered by some to be an indication for surgery (Grossman, 2000).
++
++
Birthweight over 4 kg and younger maternal age are predictors of disability lasting >1 year (Zuarez-Easton, 2017)
Sequelae deformities may result from an NBPP that does not recover. The most common deformities are humeral dislocation, pronation/supination contractures at the forearm, radial head dislocation, scapular winging, and hypoplasia of bones in the affected extremity (Plexus, 2007).
Early intervention and referral remains the best plan for a good functional outcome (Dunham, 2003).
Outcome measures:
Mallet Classification (MC): used to assess active UE motion and characterize shoulder function in infants and children; tests 5 movements (abduction, external rotation, hand behind head, hand to back, and hand to mouth. Scores can range from 5-25; scores can be affected by muscle function, joint contracture, bony deformity, and neglect of involved arm. (Bae, 2008).
Toronto Test Score (TTS): used to evaluate neurological recovery in infants (Bae, 2008).
Hospital for Sick Children Active Movement Scale (AMS): used to quantify upper limb motor function (Bae, 2008).
Pediatric Outcomes Data Collection Instrument (PODCI): a patient/parent derived measure of functional outcomes; norm referenced (Bae, 2008; Huffman, 2005).
Brachial Plexus Outcome Measure (BPOM): Assess activity and participation (Ho, 2012)
Gilbert Shoulder Classification – grades 0-5 based on active shoulder abduction and external rotation
+++
IV. Medical/Surgical Management
++
++
Early referral to a multidisciplinary center is recommended to maximize education for parents, provide access to specialized therapists, and assess for recovery and possible operative planning (Coroneos, 2017).
Prevention, early diagnosis, and early referral to physical or occupational therapy are focus of initial management (Benjamin, 2005; Jellicoe, 2008; Mami, 1997)
++
++
Because many patients with upper plexus lesions recover spontaneously within days or weeks, surgery may not be needed (Kay, 1998). Recovery has been noted up to the age of 1 year (McAbee, 2006) and surgical intervention is only necessary in approximately 10% of cases.
Timing of surgery is debatable, but most sources say it should occur between 3-8 months if no spontaneous recovery is observed (Grossman, 2000). Microsurgery will improve, but not necessarily normalize the situation (Waters, 2005).
Surgery is usually performed for exploration, evaluation, and repair (Dunham, 2003). Surgical techniques/procedures include neurolysis, neuroma resection, nerve grafting, and nerve transfer (Grossman, 2000; Waters, 2005).
Total brachial plexus lesions, with accompanied Horner's sign, can be treated surgically at 3-4 months assuming no spontaneous recovery (Kay, 1998).
Primary surgery, often used in severe injuries or when no spontaneous healing has occurred by 3-4 months, should occur 3-6 months after birth (Mayo, 2001).
Secondary surgery is used to correct the effects of incomplete nerve healing on the surrounding joints. This type of surgery is usually performed between 2 and 10 years of age. Procedures include capsule release, joint fusion, osteotomy, and muscle or tendon transfer often occurring in combination with the goal of increasing functionality (Dunham, 2003).
Internal contracture release and muscle tendon transfer for external rotation led to improved range of motion (ROM), strength, arm function, quality of life and high parental satisfaction for children aged 3-10 using latissimus dorsi/teres major (van der Holst, 2015).
+++
V. Key Aspects of Therapeutic Management
++
Achievement of full function of the hand, wrist, elbow, and shoulder with emphasis on the restoration of abduction and external rotation guides goals (Grossman, 2000; Waters, 2005; Jellicoe, 2008).
Using the ICF framework, treatment priorities shift from emphasis on body, structure and function deficits in infancy toward activity and participation as child matures. (Duff, 2015)
Physical therapy goals should focus on aiding spontaneous recovery of function and preventing joint injury, contractures and abnormal movement patterns. Gaining full ROM and normal strength to participate in daily life activities guide goal decisions for the first two years.
Intervention should address decreased ROM, decreased strength, atypical posture, sensory deficits, and prevention of joint deformity and dislocation
ROM should be done with caution immediately post-injury (neonatal period); should be pain free; if pain, warrants further investigation of associated musculoskeletal problems; emphasis on preventing should internal rotation contractures; avoid aggressive supination as can cause radial dislocation; splinting wrist in some extension can improve function of fingers; should facilitate optimal scapula-humeral rhythm (Hearse, 2015; Semel-Concepcion, 2017)
Exercise therapy is the most frequently used conservative treatment to prevent deformities and contractures, stimulate muscle and sensory function, increase awareness of the affected upper extremity and educate the parents (Bialocerkowski, 2009).
Therapeutic management of infants: focus is on maintaining passive ROM, restoring symmetry and removing areas of potential nerve impingement (Waters, 2005; Ciervo, 2009). Caution is needed in first weeks after birth to avoid further damage.
ROM: be gentle, avoiding pain and/or overstretching; strengthening: of the entire extremity; external rotators of importance in the shoulder (Waters, 2005).
Parental education should address caring for the infant and administering a home program with gentle passive ROM (Waters, 2005).
Functional training using constraint-induced movement therapy and Botox (Santamato, 2011)
Manual therapy: myofascial release for torticollis, and contractures (Ramos, 2000).
Electrical stimulation: neuromuscular electrical stimulation (NMES) during weight bearing exercises has been shown to improve shoulder function and bone mineral density in children with OBPI (Elnaggar, 2016).
Postural education: avoid compensatory maneuvers that can lead to aberrant postures (Ramos, 2000).
Splints: used to maintain shoulder abduction and external rotation and to prevent flexion contractures of wrist and fingers (Waters, 2005). Splinting no longer recommended for infants.
Botox injection: used to reduce contraction forces that overpower weaker muscles and release contractures (Waters, 2005).
+
Abzug
JM, Kozin
SH. Evaluation and management of brachial plexus birth palsy. Orthop Clin North Am. 2014;45:225–232.
+
Al-Qattan
MM, El-Sayed
AAF, Al-Zahrani
AY,
et al. Narakas classification of obstetric brachial plexus palsy revisited. J Hand Surg [Eur]. 2009;34(7):88–791.
+
Andersen
J. Perinatal brachial plexus palsy. Paediatr Child Health. 2006;11:93.
+
Bae
DS, Waters
PM, Zurakowski
D. Correlation of pediatric outcomes data collection instrument with measures of active movement in children with brachial plexus birth palsy. J Pediatr Orthop. 2008; 28:584–592.
+
Bauer
AS, Shen
PY, Nidecker
AE, Lee
PS, James
MA. Neonatal magnetic resonance imaging without sedation correlates with injury severity in brachial plexus birth palsy. J Hand Surg Am. 2017;42(5):335–343.
+
Bell-Krotoski
J, Weinstein
S, Weinstein
C. Testing sensibility, including touch pressure, two-point discrimination, point localization, and vibration. J Hand Ther. 1993;6(2):114e123.
+
Benjamin,
K. Part 1: Injuries to the brachial plexus: mechanisms of injury and identification of risk factors. Adv Neonat Care. 2005;5(4):181–189.
+
Benjamin,
K. Part 2: Distinguishing physical characteristics and management of brachial plexus injuries. Adv Neonat Care. 2005;5(5);240–251.
+
Bialocerkowski
AE, Vladusic
S, Moore
RP. Lack of effectiveness of primary conservative management for infants with brachial plexus birth palsy. JBI Lib Syst Rev. 2009;7:354–386.
+
Chauhan
SP, Rose
CH, Gherman
RB,
et al. Brachial plexus injury: a 23-year experience from a tertiary center. Am J Obstet Gynecol. 2005;192:1795–1802.
+
Ciervo
CA, Mason
DC. Brachial plexus injuries in neonates: an osteopathic approach. J Am Osteopath Assoc. 2009;109:87–91.
+
Clarke
HM, Curtis
CG. An approach to obstetrical brachial plexus injuries. Hand Clin. 1995;11: 563–580.
+
Coroneos
CJ, Voineskos
SH, Christakis
MK, Thoma
A, Bain
JR, Brouwers
MC. Obstetrical brachial plexus injury (OBPI): Canada's national clinical practice guideline. BMJ Open. 2017;7(1).
+
Curtis
D, Stephens
HM, Clarke
D, Andrews
D. The Active Movement Scale: an evaluative tool for infants with obstetrical brachial plexus palsy. J Hand Surg Am. 2002;27(3):470–478.
+
Deaver
JE, Cohen
WR. An approach to the prediction of neonatal Erb palsy. J Perinat Med. 2009;37:150–5.
+
Dodds
SD, Wolfe
SW. Perinatal brachial plexus palsy. Curr Op Pediat. 2000;12:40–47.
+
Doumouchtsis
SK, Arulkumaran
S. Are all brachial plexus injuries caused by shoulder dystocia? Obstet Gynecol Survey. 2009;64:615–623.
+
Duff
SV, DeMatteo
C. Clinical assessment of the infant and child following perinatal brachial plexus injury. J Hand Ther. 2015;28(2):126–134.
+
Dunham
E. Obstetrical brachial plexus palsy. Orthop Nurs. 2003;22:106–116.
+
Elnaggar
RK. Shoulder function and bone mineralization in children with obstetric brachial plexus injury after neuromuscular electrical stimulation during weight-bearing exercises. Am J Phys Med Rehabil. 2016;95(4):239–247.
+
Eser
F, Aktekin
LA, Bodur
H, Atan
C. Etiological factors of traumatic peripheral nerve injuries. Neurol India. 2009;57:434–437.
+
Frykman
GK. Peripheral nerve injuries in children. Orthop Clin North Am. 1976;7(3):701e716.
+
Gilbert
A. Long-term evaluation of brachial plexus surgery in obstetrical palsy. Hand Clin. 1995;111:583–594.
+
Grossman
JAI. Early operative intervention for birth injuries to the brachial plexus. Sem Pediatr Neurol. 2000;7:36–43.
+
Hankins,
G, Clark,
S. Brachial plexus palsy involving the posterior shoulder at spontaneous vaginal delivery. Obstet Gynecol Surv. 1995;50(10):44–45.
+
Heise
CO, Martins
R, Siqueira
M. Neonatal brachial plexus palsy: a permanent challenge. Arq Neuropsiquiatr. 2015;73(9):803–808.
+
Ho
E. Evaluation of pediatric upper extremity peripheral nerve injuries. J Hand Ther. 2015;28(2):135–143.
+
Ho
ES, Curtis
CG, Clarke
HM. The brachial plexus outcome measure: development, internal consistency, and construct validity. J Hand Ther. 2012;25(4):406–16.
+
Hudic
I, Fatusic
Z, Sinanovic
O, Skokic
F. Etiological risk factors for brachial plexus palsy. j matern-fetal neonat med. 2006;19:655–661.
+
Huffman
GR, Bagley
AM, James
MA, Lerman
JA, Rab
G. Assessment of children with brachial plexus birth palsy using the Pediatric Outcomes Data Collection Instrument. J Pediatr Orthop. 2005;25:400–404.
+
Jackson
ST, Hoffer
MM, Parrish
N. Brachial-plexus palsy in the newborn. J Bone Joint Surg. 1988;70:217–220.
+
Jellicoe
P, Parsons
SJ. Brachial plexus birth palsy. Current Orthop. 2008;22:289–294.
+
Kay
SPJ. Obstetrical brachial palsy. Br J Plast Surg. 1998;51:43–50.
+
Malessy
MJA, Pondaag
W, van Dijk
JG. Electromyography, nerve action potential, and compound motor action potentials in obstetric brachial plexus lesions: validation in the absence of a "gold standard." Neurosurg. 2009;65:A153–A159.
+
Mami
C, Manganaro
R, Paolata
A, Lo Monaco
I, Gemelli
M. Brachial plexus paralysis: its incidence, predisposing factors and outcome. Minerva Ginecologica. 1997;49:203–6.
+
McAbee
G, Ciervo
C. Medical and legal issues related to brachial plexus injuries in neonates. J Am Osteopath Assoc. 2006;106:209–212.
+
McRae
R. Segmental and Peripheral Nerves of the Upper Limb. Clinical Orthopaedic Examination. 3rd ed. London, UK: Churchill Livingstone; 1990:9e25.
+
Ramos
LE, Zell
JP. Rehabilitation program for children with brachial plexus and peripheral nerve injury. Sem Pediatr Neurol. 2000;7(1):52–57.
+
Rosen
B, Lundborg
G. A model instrument for the documentation of outcome after nerve repair. J Hand Surg Am. 2000;25(3):535e543.
+
Santamato
A, Panza
F, Ranieri
M, Fiore
P. Effect of botulinum toxin type A and modified constraint-induced movement therapy on motor function of upper limb in children with obstetrical brachial plexus palsy. Childs Nerv Syst [serial online]. 2011;27(12):2187–2192.
+
Semel-Concepcion, Moberg-Wolff
EA. Neonatal brachial plexus palsies treatment & management: rehabilitation program, medical issues/complications, surgical intervention. Medscape.
http://emedicine.medscape.com/article/317057-treatment. Updated June 17, 2016. Accessed April 15, 2017.
+
Spires
MC, Brown
SM, Chang
KW, Leonard
JA, Yang
LJ. Interrater reliability of electrodiagnosis in neonatal brachial plexopathy. Muscle Nerve. 2017;55 (1):69–73.
+
van der Holst
M, Vlieland
TP, Meesters
JJ, Bekkering
WP, Nagels
J, Nelissen
RG. Evaluation of shoulder function after secondary surgery in children with neonatal brachial plexus palsy. J Pediatr Rehabil Med. 2015;8(3):187–96.
+
Waters
PM. Update on management of pediatric brachial plexus palsy. J Pediatr Orthop. 2005;14:233–244.
+
Wellington
B. Quality of life issues for patients following traumatic brachial plexus injury - Part 1: literature review. J Orthop Nurs. 2009;13:194–200.
+
Zuarez-Easton
S, Zafran
N, Garmi
G, Hasanein
J, Edelstein
S, Salim
R. Risk factors for persistent disability in children with obstetric brachial plexus palsy. J Perinatol. 2017; 37(2):168–171.
+++
Cerebellar Disease and Tumor
++
A. Cerebellar Infarction:
++
Can result from occlusion in the major branches to the middle or superior cerebellar artery or to the posterior inferior cerebellar artery (PICA) as well as the basilar artery. Infarction in anterior inferior cerebellar artery (AICA) seldom happens (Amarenco, 1990)
Infarction of PICA as well as the superior cerebellar artery may occur with massive paramedian brainstem infarction and cerebellar tonsilar herniation.
There is a frequent association with vertigo and impaired hearing with AICA infarct because of common blood supply (Ogawa, 2016).
Arteriosclerosis, and embolism, can cause cerebellar infarction (Amarenco, 1990).
Risk factors for cerebellar infarction include the following:
Poor dietary habits (hyperlipidemia), hypertension, diabetes mellitus, obesity, smoking, and those who suffered previously from a stroke.
Hypertriglyceridemia and rheumatic valve disease have been identified as risk factor (Amaya, 1997).
Trauma like whiplash, MVA or closed head injuries could also contribute to infarction (Schmahmann, 2004).
Infarctions in younger adults have been associated with drug abuse, vascular dissection, infections, and hypercoagulable states (Datar, 2014).
Cerebellar infarctions are present in 2 to 3% of people who present to the emergency department with strokes (Datar, 2014).
Many patients do not report acute onset of symptoms, and they may not seek medical attention for several days (Mitoma, 2016).
++
++
Hemangioblastoma- a tumor that is typically located in the posterior cranial fossa and presents primarily as a solitary lesion with an uncertain origin in the cerebellum.
Hemangioblastomas often occur in the spinal cord and brains stem in addition to the cerebellar hemisphere (Ji, 2016).
The endothelial cells and pericytes are not neoplastic and the stromal cells are believed to be the principle tumor cell (Catteeuw, 1994).
Hemangioblastomas are rare and make up only about 2% of all intracranial neoplasms, however they are the most common primary adult tumor of the posterior fossa.
The etiology is not clear
Presence in various clinical syndromes may suggest an underlying genetic abnormality (Catteeuw, 1994).
The tumor is usually sporadic but approximately ¼ of the cases are associated with von Hippel-Lindau (VHL) disease, which is an autosomal dominant hereditary syndrome (often called Lindau's tumor) (Catteeuw, 1994).
Intermittent hemangioblastomas are typically associated with cysts and are large at the time of discovery. (Liao, 2014)
Cerebellar hemangioblastomas can cause disease symptoms similar to anorexia nervosa and appetite has been shown to return immediately following resection of the tumor (Oya, 2014)
Cerebellar Astrocytoma- a disease in which benign or malignant cells form in the tissues of the brain from brain cells called astrocytes.
Pilocytic astrocytomas are benign tumors located in the cerebellum more likely to occur in the first 20 years of life (Ye, 2014). The term "pilocytic" derives from the Greek word for hair and refers to the elongated and bipolar microscopic appearance of tumor cells. (Ye, 2014)
Although cancer is rare in children, brain tumors are the most common type of childhood cancer other than leukemia and about 15-25% of all childhood brain tumors are astrocytomas (Garcia, 1989).
Astrocytomas of the cerebellum are often cystic and well circumscribed (Schmahmann,2004).
Cerebellar pilocytic astrocytomas (Brandao, 2017)
Forty percent occur in the cerebellum. Sixty percent of all posterior fossa tumors in children are either cerebellar pilocytic astrocytomas or medulloblastomas.
Classified by the World Health Organization as a grade I tumor.
++
C. Hereditary Cerebellar degenerative disease:
++
Spincocerebellar Ataxia (SCA) or also called Autosomal dominant spinocerebellar ataxia (SCAs) (Schmahmann, 2004)
There are currently 44 different types of SCA, each with a different genetic cause but with very similar symptoms. (Smeets, 2016)
SCAs type 1,2,3,7, and 17 are due to lengthen of polyglutamine. (Schmahmann, 2004)
SCA1 is an incurable, dominantly-inherited neurodegenerative disease of the cerebellum caused by a polyglutamine-repeat expansion in the protein ATXN1 (Cvetanovic, 2015).
Disruption of Potassium or Calcium channel function cause the channelopathies, SCA 6, and episodic ataxia type 1 and 2 (Schmahmann, 2004).
SCAs 8, 10 and 12 are also called gene expression disorders. They are cause by repeat expansions outside of coding regions. (Schmahmann, 2004)
SCA type 12 is usually seen in German-American and Indian descents and is the most frequent ataxias in India (Swarup, 2012).
Machado-Joseph disease (SCA type 3)
SCA type 3 may be the most common autosomal-dominant ataxia in many regions of the world, characterized by phenotypic heterogeneity (Fahl, 2014).
SCA3 is characterized by cervical cord atrophy and flattening (Fahl, 2013).
SCA3 is characterized by cerebral cortical damage with progression of the disease, leading to motor and cognitive dysfunction (De Rezende, 2015).
Friedreich's Ataxia
Friedreich's Ataxia is an autosomal recessive neurodegenerative disorder (Vogel, 2014) and is the most common form of childhood onset ataxia with average onset at age 10 (Vogel, 2014).
The major clinical signs of Friedreich's ataxia are progressive ataxia, scoliosis, dysarthria (the most common type of speech deficiency), cardiomyopathy, diabetes mellitus and foot deformity (Vogel, 2014).
Joubert Syndrome
Incidence of between 1/80,000 and 1/100,000 births (Brancati, 2010).
Joubert Syndrome is an autosomal recessive or X-linked (rare) neurological disorder (Romani, 2013).
Joubert Syndrome: characterized by underdevelopment of the cerebellar vermis and additional brain stem differences results in hypotonia; and developmental delays/intellectual disabilities of varying degrees.
+++
II. Diagnostic Procedure:
++
A. General Signs and Symptoms:
++
Headache, nausea and vomiting, incoordination/ataxia ipsilateral to lesion side, dysmetria/pass-pointing, intention tremor, dysdiadochokinesia, movement decomposition, overcompensation, dysarthria, confusion, nystagmus, diplopia, hypotonia, dysphagia, asthenia, gait disorder, loss of balance, neuropathy, asthenia, kyphoscoliosis, anorexia and gross muscle wasting (Oya, 2014).
Sleep disturbances (DelRosso, 2014).
++
++
Imaging:
MRI is the method of choice for diagnosing cerebellar infarction (De Cocker, 2017).
On MRI, anterior inferior cerebellar artery infarction can often be mistaken for a lateral posterior inferior cerebellar artery infarction.
On MRI, subacute cerebellar infarcts may be missed during imaging methods due to a phenomenon called "fogging" where the infarct is almost invisible on MRI initially.
CT and MRI scan can be used to determine cerebral hemorrhage, but MRI has poor sensitivity for intracranial hemorrhage (Kummer, 2002).
Diffusion weighted MRI is more sensitive, which shows within the first two hours
Non-contrast head CT scan can be used to detect hyper-density of basilar artery, which can indicate a thrombotic occlusion (Datar, 2014).
Angiography and MR angiography can also be used.
Transcranial Doppler Sonography has been shown as a noninvasive method of monitoring elevated ICP in patients with large infarctions due to its ability to show cerebral cranial herniation and to decide if medical therapy or surgical therapy is to be used (Wijdicks, 2014).
Labs and Other Tests:
A complete blood count can be helpful. Levels of blood glucose, sedimentation rate, along with a test to exclude syphilis and elevated cholesterol levels should be included
Cardiac arrhythmia can be ruled out with electrocardiography.
Signs and Symptoms:
Non-equilibrium coordination tests can be used to assess cerebellar coordination function.
Having the patient fixate on the PT finger can test nystagmus. PT will be able to observe back and forth movement of the patient's eye during gaze upon a lateral placed object (PT finger) to the midline and back (Donan, 1987)
Cranial Nerve testing should also be done due to possible cranial nerve involvement, especially test for CNVII as well as CNVIII, CNIV, CNV, CNVI, CNIX-CNXII
Patients often complain of dizziness, vertigo, mild instability, or vomiting (Mitoma, 2016).
++
++
Initial observation of a hemangioblastoma/astrocytoma is made by a MRI/CT scan.
Laboratory test should also be done in order to determine red blood cell count.
Brain tumor type is confirmed by biopsy and microscopic examination of tissue/cell type (Hayostek,1993).
Some tumors are initially discovered when patient presents with anorexia nervosa symptoms to gastroenterologists due to medical referral (Oya, 2014).
Hemangioblastomas in the cerebellum can be misdiagnosed as an aneurysm or ArterioVenous Malformation (AVM) of the PICA (Ji, 2016).
D. Hereditary Cerebellar Degenerative Disease:
Spinocerebellar Ataxias (SCA)
Diagnostic Testing
Evidence of degeneration can be seen on MRI/CT scan, with location depending on the type of ataxia.
Current molecular test can be used to identify the types of SCAs (Musova, 2012).
Genetic testing by using triple repeat primed polymerase chain reaction to diagnose Friedreich's ataxia (Xunclà, 2010)
Examination of expansions of gene is part of the differential diagnosis to identify types of SCA. (Musova, 2012)
Signs & Symptoms
Nystagmus usually seen in SCA 3, 6, 10, 15, and 16 (Rossi, 2014)
Diplopia more commonly seen in SCA 1, 2, 3, 6, and 31 (Rossi, 2014)
Visual impairment and retinal degeneration mostly seen in SCA7 (Rossi, 2014).
Intention or postural tremor usually found in SCA 2 and 12 (Rossi, 2014).
Joubert Syndrome:
Presence of the "molar tooth sign" on axial MRI (T1-weighted) through the malformed pontomesencephalic junction (isthmus); two key features: enlarged superior cerebellar peduncles that do not decussate in the isthmus of the brain stem and cerebellar vermis dysplasia (Maria, 1997).
Hypotonia in 100% of infants (Parisi, 2007)
The presence of ataxia with broad-based gait, hypotonia, oculomotor disturbances, and language difficulties help to support the diagnosis of Joubert Syndrome (Bolduc, 2009; Brancati, 2010).
Developmental delay and mental retardation/intellectual disability in 100% of individuals but variable severity (Parisi, 2007; Brancati, 2010)
One or both of the following (not absolutely required but supportive of the diagnosis): (Parisi, 2007)
Irregular breathing pattern in infancy (episodic tachypnea and/or apnea)
Abnormal eye movements (including nystagmus, jerky eye movements, and oculomotor apraxia or difficulty with smooth visual pursuits)
++
Progressive disease and prognosis varies among underlying causes
Recovery from cerebellar lesions is often slow and incomplete (Deluca, 2011).
Trends of Progression
a) Early Stage
Poor balance (Schmahmann, 2004)
Inability to perform tandem gait (Schmahmann, 2004)
Nystagmus(Ataxia UK)
Dysarthria(Ataxia UK)
b) Later Stages
Wide base gait (Schmahmann, 2004)
Problem when turning around (Schmahmann, 2004)
Stepping, staggering from side to side (Schmahmann, 2004)
Dysphagia (Ataxia UK)
Parkinsonian features (Ataxia UK)
Ophthalmoplegia (Ataxia UK)
Expected Sequelae
Structural
Cerebellar atrophy (Stowe, 2012)
Disrupted motor/sensory pathways connecting cerebellum with other parts of the brain (Bastian, 1997)
Function (Walker, 1990)
Poor visual scanning, impaired visual/verbal memory, paraphasic errors
Motor problems, i.e. frequent falls due to incoordination
Difficulties with ADL such as, buttoning, donning, cooking etc.
When the disease is very severe, the patient will not be able to stand or walk even with assistance. (Schmahmann, 2004)
++
A. Cerebellar Infarction:
++
Prognosis in general always depends on the severity of the infarct and the time that lapses before intervention is initiated.
No other brain infarction has a higher fatality rate than cerebellar infarction (Donnan, 1987).
Poor Prognostic Factors
Prognosis is worse in older people and those with multiple comorbidities (Datar, 2014).
Survival is less for patients who lose consciousness due to infarction and exhibit coma.
Progressive brainstem dysfunction can be seen in some patients who had a cerebellar infarction but did not receive surgical alleviation of the cerebellar compression within 24-96 hours (Donnan, 1987).
Delayed treatment can lead to further strokes and death (Xunclà, 2010).
Patients with infarction of SCA are more impaired (postural impairment and gait ataxia) than those with infarction of PICA (Bultmann, 2014).
Surgical intervention is used in patients who are rapidly deteriorating due to large amounts of edema and has favorable outcomes based on decreased co-morbidities and time elapsed from onset of symptoms (Datar, 2014).
++
++
Hemangioblastoma
Complete resection is usually curative (Jagannathan, 2008).
Recurrence can occur locally even with total resection and is more common with von Hippel-Lindau (VHL) complex
Usually a long history of minor neurologic symptoms (Catteeuw, 1994).
Patient's with gross emaciation and anorexia nervosa symptoms generally have a worse outcome due to delayed diagnosis of the tumor because of the belief the patient had a psychological disorder (Oya, 2014).
Cerebellar Astrocytoma (childhood)
Most important factor affecting prognosis is grade of astrocytoma and whether cancer cells remain after surgery (Hayostek,1993).
Complete removal during surgery results in 90% chance of cure (Gracia, 1989).
Tumors that are not fully removed are likely to recur.
Low grade astrocytomas treated optimally have a 5 to 10 year survival rate of 100% for completely excised lesions (Gracia, 1989).
Long term quality of life prognosis after tumor excision is positive when combined with neurocognitive interventions that are carefully coordinated with parents and teachers to correctly sequence interventions (Aarsen, 2004)
Pilocytic astrocytomas have a more favorable prognosis in a younger population. (Ye, 2014)
Cerebellar high-grade astrocytomas result in worse survival rates compared to cerebral high-grade astrocytomas (Karremann, 2013).
There is little to no correlation between location of cerebellar astrocytoma and the cognitive or adaptive outcomes in a pediatric population (Beebe, 2005).
++
C. Hereditary Cerebellar Degenerative disease:
++
Friedreich's ataxia
15-20 years after the first symptoms, patients usually dependent on wheelchair. (NINDH)
Cardiac dysfunction is the most common cause of death in patients with Friedreich's Ataxia (59%) (Delatycki, 2012)
Onset typically occur before 25 years old. (Delatycki, 2012)
Spinocerebellar Ataxias Type 1, 2, 6
Mean age onset 19 to 71 years old.
Survives 10-15 years after the appearance of first symptoms (Globas, 2008)
Machado-Joseph Disease (Spinocerebellar Ataxia Type 3)
Aspiration pneumonia often cause the early death in patients with MJD (Machado-Joseph Disease Fact Sheet.)
Life expectancy ranges from the mid-30's for those with most severe form to nearly normal life expectancy for those with mild, late-onset forms.
Patients with Machado-Joseph Disease have shown extracerebellar degeneration (including the cerebral cortex) throughout the course of the disease, causing a decline in motor and cognitive function. (de Rezende, 2015)
Spinocerebellar Ataxias Type 8
Patients with SCA8 will typically have a normal lifespan (Stone, 2001)
Most of the time it is associated with sporadic cases (Musova, 2012)
Joubert Syndrome (and related disorders)
While global developmental delay is frequent, health and growth are usually not severely affected.
Commonly a degree of mild or moderate retardation.
Few cases of affected individuals with normal intelligence or learning abilities.
+++
IV. Medical and Surgical Management
++
++
Physostigmine: Active cholinerase inhibitor physostigmine to prevent further cerebellum atrophy (Ogawa, 2004).
For ataxia
Choline and choline derivatives: Lecithin/ pure phosphatidycholine may improve ataxia and muscle strength in patients with Friedreich's Ataxia (Ogawa, 2004).
Serotonergic system: hydroxytryptophan (Serotonin precursor) may improve cerebellar ataxia (Ogawa, 2004).
Thyrotrophin releasing hormone is used to improve ataxic syndrome in patients with SCAs (Ogawa, 2004).
Acetazolamide is used to improve sign and symptoms of episodic ataxia type 2. v. Cycloserine: D-serine ethylester or D-cycloserine is effective to treat truncal ataxia, and dysarthria.(Ogawa, 2004)
Varenicline(for type 3 SCA) to improve axial symptoms and rapid alternating movements (Zesiewicz, 2012)
Riluzole has been used to improve cerebellar ataxia in research and is moving towards use in clinical practice (Romano, 2015).
Sulfamethoxazole-trimethoprim: Tetrahydrobiopterin can be used to improve gait and spasticity (Ogawa, 2004).
Aminopyridines help treat downbeating nystagmus as a symptom (Ilg, 2014).
Co-enzyme Q10 may decrease seizures, developmental delay, mental retardation, and pyramidal signs (Lamperti, 2003)
The efficiency of corticosteroids (prednisone or dexamethasone) in the reduction of brain swelling is questioned (Tierney, 2003).
Idebenone (for Friedreich's Ataxia): Used as therapy for more than a decade; may provide a trend toward improved total, emotional, social, and school components of quality of life (after 1 year trial). (Brandsema, 2010)
Mannitol and hypertonic saline are used to manage cranial swelling after infarction (Datar, 2014)
To date, no medications have proven to be effective in halting disease progression (Ilg, 2014).
There is growing evidence supporting the use of coenzyme Q10 and lithium carbonate to treat Machado Joseph Disease (SCA3). However, more research is needed (Lopes-Ramos, 2016).
++
B. Possible surgical techniques
++
Tumors
High dose radiation may reduce the size of the tumor (Amaya, 1997)
Chemotherapy is sometimes used to stop the growth of cancer cells
Surgical excision of the tumor nodule (cyst wall does not have to be removed)
Infraction
Surgical decompression to decrease the intracranial pressure
Hemicraniectomy for severe cerebellar infarction (Wijdicks, 2014)
Bilateral cerebellar infarctions are best managed using immediate decompressive craniectomy and insertion of external ventricular drainage to prevent loss of life or decreased state of consciousness. (Tsitsopoulos, 2011)
Thalamotomy to treat tremor (Marmolino, 2010)
High frequency electrical stimulation (Deep Brain Stimulation) of the ventral intermediate nucleus (Marmolino, 2010)
Stem cell transplantation (Marmolino, 2010)
Spine surgery to correct neurology scoliosis to reduce ataxias symptoms (lumbar transpedicular screws, thoracic Universal Clamps, pedicle-transverse hooks at the upper end of the curve) (La Rosa, 2010)
+++
V. Therapeutic Management:
++
A. Guide Practice Patterns (APTA, 2017)
++
Pattern 5A: Primary Prevention/Risk Reduction for Loss of Balance and Falling
Pattern 5E: Impaired Motor Function and Sensory Integrity Associated With Progressive Disorders of the Central Nervous System
++
++
Therapy for cerebellar tumors will consider hypotonia, ataxia, tremors, asthenia, rebound phenomenon, nystagmus, prevention and intervention for movement disorders, and dysarthria (Konecne, 1998)
Treatment for Incoordination/Ataxia
Balance, coordination and agility training
Vestibular treatment
Encourage patients to be physically active
Strengthening exercises
Neuromuscular training
Exergame training (Synofzik, 2014)
Treatment for Difficulties in ADL
Body mechanics and postural stabilization
Neuromotor retraining for specific functional motor patterns (Synofzik, 2010)
Gait and locomotion training
PNF patterns for strengthening and stabilization
Treatment for Tremor
Gait and locomotion training
Treatment for Dysphagia
Prevent Muscle Contracture
Passive range of motion
Stretching
Treatment for Hypotonia
Use NMES to activate hypotonic muscles
Postural control training
Stretching and tapping if necessary
Considerations for Physical Therapists (Kelly, 2015)
Patients with cerebellar lesions have difficulty responding to and learning from unexpected sensory input, so consider the environment in which therapy takes place.
Cerebellar impairment results in impaired motor learning.
Patients with cerebellar lesions have difficulty consolidating movements as automatic, especially problematic during gait training
++
C. Contraindications/precautions
++
Patients should be encouraged to reduce anxiety and stress because stress will worsen the tremor.
Patients with cerebellar disease should avoid alcohol assumption because it will exacerbate the condition. (Stone, 2001).
Radiation should be delayed for children younger that 3 years of age due to the effects of radiation on the developing brain (Catteeuw, 1994).
Diabetic patients with Friedreich's Ataxia need precautions when doing exercises because they have lower glucose tolerance. (Delatycki, 2012).
++
D. Healthcare promotion/education
++
Education of lifestyle changes
a) A modified diet with lipid intake reduction
b) Control of hypertension
c) Cessation of smoking
d) Increase of physical activities with the goal on weight reduction.
e) Decreased intake of alcohol
f) Control of diabetes mellitus and other co-morbidities
+++
VI. Consumer and Professional Resources
++
American Brain Tumor Association http://www.abta.org
National Ataxia Foundation (NAF) http://www.ataxia.org
Friedreich's Ataxia Research Alliance (FARA) http://www.CureFA.org
National Organization for Rare Disorders (NORD) http://www.rarediseases.org
National Society of Genetic Counselors (NSGC) http://www.nsgc.org
Joubert Syndrome & Related Disorders Foundation http://www.jsrdf.org/
Friedreich's Ataxia Research Alliance (FARA) Patient Registry http://www.curefa.org/patient-registry
National Institute of Neurological Disorders and Stroke (NINDS) http://www.ninds.nih.gov/index.htm
+
Aarsen
F, Van Dongen
H, Paguier
P, Van Mourik
M, Catsman-Berrevoets
C. Long-term sequelae in children after cerebellar astrocytoma surgery. Neurology. 2004 (62) 8: 1311–1316.
+
Amarenco
P, Hauw
JJ. Cerebellar infarction in the territory of the anterior and inferior cerebellar artery. A clinicopathological study of 20 cases. Brain. 1990 Feb;113(1):139–55.
+
Amaya
LE, Barinagarrementeria
F. Causes and Mechanisms of Cerebellar Infarction in Young Patients. Stroke. 1997;28:2400–2404.
+
Bastian
AJ. Mechanisms of ataxia. Phys Ther. 1997;77: 672–675
+
Beebe
DW, Ris
MD, Armstrong
FD,
et al. Cognitive and adaptive outcome in low-grade pediatric cerebellar astrocytomas: evidence of diminished cognitive and adaptive functioning in national collaborative research studies (CCG 9891/POG 9130). J Clin Oncol. 2005;23(22):5198–5204.
+
Bernard
L. Maria, Karin
B.N. Hoang, Ronald
J. Tusa, Anthony
A. Mancuso, Latif
M. Hamed, Ronald
G. Quisling, Martin
T. Hove, Eileen
B. Fennell, Margaret
Booth-Jones, Deborah
M. Ringdahl, Anthony
T. Yachnis, Gwen
Creel, Beth
Frerking. "Joubert Syndrome" Revisited: Key Ocular Motor Signs With Magnetic Resonance Imaging Correlation. J Child Neurol. 1997;12(7):423–430.
+
Bolduc
ME, Limperopoulos
C. Neurodevelopmental outcomes in children with cerebellar malformations: a systematic review. Dev Med Child Neurol. 2009;51(4):256–67.
+
Brancati
F, Dallapiccola
B, Valente
EM, Brancati
F, Dallapiccola
B, Valente
EM. Joubert Syndrome and related disorders. Orphanet J Rare Dis. 2010;5:20–20.
+
Brandão
LA, Young
Poussaint T. Posterior Fossa Tumors. Neuroimaging Clin N Am. 2017;27(1):1–37.
+
Brandsema
JF, Stephens
D, Hartley
J, Yoon
G. Intermediate-Dose Idebenone and Quality of Life in Friedreich Ataxia. Pediatric Neurology. 2010;42(5):338–342.
+
Bultmann
U, Pierscianek
D, Gizewski
ER,
et al. Functional recovery and rehabilitation of postural impairment and gait ataxia in patients with acute cerebellar stroke. Gait Posture. 2014;39(1):563–569.
+
Catteeuw
R, Vanslembrouk
R, Seynaeve
et al.. Hemangioblastoma; von Hippel-Lindau disease. Radiologic Documents, 1994.
+
Cvetanovic
M, Ingram
M, Orr
H, Puneet,
O. Early activation of microglia and astrocytes in mouse models of spinocerebellar ataxia type 1. Neuroscience. 2015 Mar 19;289:289–99.
+
Datar,
S, Rabinstein,
A. Cerebellar Infarction. Neuro Clin. 2014 (32) 4: 979–991.
+
De Cocker
LJL, Le Cock
K-O, Hendrikse
J. MRI of Cerebellar Infarction. Eur Neurol. 2017;77(3-4):137–146.
+
De Rezende
TJ, D'abreu
A, Guimarães
RP, Lopez
TM, Lopes-Cendes
I, Cendes
F, Castellano
G, Franca
Jr MC. Cerebral cortex involvement in Machado-Joseph disease. Eur J Neurol. 2015;22(2):277–83, e23–4.
+
Delatycki
M, Corben
L. Clinical Features of Friedreich Ataxia. J Child Neurol. 2012 ;27(9):1133–1137.
+
Delrosso
LM, Hoque
R. The cerebellum and sleep. Neurol Clin. 2014;32(4):893–900.
+
Deluca
C, Moretto
G, Di Matteo
A,
et al. Ataxia in posterior circulation stroke: Clinical-MRI correlations. J Neurol Sci. 2011;300(1):39–46.
+
Donnan
GA. Cerebellar infarction: Natural history, Prognosis, and Pathology. Stroke. 1987;18(5): 849–55.
+
Fahl
C, Branco
L, Bergo
D'Abreu, A, Lopes-Cendes
I, Franca,
MC. Spinal cord damage in machado-joseph disease. Cerebellum. Nov 2014. DOI 10.1007/s12311-014-0619-7
+
Globas
C, du Montcel
ST, Baliko
L, Boesch
S, Depondt
C, DiDonato
S, Durr
A, Filla
A, Klockgether
T, Mariotti
C, Melegh
B, Rakowicz
M, Ribai
P, Rola
R, Schmitz-Hubsch
T, Szymanski
S, Timmann
D, Van de Warrenburg
BP, Bauer
P, Schols
L. Early symptoms in spinocerebellar ataxia type 1, 2, 3, and 6. Mov Disord. 2008;23:2232–2238.
+
Garcia
DM, Latifi
HR, Simpson
JR
et al.. Astrocytomas of the cerebellum in children. J Neurosurg. 1989; (71): 661–664.
+
Hayostek
CJ, Shaw
EG, Scheithaur
B,
et al. Astrocytomas of the cerebellum, A comprehensive clinicopathology study of pilocytic and diffuse astrocytomas. Cancer.1993; (72): 856–869.
+
Ilg
W, Bastian
AJ, Boesch
S, Burciu
RG, Celnik
P, Feil
K, Kalla
R, Miyai
I, Nachbauer
W, Schols
L, Strupp
M, Synofzik
M, Teufel
J, Timmann,
D. Consensus paper: management of degenerative cerebellar disorders. Cerebellum. 2014;13(2):248–68. doi:10.1007/s12311-013-0531-6.
+
Jagannathan
J, Lonser
RR, Smith
R, DeVroom
HL, Oldfield
EH. J Neurosurg. 2008;108:210–222.
+
Ji
Y-C, Li
Y, Hu
J-X, Zhang
H-B, Yan
P-X, Zuo
H-C. Cerebellar hemangioblastoma mimicking an aneurysm: A case report and literature review. Oncol Lett. 2016;12(4):2622.
+
Karremann
M, Rausche
U, Roth
D, Kuhn
A, Pietsch
T, Gielen
GH, Warmuth-Metz
M, Kortmann
RD, Straeter
R, Gnekow
A, Wolff
JEA, Kramm
CM. Cerebellar location may predict an unfavourable prognosis in paediatric high-grade glioma. Br J Cancer. 2013;109(4):844–51.
+
Kelly
G, Shanley
J. Rehabilitation of ataxic gait following cerebellar lesions: Applying theory to practice. Physiother Theory Pract. 2016;32(6):430–437.
+
Kummer
RV. MRI:The gold standard for Detecting Brain Hemorrhage? Stroke. 2002; 33: 1748–1749
+
La
Rosa, Giglio
G, Oggiano
L. Surgical treatment of neurological scoliosis using hybrid construct (lumbar transpedicular screws plus thoracic sublaminar acrylic loops). Eur Spine J. 2011;20(1):909–914.
+
Lamperti
C, Naini
A, Hirano
M, De Vivo
DC, Bertini
E, Servidei
S, Valeriani
M, Lynch
D, Banwell
B, Berg
M, Dubrovsky
T, Chiriboga
C, Angelini
C, Pegoraro
E. Cerebellar ataxia and coenzyme Q10 deficiency. Neurology 2003;60:1206–1208.
+
Liao
C, Huang
Y. Clinical features and surgical outcomes of sporadic cerebellar hemangioblastomas. Clin Neurol Neurosurg. 2014 (125):160–165.
+
Loh
J-K, Lieu
A-S, Chai
C-Y,
et al. Arrested growth and spontaneous tumor regression of partially resected low-grade cerebellar astrocytomas in children. Childs Nerv Syst. 2013;29(11):2051.
+
Lopes-Ramos
CM, Pereira
TC, Dogini
DB, Gilioli
R, Lopes-Cendes
I. Lithium carbonate and coenzyme Q10 reduce cell death in a cell model of Machado-Joseph disease. Braz J Med Biol Res. 2016;49(12).
+
Marmolino
D, Manto
M. (March, 2010). Past, Present and Future Therapeutics for Cerebellar Ataxias. Curr Neuropharmacol. 2010;8(1):41–61.
+
Mitoma
H, Manto
M. The physiological basis of therapies for cerebellar ataxias. Ther Adv Neurol Disord. 2016;9(5):396. doi:10.1177/1756285616648940.
+
Musova
Z, Klempir
J, Vyhnalek
M, Apltova
L. Spinocerebellar Ataxias Type 8, 12, and 17and Dentatorubro-Pallidoluysian Atrophy in Czech Ataxic Patients. Cerebellum. 2012.
+
Ogawa
M. Pharmacological treatments of cerebellar ataxia. Cerebellum. 2004;3(2):107–111.
+
Ogawa
K, Suzuki
Y, Takahashi
K, Akimoto
T, Kamei
S, Soma
M. Clinical Study of Seven Patients with Infarction in Territories of the Anterior Inferior Cerebellar Artery. J Stroke Cerebrovasc Dis. 2017 Mar 31;26(3):574–81.
+
Oya
S, Nejo
T, Indo
M, Matsul
T. Pearls & oy-sters: anorexia and emaciation in patients with cerebellar hemangioblastoma. Neurology. 2014: 83 (14) 1298–1300.
+
Parisi
MA, Doherty
D, Chance
PF, \ Glass IA. (2007). Joubert syndrome (and related disorders) (OMIM 213300). European Journal of Human Genetics. 2007; 15, 511–521.
+
Romani
M, Micalizzi
A, Valente
EM. Joubert syndrome: congenital cerebellar ataxia with the molar tooth. Lancet Neurol. 2013;12(9):894–905.
+
Romano
S, Coarelli
G, Marcotulli
C,
et al. Riluzole in patients with hereditary cerebellar ataxia: a randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2015;14(10):985–91.
+
Rossi
M, Perez-lloret
S, Doldan
L,
et al. Autosomal dominant cerebellar ataxias: a systematic review of clinical features. Eur J Neurol. 2014;21(4):607–15.
+
Schmahmann
JD. Disorders of the cerebellum: ataxia, dysmetria of thought, and the cerebellar cognitive affective syndrome. J Neuropsychiatr Clin Neurosci. 2004;16:367–378.
+
Smeets
CJ, Verbeek
DS. Climbing fibers in spinocerebellar ataxia: A mechanism for the loss of motor control. Neurobiol Dis. 2016;88:96–106.
+
Stone
J, Smith
L, Watt
K, Barron
L, Zeman
A. Incoordinated thought and emotion in spinocerebellar ataxia type 8. J Neurol.2001;248:229–232.
+
Stowe
RC, Karkare
S, Puri
V. Case Report of Subacute Cerebellar Ataxia of Adolescence With Long-Term Sequelae. J Child Neurol. 2001;.00(0):1–8.
+
Swarup
V, Srivastava
AK, Rajeswari
MR. Identification and quantification of differentially expressed proteins in plasma of spinocerebellar ataxia type 12. Neuroscience Research. 2012;(73):161–167.
+
Synofzik
M, Brötz
D, Burkard
S, Giese
MA, Schöls
L. Intensive coordinative training improves motor performance in degenerative cerebellar disease. Neurology.2010;73 (22): 1823–1830.
+
Synofzik
M, Ilg
W. Motor training in degenerative spinocerebellar disease: ataxia-specific improvements by intensive physiotherapy and exergames. Biomed Res Int. 2014;2014:583507. doi:10.1155/2014/583507
+
Tsitsopoulos
PP, Tobieson
L, Enblad
P, Marklund
N. Clinical outcome following surgical treatment for bilateral cerebellar infarction. Acta Neurol Scand. 2011; 123(5): 345–351.
+
Vogel
A, Folker
J, Poole
M. Treatment for speech disorder in friedreich ataxia and other hereditary ataxia syndromes. Cochrane Db Syst Rev. 2014:10. 0.1002/14651858.CD008953.
+
Walker
HK. The Cerebellum. In: Walker
HK, Hall
WD, Hurst
JW. Clinical Methods: The History, Physical, and Laboratory Examinations. 3rd edition. Boston: Butterworths; 1990. Chapter 69. Available at:
http://www.ncbi.nlm.nih.gov/books/NBK392/ +
Wijdicks,
E, Sheth,
K, Carter,
B, Greer,
D, Kasner,
S, Kimberly,
W, Schwab,
S, Smith,
E, Tamargo,
R, Wintermark,
M, American Heart Association Council. American recommendations for the management of cerebral and cerebellar infarction with swelling: a statement for healthcare professionals from the American Heart Association/American Stroke Association. Stroke. 2014 45(4):1222–38.
+
Xunclà
M, Rodríguez-Revenga
L, Madrigal
I, Jiménez
D, Milà
M, Badenas
C. Protocol proposal for Friedreich ataxia molecular diagnosis using fluorescent and triplet repeat primed polymerase chain reaction. Transl Res. November 2010;156(5):309–314.
+
Ye
J, Ye
M, Kranz
S, Lo
P. A 10 year retrospective study of surgical outcomes of adult intracranial pilocytic astrocytoma. J Clin Neurosci. 2014 (12): 2160–2164.
+
Zesiewicz
TA, Greenstein
MB, Sullivan
KL,
et al. A randomized trial of varenicline (Chantix) for the treatment of spinocerebellar ataxia type 3. Neurology. 2012;78(8):545–555.
++
Cerebral palsy (CP), as defined by an Executive Committee jointly sponsored by United Cerebral Palsy Research and Educational Foundation and a similar group in the United Kingdom for the definition and classification of CP (Rosenbaum, 2007; Baxter, 2007):
…describes a group of permanent disorders of the development of movement and posture, causing activity limitation, that are attributed to non-progressive disturbances that occurred in the developing fetal or infant brain. The motor disorders of cerebral palsy are often accompanied by disturbances of sensation, perception, cognition, communication, and behavior, by epilepsy, and by secondary musculoskeletal problems.
Typically the CP label is given to children who sustain damage to the brain up to the age of 2 years.
Overall prevalence is approximately 2.11 per 1,000 births with a higher prevalence in children born before 28 weeks gestation (Oskoui, 2013).
Etiology
Pathogenesis: Precursors of CP include:
Periventricular leukomalacia (PVL) especially in pre-term infants (56%) (Krägeloh-Mann, 2007)
Intraventricular hemorrhage (IVH) in infants born prematurely (Lee, 2014) • Hypoxic-ischemic encephalopathy (HIE) (Lee, 2014)
++
Signs and Symptoms:
Delayed motor development, abnormal muscle tone (spastic, hypotonic, dsykinetic), impaired movement coordination and control (ataxia, lack of isolated control), postural deficits, impaired balance, impaired gross motor function, impaired fine motor function and impaired oral motor function.
Associated findings with CP include epilepsy (~1/2 of those with CP), cognitive deficits (~2/3 of those with CP), sensory deficits, growth delays, visual impairments and hearing impairments. (Krigger, 2006; Glader, 2017)
Categorization based on primary location of central nervous system involvement:
Spastic (approximately 70-75%) – involvement of motor cortex or white matter projections to and from cortical sensorimotor areas of the brain
Further categorization based on distribution of motor impairment:
Quadriplegia – involvement of whole body with greater involvement of upper body and arms
Hemiplegia – primary involvement of one side of body
Diplegia – bilateral lower extremities more involved than upper body and extremities
Predominant in infants born preterm (Himpens, 2008)
Dyskinetic (approximately 20%)-involvement of basal ganglia
Ataxic (<10%) – involvement of cerebellum
Hypotonic – location of neuropathology uncertain
Categorization based on severity: Gross Motor Function Classification System – Expanded and Revised (GMFCS) (Palisano, 1997; Palisano, 2007); specifically developed for children with cerebral palsy:
Based on child's self-initiated movement with emphasis on sitting, transfers and mobility.
Five levels of severity within 5 age bands: before 2nd birthday, 2-4 years, 4-6 years, 6-12 years and 12-18 years
Based on usual performance of gross motor skills within home, school and community settings
Scoring forms available on CanChild website: https://www.canchild.ca/system/tenon/assets/attachments/000/000/058/original/GMFCSER_English.pdf
Example for children between ages 6-12 years:
GMFCS Level I - "Children walk at home, school, outdoors and in the community. They can climb stairs without the use of a railing. Children perform gross motor skills such as running and jumping, but speed, balance and coordination are limited."
GMFCS Level II - "Children walk in most settings and climb stairs holding onto a railing. They may experience difficulty walking long distances and balancing on uneven terrain, inclines, in crowded areas or confined spaces. Children may walk with physical assistance, a handheld mobility device or used wheeled mobility over long distances. Children have only minimal ability to perform gross motor skills such as running and jumping."
GMFCS Level III - "Children walk using a hand-held mobility device in most indoor settings. They may climb stairs holding onto a railing with supervision or assistance. Children use wheeled mobility when traveling long distances and may self-propel for shorter distances."
GMFCS Level IV - "Children use methods of mobility that require physical assistance or powered mobility in most settings. They may walk for short distances at home with physical assistance or use powered mobility or a body support walker when positioned. At school, outdoors and in the community children are transported in a manual wheelchair or use powered mobility."
GMFCS Level V - "Children are transported in a manual wheelchair in all settings. Children are limited in their ability to maintain antigravity head and trunk postures and control leg and arm movements."
Diagnostic Testing:
Differential diagnosis is based on a thorough history and examination, clinical presentation, pattern of development of signs, familial history, and other factors that may influence the probability of specific diagnoses (Krigger, 2006)
MRI provides the best evidence during investigation of possible CP, with around 89% of children with CP having an abnormal MRI (Ashwal, 2004)
Genetic testing, metabolic testing, and nerve conduction studies should also be considered in the differential diagnosis (Andersen, 2016)
Neuroimaging findings in children with CP have been shown to be associated with the CP phenotype (Towsley, 2011; Lee, 2017):
Spastic diplegia with perinatal white matter injury
Spastic quadriplegia with severe periventricular leukomalacia or bilateral cortical and deep gray matter injury
Unilateral spastic CP with sequelae of cerebrovascular events
Dyskinetic CP with deep gray matter injury; and rigidity in cases involving anoxia.
+++
III. Prognosis and Sequelae
++
Trends of Progression:
The neurological insult resulting in cerebral palsy is non-progressive, however the clinical course changes with growth and aging.
Although the signs and symptoms associated with CP are considered non-progressive, motor impairments may become more obvious as the child develops from infancy through childhood and into adulthood (Smithers-Sheedy, 2013), particularly with growth spurts and weight-gain.
Expected sequelae (secondary impairments, structural and functional)
CP leads to multifactorial impairments of muscle growth, on which adaptation of the extracellular matrix is imposed due to altered loading from abnormal muscle pull and misalignment (Gough, 2012) resulting in musculoskeletal deformity, misalignment, joint contractures, muscle length impairments, and gait deviations that increase with CP severity.
Low bone mineral density (BMD) is prevalent in children with moderate to severe CP, particularly with decreased weight-bearing through long bones, and is associated with a significant increased risk of fractures (Henderson, 2002). The low BMD is present into young adulthood and adulthood (Marciniak, 2016).
Spasticity is found in 70% of individuals with CP and is a major contributor to the development of bony deformities and contractures (Tosi, 2009).
Individuals who are mobile when they become adults show a decline in ambulation skills over time and only a few over the age of sixty who walked well in previous years were able to maintain ambulation (Strauss, 2004).
Some adults with CP experience increased spasticity, decreased mobility, fatigue, decreased endurance, fractures, osteoporosis, gastrointestinal problems, urinary problems, oral motor disease, communication difficulties, lack of medical care, sexuality concerns, and reproductive concerns (Tosi, 2009).
Prognostic predictors to consider for future ambulation include sitting independently at 2 years of age, absence of visual impairment, absence of intellectual disability, and absence of epilepsy or seizures (Keeratisiroj, 2016).
+++
IV. Medical/Surgical Management
++
Medical/Pharmaceutical
Pharmacological management focuses primarily on reducing the effects of spasticity:
Oral medications including baclofen, valium, and dantrium
Injectable medications including botulinum toxin-A and phenol
Intrathecal delivery of baclofen using an implantable pump
Non-invasive neurostimulation protocols for spasticity (Naro, 2017)
Other medications may include anticonvulsants to control seizures and bisphosphonates to prevent loss of bone mass. (Novak, 2013)
Surgical techniques
Orthopedic surgery to address adaptive shortening of muscles, misalignment of bones and incongruity of joints including tendon lengthening, tendon transfers, osteotomies, and surgical fusion.
Unilateral proximal femoral osteotomy to improve leg alignment; improved gait and walking ability in children with spastic hemiplegic CP, while hip dysplasia persisted (Rutz, 2012)
Neurological surgeries, such as selective dorsal rhizotomy and intrathecal baclofen pump implantation, are typically reserved for children with more severe spasticity in quadriplegic and diplegic distributions.
+++
V. Key Aspects of Therapeutic Management
++
A coordinated and comprehensive approach to intervention for children with disabilities can lead to effective management strategies to address impairments, functional limitations, and participation restrictions across the lifespan reducing the burden of illness and disease.
Clinical management should include preventative management of secondary musculoskeletal impairments through the use of weight-bearing activities, positioning equipment, standing mobility devices, and vibration (Gudjonsdottir, 2002).
Common stretching techniques to address shortened musculature across joints can increase overall muscle and fascicle lengths immediately post-stretch in children with CP (Theis, 2013). The brevity of the effects of stretching require the incorporation of orthotics and other positioning devices to maintain the increased length that may be attained. Hinged ankle-foot orthoses are useful in controlling dynamic equinus deformity and reducing energy expenditure of gait in children with spastic hemiplegic CP (Balaban, 2007).
Muscle strengthening exercises should be directed towards the hip power generating muscles, which include the extensors, and general coordination exercises should be focused distally on the knee and ankle (Riad, 2008).
Strategies to address gait abnormalities include shortening the gait cycle, increasing walking speed, shortening the support phase, improving motor coordination, increasing stride length, decreasing stride frequency, and maintaining speed (Wang, 2012).
Interventions should also include motor learning principles to address gait deviations, balance disturbances, coordination deficits, and functional limitations relative to the individual child's unique presentation. Intervention strategies will change throughout the stages of a child's development and throughout adulthood as their needs, activities, and participation changes.
Treatment for gross motor functional deficits through use of intensive neurodevelopmental treatment (Lee, 2017) and task-specific training (Dutt, 2016; Kumar, 2016; Dolbow, 2016).
Improve motor activity and functional use of involved hand/arm with constraint-induced movement therapy (DeLuca, 2017)
Prevent bone loss through prolonged weight-bearing (Han, 2017); various options for assistive technology
Systematic review and evidence based clinical recommendations on dosing related to supported standing; strongest evidence to positively effect BMD at some sites (but not all), lower extremity ROM, hip biomechanics, and spasticity (Paleg, 2013).
Serial casting for improving and maintaining ankle range of motion, hip surveillance for maintaining hip joint integrity (Gordon, 2006; Effgen, 2008)
Fitness training for improving fitness and decreasing cardiovascular risk (Butler, 2010)
Personalized home programs for improving motor activity performance (Novak, 2006)
Strength training intervention for weakness that inhibits function (Dodd, 2002), particularly after selective dorsal rhizotomy surgery or intrathecal baclofen implantation (Graham, 2016; Bales, 2016). Evidence supports increase in strength but small to medium effect on improving functional activities.
Sensory intervention to improve tactile dysfunction and other possible sensory processing problems (Auld, 2016).
Hippotherapy, according to the American Hippotherapy Association, is therapist-directed use of equine movement to achieve functional outcomes for patients by engaging neuromotor, sensory and cognitive systems; moderate evidence exists to support improvement in functional tasks and activities (Kwon, 2015; Snider, 2007) and increased postural control during gait (Encheff, 2012). The activity on the horse may provide cardiovascular benefits, especially in children with severe impairments (Bongers, 2012). Interface with the horse also seems to provide psychosocial benefits including improved self-esteem and confidence (Frank, 2011).
Constraint-induced movement training (CIMT) has been used extensively to improve upper limb function in children who have hemiplegic CP. In a systematic review and meta-analysis of 27 randomized controlled trials on the subject, Chen et al (2014) found considerable variability in the type of constraint used (e.g., slings, gloves, mittens and casts), the length of time each day constraints were applied (ranging from 30 minutes to 6 hours) and the length of treatment (ranging from 2-10 weeks). Comparisons were complicated by a lack of consistency in outcome measurement and categorization of the participants. Chen used the International Classification of Functioning, Disability and Health (ICF) model to structure selected outcomes and reported medium effects at activity level, but only small effects at the body function and participation levels. Home-based treatment demonstrated better outcomes than clinic or camp-based settings. Zipp et al (2012) examined the effects of intensive CIMT on lower extremity function and found improvements in functional mobility, spatial temporal gait measures and balance.
Electrical stimulation has been used with mixed result (Chiu, 2014); systematic review of randomized controlled studies is confounded for multiple reasons, including categorization of participants, outcome measures used, terminology, muscle(s) stimulated, parameters used, and application (Carmick, 2014).
Focus on prevention of loss of function in adults with CP by monitoring mobility and ability to perform activities of daily living over time.
TheraTogs, orthotic garments with an optional applied strapping system (www.theratogs.com), have been shown to improve gross motor skills, balance, joint motion, posture, and a variety of gait kinematics in children with CP (Flanagan, 2009; El-Kafy, 2014)
TheraSuits, orthotic garments with an extensive external framework of elastic bands (www.suittherapy.com), combined with comprehensive therapy, have been shown to improve gross motor skills, balance and a variety of gait kinematics in children with CP (Bailes, 2010; Bailes, 2011).
Partial body weight supported treadmill training has been shown to improve gait parameters and gross motor skills in children with CP, especially in children with moderate to severe involvement and with more frequent and longer duration intervention (Mattern-Baxter, 2009; Damiano, 2009).
Variable evidence on the effectiveness of therapeutic taping to improve gross motor function and gait parameters in children with CP; no functional change in seated postural control in children with spastic quadriplegia GMFCS levels IV and V (Footer, 2006); no improvement in gross motor function and functional independence but positive effect on head, neck and foot position and arm, hand function in sitting (Simsek, 2011); In a systematic review Shamsoddini et al (2016) found positive impact on fine and gross motor abilities and functional independence in ADL, sitting/standing control and balance when used with children who have mild to moderate CP and when used as an adjunct to therapy.
Aquatic therapy has been shown to have positive effects on gait efficiency (Ballaz, 2011), gross motor function and joint range of motion (Chrysagis, 2009), gross motor function and physical activity enjoyment scores (Lai, 2015) across various types of CP and levels of severity.
+
Andersen
EW, Leventer
RJ, Reddihough
DS, Davis
MR, Ryan
MM. Cerebral palsy is not a diagnosis: A case report of a novel atlastin-1 mutation. J Paediatr Child Health. 2016;52:669–671.
+
Ashwal
S, Russman
BS, Blasco
PA,
et al. Practice parameter: diagnostic assessment of the child with cerebral palsy: Report of the Quality Standards Subcommittee of the American Academy of Neurology and the Practice Committee of the Child Neurology Society. Neurology. 2004;62:851–63.
+
Auld
ML, Johnston
LM, Russo
RN, Moseley
GL. A single session of mirror-based tactile and motor training improves tactile dysfunction in children with unilateral cerebral palsy: a replicated randomized controlled case series. Physiother Res Int. 2017;22(4). doi: 10.1002/pri.1674. Epub 2016 Aug 17.
+
Bailes
AF, Greve
K, Schmitt
LC. Changes in two children with cerebral palsy after intensive suit therapy: a case report. Pediatr Phys Ther. 2010;22(1):76–85.
+
Bailes
AF, Greve
K, Burch
CK, Reder
R, Lin
L, Huth
MM. The effect of suit wear during an intensive therapy program in children with cerebral palsy. Pediatr Phys Ther. 2011;23(2):136–142.
+
Balaban
B, Yasar
E, Dal
U, Yazicioglu
K, Mohur
H, Kalyon
TA. The effect of hinged ankle-foot orthosis on gait and energy expenditure in spastic hemiplegic cerebral palsy. Disabil Rehabil. 2007;29(2):139–144.
+
Bales
J, Apkon
S, Osorio
M, Kinney
G, Robison
RA, Hooper
E, Browd
S. Infra-conus single-level laminectomy for selective dorsal rhizotomy: technical advance. Pediatr Neurosurg. 2016;51(6):284–91.
+
Ballaz
L, Plamondon
S, Lemay
M. Group aquatic training improves gait efficiency in adolescents with cerebral palsy. Disabil Rehabil. 2011;33(17-18):1616–1624.
+
Baxter
P, Morris
C, Rosenbaum
P, Paneth
N, Leviton
A, Goldstein
M, Bax
M, Colver
A, Damiano
D, Graham
HK, Brien
GO. The definition and classification of cerebral palsy. Dev Med Child Neurol. 2007;49:1–44.
+
Bongers
B, Takken
T. Physiological demands of therapeutic horseback riding in children with moderate to severe motor impairments: an exploratory study. Pediatr Phys Ther. 2012;24(3): 252–257.
+
Butler
JM, Scianni
A, Ada
L. Effect of cardiorespiratory training on aerobic fitness and carryover to activity in children with cerebral palsy: a systematic review. Int J Rehabil Res. 2010;33:97–103.
+
Carmick
J. Letter to editor. Pediatr Phys Ther. 2014;26(4):487–488.
+
Chen
YP, Pope
S, Tyler
D, Warren
GL. Effectiveness of constraint-induced movement therapy on upper-extremity function in children with cerebral palsy: a systematic review and meta-analysis of randomized controlled trials. Clin Rehabil. 2014;28(10):939–953.
+
Chiu
HC, Ada
L. Effect of functional electrical stimulation on activity in children with cerebral palsy: a systematic review. Pediatr Phys Ther. 2014;26(3):283–288.
+
Chrysagis
N, Douka
A, Nikopoulos
M, Apostolopoulou
F, Koutsouki
D. Effects of an aquatic program on gross motor function of children with spasti cerebral palsy. Biol Exer. 2009;5(2):13–25.
+
Damiano
DL, DeJong
SL. A systematic review of the effectiveness of treadmill training and body weight support in pediatric rehabilitation. J Neurol Phys Ther. 2009;33(1):27–44.
+
DeLuca
SC, Trucks
MR, Wallace
DA, Ramey
SL. Practice-based evidence from a clinical cohort that received pediatric constraint- induced movement therapy. J Pediatr Rehabil Med. 2017;10(1):37–46.
+
Dodd
KJ, Taylor
NF, Damiano
DL. A systematic review of the effectiveness of strength-training programs for people with cerebral palsy. Arch Phys Med Rehabil. 2002;83:1157–64.
+
Dolbow
JD, Mehler
C, Stevens
SL, Hinojosa
J. Robotic-assisted gait training therapies for pediatric cerebral palsy: a review. J Rehabil Robotics. 2016;4:14–21.
+
Dutt
A, Kumar
BP. Efficacy of task-oriented training on balance in children with cerebral palsy. Ind J Cereb Palsy. 2016;2(2):94.
+
Effgen
S, McEwen
I. Review of selected physical therapy interventions for school age children with disabilities. Phys Ther Rev, 2008;13:297–312.
+
El-Kafy
E. The clinical impact of orthotic correction of lower limb rotational deformities in children with cerebral palsy: a randomized controlled trial. Clin Rehabil. 2014;28(10):1004–1014.
+
Encheff
J, Armstrong
C, Masterson
M, Fox
C, Gribble
P. Hippotherapy effects on trunk, pelvic, and hip motion during ambulation in children with neurological impairments. Pediatr Phys Ther. 2012;24(3):242–250.
+
Flanagan
A, Krzak
J, Peer
M, Johnson
P, Urban
M. Evaluation of short-term intensive orthotic garment use in children who have cerebral palsy. Pediatr Phys Ther. 2009;21(2):201–204.
+
Footer
CB. The effects of therapeutic taping on gross motor function in children with cerebral palsy. Pediatr Phys Ther. 2006;18(4):245–252.
+
Frank
A, McCloskey
S, Dole
R. Effect of hippotherapy on perceived self-competence and participation in a child with cerebral palsy. Pediatr Phys Ther. 2011;23:301–308.
+
Glader
L, Barkoudah
E. Epidemiology, etiology, and prevention of cerebral palsy. Wolters-Kluwer;
www.uptodate.com. Updated 2017. Accessed August 3, 2017.
+
Gordon
GS, Simkiss
DE. A systematic review of the evidence for hip surveillance in children with cerebral palsy. J Bone Joint Surg Br. 2006;88:1492–1496.
+
Gough
M, Shortland
AP. Could muscle deformity in children with spastic cerebral palsy be related to an impairment of muscle growth and altered adaptation? Dev Med Child Neurol. 2012;54(6):495–499.
+
Graham
D, Aquilina
K, Cawker
S, Paget
S, Wimalasundera
N. Single-level selective dorsal rhizotomy for spastic cerebral palsy. J Spine Surg. 2016;2(3):195–201.
+
Gudjonsdottir
B, Stemmons
Mercer V. Effects of a dynamic versus a static prone stander on bone mineral density and behavior in four children with severe cerebral palsy. Pediatr Phys Ther. 2002;14(1):38–46.
+
Han
EY, Choi
JH, Kim
SH, Im
SH. The effect of weight bearing on bone mineral density and bone growth in children with cerebral palsy: a randomized controlled preliminary trial. Medicine (Baltimore). 2017;96(10):e5896.
+
Hendersen
RC, Lark
RK, Gurka
MJ,
et al. Bone density and metabolism in children and adolescents with moderate to severe cerebral palsy. Pediatrics. 2002;110(1 Pt 1):e5.
+
Himpens
E, Van den Broeck
C, Oostra
A, Calders
P, Vanhaesebrouck
P. Prevalence, type, distribution, and severity of cerebral palsy in relation to gestational age: a meta-analytic review. Dev Med Child Neurol. 2008;50(5):334–40.
+
Keeratisiroj
O, Thawinchai
N, Siritaratiwat
W, Buntragulpoontawee
M, Pratoomsoot
C. Prognostic predictors for ambulation in children with cerebral palsy: a systematic review and meta-analysis of observational studies. Disabil Rehabil. 2017;40(2):135–143.
+
Krägeloh-Mann
I, Horber
V. The role of magnetic resonance imaging in elucidating the pathogenesis of cerebral palsy: a systematic review. Dev Med Child Neurol. 2007;49(2):144–151.
+
Krigger
K. Cerebral palsy: an overview. Am Fam Physician. 2006;73(1):91–100.
+
Kumar
C, Ostwal
P. Comparison between task-oriented training and proprioceptive neuromuscular facilitation exercises on lower extremity function in cerebral palsy: a randomized clinical trial. J Novel Physiother. 2016;6(291):2.
+
Kwon
J, Chang
H, Yi
S, Lee
J, Shin
H, Kim
Y. Effect of hippotherapy on gross motor function in children with cerebral palsy: A randomized controlled trial. J Alter Complement Med. 2015;21(1):15–21.
+
Lai
CJ, Liu
WY, Yang
TF, Chen
CL, Wu
CY, Chan
RC. Pediatric aquatic therapy on motor function and enjoyment in children diagnosed with cerebral palsy of various motor severities. J Child Neurol. 2015;30(2):200–208.
+
Lee
KH, Park
JW, Lee
HJ,
et al. Efficacy of intensive neurodevelopmental treatment for children with developmental delay, with or without cerebral palsy. Ann Rehabil Med. 2017;41(1):90–96.
+
Lee
RW, Poretti
A, Cohen
JS,
et al. A diagnostic approach for cerebral palsy in the genomic era. Neuromol Med. 2014;16(4):821–844.
+
Marciniak
C, Gabet
J, Lee
J, Ma
M, Brander
K, Wysocki
N. Osteoporosis in adults with cerebral palsy: feasibility of DXA screening and risk factors for low bone density. Osteoporosis Int. 2016;27(4):1477–84.
+
Mattern-Baxter
K. Effect of body weight supported treadmill training on children with cerebral palsy. Pediatr Phys Ther. 2009;21(1):12–22.
+
Miller
B. Cerebral palsy linked to prenatal causes. Fam Pract News. 2003;20(12):3–4.
+
Naro
A, Leo
A, Russo
M,
et al. Breakthroughs in spasticity management: are non-pharmacological treatments the future? J Clin Neurosci. 2017;39:16–27.
+
Novak
I, Cusick
A. Home programs in paediatric occupational therapy for children with cerebral palsy: where to start? Aust Occup Ther J. 2006;53:251–64.
+
Novak
I, McIntyre
S, Morgan
C, Campbell
L, Dark
L, Morton
N, Stumbles
E, Wilson
SA, Goldsmith
S. A systematic review of interventions for children with cerebral palsy: state of the evidence. Dev Med Child Neurol. 2013;55:885–910.
+
Oskoui
M, Coutinho
F, Dykeman
J, Jetté
N, Pringsheim
T. An update on the prevalence of cerebral palsy: a systematic review and meta-analysis. Dev Med Child Neurol. 2013;55(6):509–519.
+
Paleg
GS, Smith
BA, Glickman
LB. Systematic review and evidence-based clinical recommendations for dosing of pediatric supported standing programs. Pediatr Phys Ther. 2013;25(3):232–247.
+
Palisano
R, Rosenbaum
P, Walter
S, Russell
D, Wood
E, Galuppi
B. Development and reliability of a system to classify gross motor function in children with cerebral palsy. Dev Med Child Neurol. 1997;39(4):214–23.
+
Palisano
R, Rosenbaum
P, Bartlett
D, Livingston
M. Gross Motor Function Classification System – Expanded and Revised. Hamilton, Ontario, Canada: CanChild Centre for Childhood Disability Research, McMaster University; 2007.
+
Riad
J, Haglund-Akerlind
Y, Miller
F. Power generation in children with spastic hemiplegic cerebral palsy. Gait Posture. 2008;27(4):641–647.
+
Rosenbaum
P, Paneth
N, Leviton
A,
et al. A report: the definition and classification of cerebral palsy April 2006. Dev Med Child Neurol. 2007;49(SUPPL.109):8–14.
+
Rutz
E, Passmore
E, Baker
R, Graham
HK. Multilevel surgery improves gait in spastic hemiplegia but does not resolve hip dysplasia. Clin Orthop Relat Res. 2012;470(5):1294–1302.
+
Shamsoddini
A, Rasti
Z, Kalantari
M,
et al. The impact of Kinesio taping technique on children with cerebral palsy. Iran J Neurol. 2016;15(4):219–27.
+
Simsek
TT, Turkucuoglu
B, Cokal
N, Ustunbas
G, Simsek
IE. The effects of Kinesiotaping on sitting posture, functional independence and gross motor function in children with cerebral palsy. Disabili Rehabil. 2011;33(21-22):2058–2063.
+
Smithers-Sheedy
H, Badawi
N, Blair
E,
et al. What constitutes cerebral palsy in the twenty-first century? Dev Med Child Neurol. 2014;56(4):323–328.
+
Snider
L, Korner-Bitensky
N, Kammann
C, Warner
S, Saleh
M. Horseback riding as therapy for children with cerebral palsy. Phys Occup Ther Peds. 2007;27:5–23.
+
Stavsky
M, Mor
O, Mastrolia
SA, Greenbaum
S, Than
NG, Erez
O. Cerebral Palsy—Trends in epidemiology and recent development in prenatal mechanisms of disease, treatment, and prevention. Front Pediatr. 2017; 5:21.
+
Strauss
D, Ojdana
K, Shavelle
R, Rosenbloom
L. Decline in function and life expectancy of older persons with cerebral palsy. Neuro Rehabil. 2004;19(1):69–78.
+
Theis
N, Korff
T, Kairon
H, Mohagheghi
A. Does acute passive stretching increase muscle length in children with cerebral palsy? Clin Biomech. 2013;28(9-10):1061–1067.
+
Tosi
LL, Maher
N, Moore
DW, Goldstein
M, Aisen
ML. Adults with cerebral palsy: a workshop to define the challenges of treating and preventing secondary musculoskeletal and neuromuscular complications in this rapidly growing population. Dev Med Child Neurol. 2009;51(Suppl 4):2–11.
+
Towsley
K, Shevell
MI, Dagenais
L, Consortium
R. Population-based study of neuroimaging findings in children with cerebral palsy. Eur J Paediatr Neurol. 2011;15(1):29–35.
+
Wang
X, Wang
Y. Gait analysis of children with spastic hemiplegic cerebral palsy. Neural Regen Res. 2012;7(20):1578–1584.
+
Zipp
G, Winning
S. Effects of constraint-induced movement therapy on gait, balance, and functional locomotor mobility. Pediatr Phys Ther. 2012;24(1):64–68.
++
Two Types of Central Nervous System (CNS) tumors
Primary (Intrinsic) Tumors: arise within the CNS or its coverings and can be neuroepithelial or non-neuroepithelial tumors; gliomas are most common primary tumor
Rarely metastasize outside the CNS but may spread widely within.
Can be classified as benign or malignant
Metastatic tumors: arise from a distant site or locally invasive tumor (Ironside, 2012).
Metastatic brain tumors occur 4x's more often than primary brain tumors
Occur most commonly from lung, breast, kidney, and malignant melanomas. (Bondy, 1994)
The primary cause of CNS tumors is unknown, but the current theory is that they result from a combination of genetic and environmental factors
Genetic Factors
Environmental Risk Factors
Physical agents:
Low frequency electromagnetic fields (cell phones) not proven; exposure to cellphone 3G signal does not affect the biological features of brain tumor cells (Lui, 2015)
Ionizing radiation (X-ray diagnostics and high dose radiation therapy) high dose has been linked to secondary brain CA,
Chemical agents:
Research has just scratched the surface as to causal relationships (Bondy, 1994)
Many chemicals have been linked to tumors at industries, agricultural farms, and home
Exposure to vinyl chloride increases risk of glioma (National Cancer Institute, 2015)
Biologic agents:
+++
II. Diagnostic Procedures
++
General Signs and Symptoms:
Headaches: common initial symptom typically worse in the morning and with activity
Seizures: with convulsions, unusual sensations, loss of consciousness, occur in 55.3% of patients with low grade gliomas and 15.4% of patients with high grade glioms (Huang, 2017)
Mental change: behavior and intellect
Focal symptoms: help to identify location of tumor (Resources, 2001)
Frontal lobe: controls voluntary movement on the opposite side of the body, dominant hemisphere controls language and writing, and also controls intellectual function, thought processes, behavior and memory
Symptoms – "silent growers", hemiparalysis, seizures, short-term memory loss, impaired judgment, personality change, urinary frequency and urgency, gait disturbances, communication problems, behavioral changes, and impaired senses;
If the tumor is at the base of the frontal lobe, smell and vision can be impaired
Occipital lobe: involved in understanding of visual images and meaning of written words
Parietal lobe: interprets pain, temperature, touch, pressure, size, shape, body awareness, and is involved in hearing, reasoning and memory
Symptoms– seizures, language disturbances if tumor is on dominant side, inability to read, impaired speech, inability to write
Temporal lobe: involved in understanding of sounds, spoken words, emotion, and memory
Symptoms: seizures, difficulty with recognition of sounds, problems with memory, and vision impairment
Basal Ganglion: involved in procedural learning, voluntary movement control
Corpus Callosum: Provides communication between the two cerebral hemispheres
Symptoms: impaired judgment, defective memory, behavioral changes
Will invade other areas of brain
Seizures are uncommon
Diagnostic Testing:
Neurologic Exam including ophthalmoscopy to check for papilledema (edema of optic disc) resulting from increased ICP
Imaging: includes CT and MRI
MRI is the preferred method for diagnosing brain tumors (Chourmouzi, 2014)
Others: Angiography, holography, MRS- magnetic resonance spectroscopy, PET- position emission tomography, SPECT – single photon Emission computerized tomography, MEG- magnetoencephalography
Lab Test:
Lumbar puncture, myelogram, evoked-potentials, audiometry, endocrine evaluation, perimetry
Biopsy: needle and stereotaxic biopsy (Resources, Research, Information, 2001).
++
Primary Tumors
Benign Tumors–
Have a slow growth rate and are relatively noninvasive
If surgically inaccessible, growth will lead to increased intracranial pressure (ICP), neurological deficits, herniation syndromes and finally death. (Konecne, 1998)
Malignant Tumors –
Have a high growth rate and are usually invasive and infiltrative
Prognosis is better with younger age, high functional status, and radical surgical resection (Hardwidge, 2012)
Type of tumor (American Brain Tumor Association, 2015)
With standard treatment, median survival for adults with an anaplastic astrocytoma is about two to three years. For adults with more aggressive glioblastoma, treated with concurrent temozolamide and radiation therapy, median survival is about 14.6 months and two-year survival is 30%. However, a 2009 study reported that almost 10% of patients with glioblastoma live five years or longer.
Children with high-grade tumors (grades III and IV) tend to do better than adults; five-year survival for children is about 25%.
In addition, glioblastoma patients who have had their MGMT gene shut off by a process called methylation also have prolonged survival rates. The MGMT gene is thought to be a significant predictor of response.
Metastatic Tumor:
Associated with a poor prognosis as metastasis indicates primary site has already escaped control (Konecne, 1998)
Location of brain metastases:
80% cerebral hemispheres, 15 % cerebellum, 5% brainstem, (Eichler, 2007)
Prognostic factors:
Age, performance on the Karnofsky performance status (KPS) score, number of brain metastases, type of primary tumor, and systemic tumor activity
KPS score is the major determinant of survival, second to treatment regimen (Eichler, 2007)
WHO Tumor Grading System: type and grade of tumor affect prognosis (National Cancer Institute, 2015)
Grade I (low grade): tumor cells resemble normal cells, grow slowly, rarely spread to surrounding tissues, typically cured when tumor is removed by surgery
Grade II: tumor cells grow and spread slower than grade III and IV, may spread to surrounding tissues, may recur, may become a higher grade tumor
Grade III: tumor cells appear different that normal cells, they grow more quickly and are likely to spread to surrounding tissues
Grade IV (high grade): tumor cells do not resemble normal cells, grow and spread quickly, areas of dead cells may exist, usually cannot be cured
+++
IV. Medical and Surgical Management
++
Active Surveillance: Involves closely watching the person's condition without treatment unless the condition gets worse. Routine tests are performed. It is used to avoid or delay other treatment side effects and is only used for slow growing, asymptomatic tumors (National Cancer Institute, 2015)
Surgery: Goal is to remove tumor mass and to decrease ICP
Radiation Therapy: Can follow surgery or be used as an alternative. It can be delivered to localized area of the brain (Fine, 1993)
Whole brain radiation therapy (WBRT) + corticosteroids is most common for brain metastases (Eichler, 2007)
WBRT + corticosteroids increases survival time by 3-4 months
Stereotactic radiosurgery – halo device that uses a finely collimated beam with 1 mm accuracy
Chemotherapy: provides some increase in survival but no dramatic difference in management proven (Fine, 1993); but keep in mind blood brain barrier impedes penetration of drugs to the brain.
Chemo can be administered by oral, intravenous, direct tumor bed, or direct carotid perfusion routes
Immunotherapy: is a growing focus in cancer intervention (Gavin, 2012). There is a theoretical potential for biologic response modifiers, but participation in a clinical trial is strongly encouraged (Nabors, 2015)
Pain Management: quality of life is the highest priority and should guide clinical decisions (Nabors, 2015)
Recommend regular schedule of opioid with rescue dose as needed, education and psychosocial support, and adjunct analgesics as needed(Swarm, 2015)
Treatment being tested in clinical trials: proton beam radiation therapy, biologic therapy (National Cancer Institute, 2015)
Genomic Characterization: more than 500 glioblastoma tumors have been identified as a mutation. In one glioblastoma subtype, potential biomarker of treatment response is predictive (Brennan, 2013)
+++
V. Therapeutic Management
++
Health Care Team – can include nursing, nutrition, respiratory therapy, physical therapy, occupational therapy, speech therapy, social worker, and physician office staff
Oncologic Physical Therapy
Provide skilled therapy services for patients who have cancer of various stages
Practice in acute care, rehabilitation hospitals, skilled nursing facilities, home health, outpatient clinics, and wellness centers (APTA, 2011)
The therapist should be aware of expected symptoms in relation to tumor location in order to anticipate functional changes that may require PT modifications. Goal setting with client and family may be significantly different when cure is possible compared to when rapid decline is anticipated. Anticipated functional changes may include focusing therapy on restoring limb, movement, compensation techniques, sitting balance, standing balance, transfer training, and strength training (American Brain Tumor Association, 2014)
Intervention for Cerebral Hemisphere Tumors
Will vary according to location and may include spastic hemiplegia
Frontal Lobe – therapy for weakness, ataxia, urinary incontinence, hemiparalysis, seizures, short-term memory loss, impaired judgment, personality changes, urinary frequency and urgency, gait disturbances, communication problems, behavioral changes, and impaired senses
Occipital Lobe – therapy for partial seizures with visual phenomena, and homonymous hemianopsia or other visual disturbances
Parietal Lobe - therapy for partial sensory seizures, of cortical sensation, tactile localization, sterognosis, graphism, autopagnosia, anosognosia, and aphasia
Temporal Lobe – therapy for seizures, difficulty with recognition of sounds, problems with memory, and vision impairment
Basal Ganglia– therapy for contralateral choreooathetosis, contralateral dystonia, or other movement disorders
Corpus Callosum– Therapy for and prevention of tonic-clonic seizures, apraxia, and agraphia
+
Bondy, Wiencke, Wrensh, Kyritsis. "Genetics of primary brain tumors: a review." Journal of Neuro-Oncology. 18:69–81, 1994.
+
Brennan
CW, Verhaak
RGW, McKenna
A,
et al. The Somatic Genomic Landscape of Glioblastoma. Cell. 2013;155(2):462–477.
+
Chourmouzi,
Danai. "Imaging of Brain Tumors." Surgical Oncology Clinics of North America. Elsevier 23:629–684, 2014.
+
Dunn
GP, Fecci
PE, Curry
WT. Cancer Immunoediting in Malignant Glioma. Neurosurgery. 2012;71:201–223.
+
Eichler
AF, Loeffler
JS. Multidisciplinary management of brain metastases. Oncologist. 2007; 12:884–898.
+
Fine
HA, Dear
KB, Loeffler
JS, McBlack
PL, Canellos
GP. Meta-analysis of radiation therapy with and without adjuvant chemotherapy for malignant gliomas in adults. Cancer. 1993;71(8):2585–2597.
+
Hardwidge,
Hettige. "Tumours of the Central Nervous System". Surgery (Oxford). Elsevier 30: 155–161, 2012.
+
Huang
C, Chi
X, Hu
X
et al.. Predictors and mechanisms of epilepsy occurrence in cerebral gliomas: What to look for in clinicopathology. Exp Mol Pathol. 2017;102(1):115–122.
+
Ironside, Alasatair, James. "Pathology of tumours of the central nervous system." Surgery (Oxford). Elsevier 30:107–115, 2012.
+
Khurana, Teo, Kundi, Hardell, Carlberg. "Cell phones and brain tumors:a review including long-term epidemiologic data." Surgical Neurology. 2009: 2015–215
+
Kitai
R, Matsuda
K, Adachi
E, Saito
Y, Nakajima
T, Takeuchi
H, Kazufumi
S, Imamura
Y, Kubota
T . Epstein-Barr Virus-Associated Primary Central Nervous System Lymphoma in the Japanese Population. Neurologia medico-chirurgica. 2010;50(2):114–118.
+
Liu
Y, Li
G, Fu
X
et al.. Exposure to 3G mobile phone signals does not affect the biological features of brain tumor cells. BMC Public Health. 2015;15(1):764. doi:10.1186/s12889-015-1996-7.
+
McNeil, Caroline. Vitamins during Pregnancy Linked to Lower Risk of Childhood Brain Tumors. Journal of the National Cancer Institute. 1997;89:1481–1482.
+
Wu,
Haber. Molecular Biology of Brain Tumors. Neurosurgery Clinics of North America. 1994;5:127–132.
+++
Cerebrovascular Accident (CVA) - ischemic/hemorrhagic - "Stroke"
++
A stroke or a cerebrovascular accident (CVA), occurs when blood supply to the brain is blocked or when a vessel in the brain bursts, depriving part of the brain of oxygen. Lack of oxygen and nutrients cause brain cells to die (NIH, 2006).
Incidence
Stroke is the US fourth leading cause of death (NHLBI, 2012)
Approximately 795,000 new or recurrent cerebral vascular accidents occur per year in the US (NHLBI, 2012)
About every 40 seconds in the US someone has a stroke (American Heart Association)
Approximately 130,000 Americans die from stroke every year (CDC, 2014).
In the U.S., someone dies from a stroke approximately every 4 minutes (AHA, 2015), but rate of stroke death fell by 21.2% from 2001-2011 (AHA, 2015).
Prevalence (NHLBI, 2008)
- Increases markedly with age
- Higher in blacks than in whites at all ages
Incidence among African-Americans is double that of white Americans, whereas, Hispanics, Native, and Asian Americans incidence and mortality rates similar to white (NIH, 2006).
- Higher in males than in females but stroke tends to be more severe in women, with a 1-month case fatality of 24.7% compared with 19.7% for men (Appelros, 2009)
- "Stroke belt and stroke buckle": 12 states and DC --stroke death rates >10 % higher than the rest of the country (NIH, 2006; NSA, 2006).
Stroke Precursor: Transient ischemic attack (TIA) –
Caused by a temporary cerebral artery blockage that causes symptoms to occur rapidly and last less than five minutes causing no permanent injury to the brain
Not a stroke, but precursor to stroke and a warning of potentially a more serious and debilitating stroke (NIH, 2006).
Two forms of CVA:
Ischemic stroke (88%) (Jorge, 2013) in which blood flow is blocked
Mechanisms: blockage/atherosclerosis to one of the cerebral arteries, embolism, decreased perfusion, and thrombosis (NIH, 2006; Stroke Center, 2006; Mohr, 1997).
Hemorrhagic stroke (12%) (Jorge, 2013) in which blood leaks at a rupture site, depriving the intended tissue
Risk Factors
- Non-modifiable risk factors:
Age, gender, and race (NIH, 2006; Sacco et al, 2006)
Family history of stroke in a first-degree relative increases risk by 50% (Liao, 1997).
Individuals that have had a stroke are at higher risk and are more likely to have another stroke or TIA. Approximately 60% of strokes occur in patients who have had a prior stroke. (NSA, 2017)
- Modifiable risk factors:
Hypertension, cardiac disease, diabetes, hyperlipidemia, cigarette smoking, alcohol, atrial fibrillation.
Modifiable lifestyle factors: smoking, obesity, decreased physical activity, poor diet, and acute triggers (emotional stress)
- Most patients who experience a stroke have more than one risk factor; Having multiple risk factors could also predispose a patient to have recurrent strokes (Mohr, 1997).
- Predictors of stroke include: age over 60 years, having diabetes, focal symptoms that include weakness or speech impairment, and having a TIA that lasts over 10 minutes (Go, 2013).
Poor public awareness of stroke: among adults >50, 38% did not know stroke occurs in the brain (NSA, 2006).
- Among those who are at highest risk for coronary heart disease, black men were the least aware of hyperlipidemia. Black women were the least likely to be treated for hyperlipidemia, while white men were the most likely. These findings could explain the difference between races as far as prevalence of stroke (Safford, 2015)
The direct and indirect cost of stroke in 2009 was $38.6 billion (Go, 2013)
Costs: On average, stroke costs the US an estimated $34 billion total each year including the cost of health care services, medications to treat stroke, and missed days of work (Mozaffarian, 2015).
+++
II. Diagnostic Procedures
++
The diagnosis of a stroke is based on history and physical examination (Ferro, 1998).
Signs and Symptoms by location (NSA, 2006):
Cerebral stroke (depending on specific location of infarction)
Contralateral hemiplegia, hemiparesis, and hemianesthesia, aphasia, depression, post-stroke fatigue, memory deficits, severe headaches, behavioral changes (i.e., personality changes or emotional lability), balance impairment (McGeough, 2009, Stroke Association, 2012), perceptual impairment;
Posterior cerebral artery stroke
Visual field deficits (contralateral homonomous hemianopsia) if occipital lobe or optic radiations are affected, Visual inattention; L hemisphere damage may result in alexia (inability to read)
most common impairments: motor paresis (65%), followed by visual field deficits (54%) and confusion or agitation (43%) (Ng, 2005).
Anterior cerebral artery stroke
Predominant lower extremity contralateral hemiparesis/hemiplegia, some sensory loss possible in lower extremities, incontinence, problems with bimanual tasks, apraxia
Middle cerebral artery stroke (most common)
Predominant contralateral hemiparesis and hemisensory loss of upper extremity, trunk and face (may also affect lower extremities if deep white matter of corona radiata and posterior limb of internal capsule affected)
Possible dysarthria and dysphagia
Contalateral homonymous hemianopsia (may occur from optic radiation damage) • R hemisphere: visual-spatial or somatic perceptual impairment
L hemisphere: communication impairment (various aphasias)
Cerebellar stroke
Dizziness, nausea and vomiting; ipsilateral ataxia and hypotonia; nystagmus, impaired balance, incoordinated gait and speech.
Brainstem stroke
Small isolated lesions may result in alternating hemiplegia/hemiparesis (motor symptoms in ipsilateral motor cranial nerve distribution – facial and jaw weakness, eye muscle weakness - but in contralateral limbs and trunk) with unilateral medial brainstem damage, or alternating hemianesthesia (sensory impairment in ipsilateral face and in contralateral limbs and trunk) with unilateral lateral brainstem damage. Other symptoms could include impaired breathing, heart rate, and blood pressure; impaired eye movements/strabismus; hearing, speech and swallowing impairment; vestibular dysfunction and related balance problems
Diagnostic Testing
Tests include blood tests (Doppler test and duplex scanning), Evoked response tests (stimulated by hearing, body sensation, and vision), electrocardiogram, head CT, MRI (NIH, 2006; Stroke Center, 2006, Senelick 1994).
Confirmed by imaging:
CT of brain, can identify ischemic vs hemorrhagic CVA, including use of contrast to clearly demonstrate cerebral blood flow, leaks (acute blood is bright on CT), blockage or ischemia;[tip: on CT, the brain tissue appears immediately adjacent to the white skull]
MRI of brain can identify CVA earlier than CT (within 8-12 minutes will see absent flow void) [tip: on MRI, there is a dark layer between the white skull and very detailed cortex; on T1-weighted image, fat is bright and CSF is dark; on T2-weighted image, water (and CSF) is bright and most pathology is bright.]
CTA and MRA can reveal the large blood vessels in the brain to help localize the site of the clot and the flow of blood through the brain. (NHLBI, 2014).
Carotid ultrasound can reveal plaque that narrows or blocks the carotid arteries (NHLBI, 2014).
Echocardiogram to demonstrate a clot in the heart, that may be sending emboli to the brain as a cause of stroke can diagnose and prevent the occurrence of cerebrovascular accidents (Golipuor, 2011).
Employing various MR sequences and a multimodal approach to imaging can correctly identify those patients who have experienced an acute stroke that may or may not benefit from treatment and/or therapy (Naggara, 2014).
Neuroangiography is an invasive technique that can be used for extensive diagnosis of cerebral artery disease and to aid in the treatment of various neurological disorders, such as an acute CVA (Ahn, 2013).
NIH Stroke Scale, Glasgow Coma Scale, Hunt and Hess Scale, Modified Rankin Scale, and Barthel Index (NIH, 2006).
++
Stroke is nonprogressive, and is a leading cause of long term disability in the US (CDC, 2009; Smith, 2016)
50% of stroke survivors experience ongoing disability, and 30% require assistance for activities of daily living (Smith, 2016)
Following a cerebral insult, the CNS can undergo reorganization or plasticity during the recovery process, with variable recovery. Prognosis depends on patient's pre-morbid condition, age, sex, lesion location, degree of damage, and rehabilitation treatment. Other long-term deficits may include:
Physical deficits
Hemiparesis on contralateral side, speech deficits, incoordination, weakness, gait deviations, pain
Some studies have shown that up to 85% of stroke survivors experience long-lasting hemiparesis (Yavuzer, 2008)
Following a stroke, women have greater disability than men (American Heart Association)
Muscle tone
Muscle performance
UE usually biased toward flexion with LE biased toward extension movement patterns
Impaired force production (eccentric and concentric), speed and power impaired
Abnormal synergies may develop
Absence of voluntary control of the LE within the first week following the stroke and no development of normal UE synergies after 4 weeks is associated with a poor outcome at 6 months (Kwakkel, 2003)
Up to 2 years following the stroke there may still be some potential for improvements (Donaldson, 2009)
Emotional Deficits
Anxiety, Panic attacks, flat affect, sleeping problems, lower self-esteem, depression, lethargy, withdrawal, labiality (Senelick 1994, Coffey 2000, Villarosa 1993).
Cognitive Deficits
Aphasia, dementia, hemispatial neglect, trouble with memory and attention (Reith 1997, Burn 1997)
Secondary Impairments
Development of contractures
UE: Finger, wrist and elbow flexion contractures
Cardiovascular secondary impairments due to immobility (Stroller, 2013)
Pusher syndrome
Patients who have a stroke may be at risk for this secondary impairment, associated with an abnormal vertical orientation of the body toward the side of the lesion. The person perceives that he/she is oriented vertically in relation to gravity, when in fact, the body is "pushed" toward the affected side. The focus of the physical therapist in treating this disorder is the visual capability of the patient to correct himself/herself, which has usually not been compromised (Karnath, 2008).
Wallenberg's syndrome
This syndrome is usually secondary to a lateral medullary infarction and involves the cranial nerves in that area (usually CN IX and X). The damage to these nerves causes dysphagia in 51-94% of patients who experience this type of infarction. If intervention is begun soon enough, the dysphagia can be reversed; if no intervention is provided, the patient could progress to nonoral feeding status (Aydogdu, 2001).
Hemispatial neglect
This perceptual impairment usually occurs after a right hemisphere infarction; the person tends to neglect items on the side contralateral to his/her lesion (left side neglect in a person with right CVA). Sometimes the person doesn't acknowledge or even rejects their own body on the affected side (Parton, 2004).
Among survivors of ischemic stroke who were 65 years of age, these disabilities were observed 6 months after stroke (Kelly-Hayes, 2003):
- 50% had some hemiparesis
- 30% were unable to walk without some assistance
- 26% were dependent in activities of daily living
- 19% had aphasia
- 35% had depressive symptoms
- 26% were institutionalized in a nursing home
- 46% had cognitive deficits
According to the National Stroke Association (NSA, 2014):
- 10% of stroke survivors recover almost completely
- 25% recover with minor impairments
- 40% experience moderate to severe impairments requiring special care
- 10% require care in a nursing home or other long-term care facility
- 15% die shortly after the stroke
Reoccurrence Rates
Men have a 30-80% higher chance of reoccurrence than women
25% who recover from first stroke will have another stroke within 5 years (NIH, 2006).
Estimated US Mortality Rates
8-12% of ischemic strokes and 37-38% of hemorrhagic strokes result in death within 30 days in patients aged 45-64. (Rosamond, 1999).
From 1998 to 2008, the annual stroke death rate decreased 34.8%, and the actual number of stroke deaths declined 19.4% (NHLBI, 2008)
Comorbidities, like diabetes, can increase the risk of death after a stroke, recurrent stroke, and myocardial infarction.
+++
IV. Medical and Surgical Management
++
Most important intervention is education and prevention
Acute stroke needs immediate medical intervention-dissolving the blood clot if ischemic stroke or stopping the bleeding of a hemorrhagic stroke (NIH, 2006)
Before administering any type of treatment for an acute stroke, the physician must determine whether the event is an ischemic stroke or hemorrhagic stroke using bloodwork, imaging, and evaluating signs/symptoms. Some conditions, such as sepsis, space-occupying lesions, and seizures, can mimic stroke, and should be ruled out (Goldstein, 2014).
Ischemic Stroke
Emergency Room Procedures
Thrombolytic: Tissue Plasminogen Activator (tPA) for ischemic stroke only (Stanford, 2006)
tPA works by dissolving the clot and restoring some blood flow to the part of the ischemic brain. Must be given emergently within 3-4.5 hours of onset of symptoms.
Brain hemorrhage is a possible complication of tPA administration in an acute ischemic stroke (Goldstein, 2014).
Anticoagulant: heparin and warfarin
Antiplatelet: Aspirin
"Treatment with aspirin is associated with nine fewer deaths and nonfatal strokes over the ensuing few weeks per 1,000 persons treated." (Goldstein, 2014).
Low-dose aspirin therapy (anti-platelet) to prevent recurrent ischemic stroke (Mant, 2007)
Surgical Treatment:
Embolectomy and revascularization
Carotid Endarterectomy (Preventive):
Thrombectomy is another treatment option in which the large blood clot is removed by a stent retriever. Results show that mechanical thrombectomy in acute stroke due to internal carotid artery and middle cerebral artery occlusion displays high rate of recanalization and favorable functional outcomes (Yu, 2016).
Hemorrhagic Stroke
Blood transfusion and medication to coagulate and stop the bleeding
Medication may be used to decrease intracranial pressure, lower the pts BP, prevent seizures
Surgical treatment
Surgical clipping to prevent the vessel from bleeding, Endovascular embolization or coiling to prevent the bleeding from getting worse (Olson, 1990, Mayo Clinic 2013).
Neuroprotectants to protect the brain from secondary injury caused by stroke (NIH, 2006).
Treat and reduce current risk:
Hypertension, diabetes, and heart disease, changing lifestyle (NSA, 2006).
Controlling blood pressure consistently reduces the risk for recurrent stroke, myocardial infarction, and vascular death (Towfighi, 2014). Stroke incidence is reduced by 36% with a 10mm Hg decrease in systolic blood pressure or a 5mm Hg decrease in diastolic blood pressure (Towfighi, 2014).
Some patients with a history of stroke and other predisposing risk factors may exhibit an apparently treatment-resistant type of hypertension. This fact emphasizes the importance of an individualized blood pressure control approach. (Howard, 2015).
+++
V. Implications for Therapeutic Management
++
Possible Physical Therapy Preferred Practice Patterns (APTA, 2017):
5D- Impaired Motor Function and Sensor Integrity Associated with Non-Progressive Disorders of the Central Nervous System – Acquired in Adolescence or Adulthood
5A: Primary prevention/risk prevention for loss of balance and falling
5I- Impaired Arousal, Range of Motion, and Motor Control Associated with Coma, Near Coma, or Vegetative State.
Role of the PT:
Treat symptoms of stroke including underlying impairments and their impact on functional activity, Education, Maintain ROM, skin integrity, posture, strength; enhance motor control, self-care, ADLs. Monitor ventilation and pain. Assess for assistive device and/or splinting. (Furie 2010, Rodgers 1999)
Therapeutic treatment goal-- functional recovery.
General Physical Therapy Guidelines
Early rehabilitation intervention enhances recovery process and to prevent a quick decline (Duncan et al, 2004;Pollock, 2006).
"Initiating post-stroke rehabilitation soon after the onset of an acute stroke appears to be the most important factor associated with early discharge from the hospital; in addition, earlier admission is recognized as a relevant and favorable prognostic factor." (Safer, 2015)
Late rehabilitation using reinforced feedback (6 months post-stroke) may also be effective in improving motor performance. (Piron, 2010)
Multidisciplinary Team Approach to Rehabilitation
Speech, occupational and psychological therapy. Stroke unit care, Home health (Stroke Unit, 2006, Schouten 2008).
Multidisciplinary team meetings with efforts to make them more effective can result in improved patient outcomes and decreased length of stay after stroke. The model includes a planned agenda for each team meeting, with structured topics to discuss to limit distraction or extraneous discussion. Each patient is discussed methodically, with the goal being functional improvement of the patient and discharge planning based on independence in daily life (Tyson, 2015).
Can include physiatrist, neurologist, nurse, PT, OT, speech pathologist, social worker (NSA, 2017)
Intensity of rehabilitation
Patients with stroke who are treated with very high-intensity rehabilitation protocols have a lower rate of readmission to a hospital within 30 days than do those who are treated with a very low-intensity protocol. Patients with comorbidities and higher severity of illness were determined suitable for high-intensity rehabilitation. Intensity in this study was designated using charges billed by the physical therapist in one visit (higher-intensity treatments included more charges than the low-intensity treatments) (Andrews, 2015).
Evaluation and ongoing reassessment to include patient and family education, exercises (muscle length, ROM, stretching), ADL, device and equipment training, injury prevention, soft tissue mobilization/ manipulation, airway clearance, use of physical agents, repetitive task training for lower extremities (French et al, 2007),
Gait training overland (small gains in 6 MWT and gait speed) or with electromechanical or robot-assist, postural training, balance training, fall prevention (Merholtz, 2007; States, 2009)
Treatment for coordination of movement, balance, motor relearning, and muscle reeducation. Forced use / Constraint-Induced Therapy (Hammer, 2009).
Treatment for spasticity and rigidity of affected muscles. Prevention of disuse muscle atrophy.
For patients who have finger and wrist flexor spasticity following stroke, injection of botulinum toxin type A followed by prolonged stretch with adhesive tape can decrease spasticity in the wrist and finger flexor muscles (Santamato, 2015).
Use of orthotics in order to prevent development of contractures and compensate for motor deficits (Lannin, 2011)
Choice of appropriate orthotic should be individualized to patient
Hinged ankle foot orthoses (HAFOs) have been shown to decrease the fall risk in adults with longstanding hemiplegia following a stroke, as well as improve static postural control (Lusardi, 2013).
Researchers studied a group of patients undergoing stoke therapy who had unilateral hemiplegia with weakness or paralysis to the dorsiflexor muscles and found that using an anterior ankle-foot orthosis (AAFO) significantly increased walking speed during the timed up and go (TUG) and timed up and down stairs (TUDS) and may also increase the patients' ability to perform these tasks safely (Chen, 2014).
Cardiovascular fitness: determined to be beneficial to pt's as an intervention post-CVA
Must be aware of
Risk of seizures, A diagnosis of paroxysmal non-valvular atrial fibrillation (NVAF), Metabolic syndrome, personal contextual factors like age, height, weight, activity level pre-stroke, impairments that he/she had pre or post stroke to address in treatment (MacKay-Lyons et. al).
Cognitive and mood screening is essential to guide functional training through physical and occupational therapy. Psychological well-being influences every facet of stroke treatment and should always be considered by every healthcare professional; referrals should be completed when appropriate (Gurr, 2015).
Cognitive treatment to increase alertness and attention (Lincoln, 2000/2009).
The healthcare provider should be aware of the possibility of depression after a person experiences a stroke. The age groups of stroke survivors who are most likely to experience depression are ages 25-54 and 55-64. Also, those of any age who are experiencing other stressors (financial, family, social, health issues) are at higher risk for developing depression after surviving a stroke (McCarthy, 2015).
Others
Mirror therapy can be used to improve hand functioning (Yavuzer, 2008).
Transcranial Direct Current Stimulation (TDCS)
Emerging technique to either stimulate the affected side after a stroke, or to decrease the excitability of the unaffected side so that it does not compensate for the affected side during rehabilitation (Schlaug, 2008). This allows for the affected side to adapt to the changes in the brain that follow a stroke which makes this new technology a promising tool for clinicians in the coming future.
TDCS combined with Virtual Reality Therapy may be a beneficial method to enhance recovery of the paretic upper extremity in patients following a stroke (Lee and Chun, 2014).
Education of how and why the CVA occurred and what to expect in the future, minimizes functional disability (Duncan et al, 2004;Pollock, 2006)
+++
VI. Consumer and Professional Resources
++
American Stroke Association
Evidence-Based Review of Stroke Rehabilitation
National Stroke Association
- http://www.stroke.org/site/PageNavigator/HOME
- A website designed to describe a stroke and to help raise awareness about the diagnosis.
- Support group contact available
- StrokeSmart Magazine provides tips on money, life after a stroke, and wellness.
- Stroke Help Line to answer questions about stroke prevention, treatment, and recovery.
http://www.neuropt.org/special-interest-groups/stroke/resources
- A special interest group of the Neurology Section of the APTA
- Has information from the National Stroke Association, articles dealing with stroke, assessment tools and outcome measures, as well as several other resources for physical therapists treating patients who have had a stroke.
http://strokengine.ca/assess/
http://strokengine.ca/intervention/
+
Ahn
SH, Prince
EA, Dubel
GJ. Basic neuroangiography: review of technique and perioperative patient care. Semin Interv Radiol. 2013;30(3):225–233.
+
American Heart Association. 1999 Heart and Stroke Statistical Update. Dallas, Texas: American Heart Association. 1998.
+
Andrews
A, Li
D, Freburger
J. Association of rehabilitation intensity for stroke and risk of hospital readmission. PhysTher December 2015;95(12):1660–1667 8p.
+
Appelros
P, Stegmayr
B, Terrent
A. Sex differences in stroke epidemiology: a systematic review. Stroke. 2009: 40:1082–90
+
APTA, Guide to Physical Therapist Practice 2nd ed.,2003.
+
Aydogdu
I, Ertekin
C, Tarlaci
S, Turman
B, Kiylioglu
N, Secil
Y. Dysphagia in lateral medullary infarction (Wallenberg's syndrome): An acute disconnection syndrome in premotor neurons related to swallowing activity? Stroke. 2001:32:2081–2087.
+
Boffeli
TJ, Collier
RC. Minimally Invasive Soft Tissue Release of Foot and Ankle Contracture Secondary to Stroke. J Foot Ankle Surg/j.jfas.2013.06.008.
+
Burn
J, Dennis
M, Bamford
J, Sandercock
P, Wade
D, Warlow
C (December 1997)."Epileptic seizures after a first stroke: the Oxfordshire Community Stroke Project". BMJ 315 (7122): 1582–7
+
Centers for Disease Control and Prevention (CDC). Prevalence and most common causes of disability among adults--United States, 2005. MMWR Morb Mortal Wkly Rep. 2009;58(16):421–426.
+
Chen
C-L, Teng
Y-L, Lou
S-Z, Chang
H-Y, Chen
F-F, Yeung
K-T. Effects of an Anterior Ankle-Foot Orthosis on Walking Mobility in Stroke Patients: Get Up and Go and Stair Walking. Arch Phys Med Rehabil. 2014;95(11):2167–2171.
+
Coffey
C. Edward, Cummings
Jeffrey L, Starkstein
Sergio, Robinson
Robert (2000). Stroke - the American Psychiatric Press Textbook of Geriatric Neuropsychiatry (Second ed.). Washington DC: American Psychiatric Press. pp. 601–617.
+
Donaldson
C, Tallis
R, Miller
S, Sunderland
A, Lemon
R, Pomeroy
V. Effects of Conventional Physical Therapy and Functional Strength Training on Upper Limb Motor Recovery After Stroke: A Randomized Phase II Study. Neurorehabil Neural Repair. 2009;23(4):389–397.
+
Duncan
PW, Zorowitz
R, Bates
B, Choi
JY, Glasberg
JJ, Graham
GD
et al.. Management of adult stroke rehabilitation care. A clinical practice guideline. 2005;36 e100.
+
Ferro
J, Pinto
A, Falcão
I, Rodrigues
G, Ferreira
J, Falcão
F,
et al. Diagnosis of Stroke by the Nonneurologist. Stroke. 1998; 29:1106–1109.
+
French
B, Thomas
LH, Leathley
MJ, Sutton
CJ, McAdam
J, Forster
A, Langhorne
P, Price
CIM, Walker
A, Watkins
CL. Repetitive task training for improving functional ability after stroke. Cochrane Database of Systematic Reviews 2007, Issue 4. Art. No.: CD006073. DOI: 10.1002/14651858.CD006073.pub2
+
Furie
K, Kasner
S, Adams
R, Albers
G, Bush
R, Fagan
S, Halperin
J, Johnston
C, Katzam
I, Kernan
W, Mitchell
P, Ovbiagele
B, Palesch
Y, Sacco
R, Schwamm
L, Wassertheil-Smoller
S, Turan
T, Wentworth
D. Guidelines for the Prevention of Stroke in Patients With Stroke or Transient Ischemic Attack: A Guideline for Healthcare Professionals From the American Heart Association/American Stroke Association. American Heart Association Journal. 2010.
http://stroke.ahajournals.org/content/42/1/227.full +
Go
AS, Mozaffarian
D, Roger
VL, Benjamin
EJ, Berry
JD, Borden
WB, Bravata
DM, Dai
S, Ford
ES, Fox
CS, Franco
S, Fullerton
HJ, Gillespie
C, Hailpern
SM, Heit
JA, Howard
VJ, Huffman
MD, Kissela
BM, Kittner
SJ, Lackland
DT, Lichtman
JH, Lisabeth
LD, Magid
D, Marcus
GM, Marelli
A, Matchar
DB, McGuire
DK, Mohler
ER, Moy
CS, Mussolino
ME, Nichol
G, Paynter
NP, Schreiner
PJ, Sorlie
PD, Stein
J, Turan
TN, Virani
SS, Wong
ND, Woo
D, Turner
MB. Heart Disease and Stroke Statistics—2013 Update A Report From the American Heart Association. Circulation. 2013;127(1):e6–e245.
+
Goldstein
L. Modern medical management of acute ischemic stroke. Methodist Debakey Cardiovasc J. April 2014;10(2):99–104.
+
Golipuor
F, Zamani
A, Zarrineh
M. Evaluation of Echocardiography Results in Patients with Acute Cerebrovascular Accidents (CVA). JUMS Journal. June 2011;19(75):58–65p.
+
Gurr
B, Dendle
J. Assessment of stroke survivors on an inpatient rehabilitation unit. Br J Neuro Nurs. August 2015;11(4):179–184 6p.
+
Hammer
AM, Lindmark
B. Effects of forced use on arm function in the subacute phase. Phys Ther.2009; 89: 993–995
+
Howard
V, Tanner
R, Muntner
P,
et al. Apparent treatment-resistant hypertension among individuals with history of stroke or transient ischemic attack. Am J Med. 2015:128(7):707.
+
Hoyert
DL, Kochanek
KD, Murphy
SL. Deathes: Final Data for 1997. National Vital Statistics Reports; Vol. 47 no. 19. Hyattsville, Maryland: National Center for Health Statistics. 1999.
+
Jorge
M, Lusardi
M, Nielson
C, Orthotics and Prosthetics in Rehabilitation. Elsevier Health Sciences; 2012.
+
Karnath
H, Broetz
D. Understanding and treating "pusher syndrome". Phys Ther. December 2008:83(12):1119–1125.
+
Kelly-Hayes
M, Beiser
A, Kase
CS, Scaramucci
A, D'Agostino
RB, Wolf
PA. The influence of gender and age on disability following ischemic stroke: the Framingham study. J Stroke Cerebrovasc Dis.2003;12:119–126.
+
Kwakkel
G, Kollen
BJ, van der Grond
J, Prevo
AJH. Probability of regaining dexterity in the flaccid upper limb: impact of severity of paresis and time since onset in acute stroke. Stroke J Cereb Circ. 2003;34(9):2181–2186.
+
Lannin
N, Lannin
NA, Ada
L. Neurorehabilitation splinting: Theory and principles of clinical use. NeuroRehabilitation. 2011;28(1):21–28.
+
Lee
SJ, Chun
MH. Combination Transcranial Direct Current Stimulation and Virtual Reality Therapy for Upper Extremity Training in Patients With Subacute Stroke. Arch Phys Med Rehabil. 2014;95(3):431–438.
+
Liao
D, Myers
R, Heiss
G,
et al. Familial history of stroke and stroke risk. The Family Heart Study. Stroke. 1997;28(10):1908–1912.
+
Lincoln
N, Majid
M, Weyman
N. Cognitive rehabilitation for attention deficits following stroke. Cochrane Database of Systematic Reviews 2000, Issue 4. Art. No.: CD002842. DOI: 10.1002/14651858.CD00284
+
Lusardi
M, Jorge
M, Nielsen
C. Orthotics and Prosthetics in Rehabilitation, 3rd edition. St. Louis, Missouri: Elsevier; 2013: 235
+
Mant
J, Hobbs
FDR, Fletcher
K, Roalfe
A, Fitzmaurice
D, Lip
GYH, Murray
E. Warfarin versus aspirin for stroke prevention in an elderly community population with atrial fibrillation (the Birmingham Atrial Fibrillation Treatment of the Aged Study, BAFTA): a randomised controlled trial. Lancet. 2007;370(9586):493–503.
+
McCarthy
M, Sucharew
H, Kissela
B,
et al. Age, subjective stress, and depression after ischemic stroke. J Behav Med. August 2015.
+
McGeough
E, Pollock
A, Smith
LN, Dennis
M, Sharpe
M, Lewis
S, Mead
GE. Interventions for post-stroke fatigue. Cochrane Database of Systematic Reviews 2009, Issue 3. Art. No.: CD007030. DOI: 10.1002/14651858.CD007030.pub2
+
Mehrholz
J, Werner
C, Kugler
J, Pohl
M. Electromechanical-assisted training for walking after stroke. Cochrane Database of Systematic Reviews 2007, Issue 4. Art. No.: CD006185. DOI: 10.1002/14651858.CD006185.pub2
+
Mohr
JP, Albers
GW, Amarenco
P, Babikian
VL, Biller
J, Brey
RL, Coull
B, Easton
JD, Gomez
CR, Helgason
CM, Kase
CS, Pullicino
PM, Turpie
AGG. Etiology of. Stroke. 1997;28(7):1501–1506.
+
Mohr
J, Gregory
W, Pierre
A, Robin
L, Bruce
C, Easton
J
et al.. Etiology of Stroke. Stroke. 1997; 28:1501–1506.
+
Mozaffarian
D, Benjamin
EJ, Go
AS
et al.. American Heart Association. Executive summary: Heart disease and stroke statistics--2015 update. Circulation. January 27, 2015;131(4):434–441.
+
Naggara
O, Meder
J, Oppenheim
C. MRI is the cornerstone of the actual and future medical management in stroke patients. Diagn Imaging. December 2014;95(12):1127–1128.
+
National Heart, Lung, and Blood Institute. Unpublished Tabulation of the National Health and Nutrition Examination Survey, 1971-1975, 1976-1980, 1988-1994,1999-2004, and 2005-2008 and Extrapolation to the U.S. Population, 2008.
+
Ng
Y, Stein
J, Salles
S, Black-Schaffer
R. Clinical characteristics and rehabilitation outcomes of patients with posterior cerebral artery stroke. Arch Phys Med Rehabil. November 2005;86(11):2138–2143.
+
Olsson
T, Viitanen
M, Asplund
K, Eriksson
S, Hagg
E. Prognosis after stroke in diabetic patients. A controlled prospective study. Diabetologia. 1990; 33(4): 244–249.
+
Parton
A, Malhotra
P, Husain
M. Hemispatial neglect. J Neurol Neurosurg Psychiatry. 2004:75:13–21.
+
Piron
L, Turolla
A, Agostini
M,
et al. Motor learning principles for rehabilitation: a pilot randomized controlled study in poststroke patients. Neurorehabil Neural Repair. 2010;24(6):501–508.
+
Pollock
A, Baer
G, Pomeroy
VM, Langhorne
P. Physiotherapy treatment approaches for the recovery of postural control and lower limb function following stroke. Cochrane Database of Systematic Reviews 2006, Issue 1. Art. No.: CD001920. DOI: 10.1002/14651858.CD001920.pub2
+
Reith
J, Jørgensen
HS, Nakayama
H, Raaschou
HO, Olsen
TS (August 1997)."Seizures in acute stroke: predictors and prognostic significance. The Copenhagen Stroke Study". Stroke 28 (8): 1585–9
+
Rodgers
H, Atkinson
C, Bond
S, Suddes
M, Dobson
R, Curless
R. Randomized controlled trial of a comprehensive stroke education program for patients and caregivers. Stroke Journal. December 1999. Ed 30 Vol 12 pgs. 2585–91.
+
Roger
VL, Go
AS, Lloyd-Jones
DM. Heart Disease and Stroke Statistics—2012 Update A Report From the American Heart Association. Circulation. 2012;125(1):e2–e220.
+
Rosamond
WD, Folsom
AR, Chambless
LE. Stroke Incidence and Survival Among Middle-Aged Adults 9-Year Follow-Up of the Atherosclerosis Risk in Communities (ARIC) Cohort. Stroke. 1999;30(4):736–743.
+
Safer
V, Koseoglu
B. Timing of inpatient rehabilitation initiation in stroke patients: factors influencing early admission. J Phys Ther Sci. June 2015;27(6):1913–1917 5p.
+
Safford
M, Gamboa
C, Muntner
P,
et al. Race-sex differences in the management of hyperlipidemia the reasons for geographic and racial differences in stroke study. Am J Prev Med. 2015;48(5):520–527.
+
Santamato
A, Micello
MF, Panza
F, Fortunato
F, Picelli
A, Smania
N, Logrscino
G, Fiore
P, Ranieri
M. Adhesive taping vs. daily manual muscle stretching and splinting after botulinum toxin type A injection for wrist and fingers spastic overactivity in stroke patients: a randomized controlled trial. Clin Rehabil. 2015;29(1):50–58.
+
Schouten
L, Hulscher
M, Akkermans
R, van Everdingen
J, Grol
R, Huijsman
R. Factors That Influence the Stroke Care Team's Effectiveness in Reducing the Length of Hospital Stay. Stroke. 2008 Sep 1;39(9):2515–21.
+
Senelick
RC, Rossi
PW, Dougherty
K. Living with Stroke: A Guide for Families. Contemporary Books, Chicago. 1994.
+
Smith
M, Stinear
C. Plasticity and motor recovery after stroke: Implications for physiotherapy. New Zealand J Physiotherapy. 2016;44(3): 166–173.
+
States
RA, Pappas
E, Salem
Y. Overground physical therapy gait training for chronic stroke patients with mobility deficits. Cochrane Database of Systematic Reviews 2009, Issue 3. Art. No.: CD006075. DOI:10.1002/14651858.CD006075.pub
+
Stoller
O, de Bruin
ED, Schuster-Amft
C, Schindelholz
M, de Bie
RA, Hunt
KJ. Cardiovascular rehabilitation soon after stroke using feedback-controlled robotics-assisted treadmill exercise: study protocol of a randomised controlled pilot trial. Trials. 2013;14:304.
+
Sandercock
PAG, Gibson
LM, Liu
M. Anticoagulants for preventing recurrence following presumed non-cardioembolic ischaemic stroke or transient ischaemic attack. Cochrane Database of Systematic Reviews 2008, Issue 3. Art. No.: CD000248. DOI: 10.1002/14651858.CD000248.pub2
+
Towfighi
A, Markovic
D, Ovbiagele
B. Consistency of blood pressure control after ischemic stroke: prevalence and prognosis. Stroke. May 2014;45(5):1313–1317.
+
Tyson
S, Burton
L, McGovern
A. The effect of a structured model for stroke rehabilitation multi-disciplinary team meetings on functional recovery and productivity: a Phase I/II proof of concept study. Clin Rehabil. September 2015;29(9):920–925.
+
Villarosa, Linda, Singleton, LaFayette, MD, Johnson, Kirk A. (1993). Black Health Library Guide to Stroke. Henry Holt and Company, New York.
+
Yavuzer
G, Selles
R, Sezer
N, Sutbeyaz
S, Busssmann
J, Koseoglu
F, Atay
M, Stam
H. Mirror Therapy Improves Hand Function in Subacute Stroke: A Randomized Controlled Trial. Arch Phys Med Rehabil. 2008;89(3):393– 398.
+
Yu-Dong
M, Jun
W, Zhi-Hua
D, Xiang
Yu C, Ding-Biao
Z, Bao-Min
L. Mechanical Thrombectomy with Solitare Stent for Acute Internal Carotid Artery Occlusion without Atherosclerotic Stenosis: Dissection of Cardiogenic Thromboembolism. International Medical Journal. August 2016;23(4):373–378.
+++
Cerebrovascular Accident –Arteriovenous Malformation
++
ArterioVenous Malformation (AVM)s are congenital vascular deformities in intracranial vessels, caused by failure of the embryonic capillary bed to develop appropriately, but usually not detected or problematic until adulthood.
++
++
Abnormal expressions of angiogenic and inflammatory proteins, along with the high level of growth factors found in cerebral AVMs, may indicate that AVMs could be the result of a biologically active lesion (Ferrara, 2011). Though AVMs occur throughout the human body, this outline is focused on cerebrovascular accidents (CVAs) caused by bleeding from weakened cerebral AVMs.
++
++
Arteriovenous malformation (AVM) occurs when arterial blood is shunted directly into the venous system. These malformations form a central nidus, or tangled mass of improperly developed vessels, prone to either subarachnoid or intracranial bleeding. The bleeding can be sudden and severe, possibly caused by a weak-walled aneurysm with rupture.
Aneurysms in or due to AVMs can also cause multiple signs and symptoms of CNS distress with an increase in intracranial pressure (ICP). Aneurysms are most commonly found in the artery that feeds the AVM (Altschul, 2002). Bleeding can result in hemorrhagic CVA, or ischemia to the tissues that should have received the lost blood. Cardiac insufficiency may cause latent AVMs to become problematic by gradually decreasing supply to the surrounding tissues.
Surgical removal is enhanced because AVMs are usually encased by a membrane/layer of abnormal tissue, separating the AVM from the surrounding functioning tissue. 90% of AVMs in the brain are located superior to the tentorium cerebelli (Rinaldi, 2015).
Intracranial AVMs present as hemorrhagic CVA in approximately 40-50%, which accounts for between 12% and 24% of all strokes (Dursaut, 2015; American Stroke Association, 2014).
++
++
AVMs are frequently not detected until the patient is 20-50 years old (mean detection 30.2 - 32.2 years of age) (Hofmeister, 2000).
AVMs occur in approximately one out of every 200-500 people, while the annual incidence of discovery of symptomatic AVMs is one per 100,000 (American Stroke Association, 2014; Dursaut, 2015).
+++
II. Diagnostic Procedures
++
++
A common symptom of an existing, unruptured AVM is headache. Approximately 33% of patients with an existing AVM experienced migraine-like headache and 51% of participants present with tension-type headaches. Migraines were associated with AVMs located predominantly in the occipital lobe while tension headaches were most often linked to frontal and temporal AVMs (Lai, 2017).
Often the first symptoms of ruptured AVMs are those of transient ischemic attack (TIA) or CVA. The New York Islands AVM Hemorrhage Study showed that the symptoms of hemorrhage, such as dizziness and paralysis, were the first symptoms in 0.51 per 100,000 persons per year (Ferrara, 2011).
Hemorrhage, headache, and seizure (Dursaut, 2015; Ferrara, 2011) are most often reported (Stapf, 2000). Other symptoms may include paralysis, nausea and vomiting, loss of consciousness, speech, memory, and vision (NINDS, 2001; Altschul, 2002).
++
++
Signs of AVMs are often observed as a change in vision, loss or difficulty with speech, loss of memory, and paralysis. Patients also present with difficulty in movement or focal neurological deficits (Dursaut, 2015).
A small number of cases even present with orbital drainage (Volpe, 2000).
++
++
Prior to surgery, the main diagnostic tool to identify AVM is cerebral angiography. Angiography provides detailed information regarding size, pattern and precise location of the AVM (Altschul, 2002).
MRI is the leading non-invasive tool for diagnosis of AVMs; MRI is very sensitive and provides critical details regarding size and location.
3-D multidetector CT (MDCT) angiography can show active contrast leakage outside of the blood vessels, called a spot sign. Spot signs may predict poor outcomes and/or a high risk of death, but it is difficult to determine the difference between AVM or aneurysm (Ferrara, 2011).
Other tests frequently used during the diagnostic process may include CT scan, digital subtraction angiography (DSA), radiographs, ultrasound, electroencephalography (EEG), and arteriograms (Altschul, 2002).
++
Diagnostic Classification:
++
AVMs are commonly classified using the Spetzler-Martin Grading System (SMGS), a tool used to improve preoperative risk prediction and identifying patients who are appropriate for the surgical procedure (Kim, 2014). Using this system, AVMs are graded based on the 1) size of the AVM, 2) eloquence of adjacent brain, and 3) pattern of venous drainage (Altschul, 2002; Shotar, 2017). Total SMGS grade is calculated based on adding score for size, eloquence and drainage pattern, with a max grade of five for large (3), eloquent (1) and deep (1) (Altschul, 2002).
Size is designated as small (<3 cm), medium (3-6 cm), or large (>6 cm).
Eloquence refers to location in sensorimotor, language, visual, thalamus, hypothalamus, internal capsule, brain stem, cerebellar peduncles, and deep cerebellar nuclei.
Pattern of venous drainage is superficial only if all drainage is via the cortical system.
In addition to SMGS classification, points are also assigned for the ABCs of AVMs: 1) patient Age, 2) Bleeding or hemorrhage presentation, and 3) AVM Compactness. Total scores are analogous with the Spetzler-Martin System.
Patients' ages are assigned 1 point (<20 years old), 2 points (20-40 years old), or 3 points (>40 years old).
Bleeding receives 0 points for a "no" answer and 1 point for a "yes" answer.
Compactness receives 0 points for a "yes" answer and 1 point for a "no" answer (Kim, 2014).
++
A ruptured brain AVM has a high morbidity and mortality rate. Survival outcomes can vary greatly. Intraventricular hemorrhage is the strongest predictor of a poor outcome. Other factors associated with poor outcomes include: blocked deep venous drainage, systemic hypertension, midline shift, high intracranial hemorrhage volume, hydrocephalus, and hyperglycemia (Shotar, 2017).
More than 50% of AVMs will eventually bleed, which may lead to stroke and/or death. Once the condition is diagnosed, severe restrictions are likely to be placed on activities, with the ever-present psychologically debilitating threat of hemorrhagic CVA or neurological problems brought on by increased ICP.
Surgical removal of an AVM can require a significant period (up to three years) of guarded rehabilitation requiring the strict maintenance of normal blood pressure. Successful surgical removal usually permanently corrects the problem, though a new occurrence 17 years after initial treatment has been reported in one case (Akimoto, 2003). Results of surgical correction seem to be very good for AVMs of less than 3 cm, though neurological complications can and do occur in a small percentage of cases (Pik, 2000).
Radiosurgery for AVMs in children achieve complete obliteration in 70% of the children, and subsequent hemorrhaging correlated mainly to high-volume AVMs (Levy, 2000).
High-grade SMGS deficits are more difficult to surgically correct and typically lead to greater risk of neurological complications from surgery (Altschul, 2002). However, individual characteristics including increased age, decreased health, and brain anatomy variations can also contribute to the increased risk of surgical complications (Ferarra, 2011). The results from surgery are reported as excellent or good for 105/144 patients, even though 112 of the patients were Grade 3 or higher (Pollock, 2003).
Patients who have an AVM with single venous drainage anatomy are more likely to have hemorrhage compared to patients with multiple draining veins without stenosis (Sahlein, 2014).
+++
IV. Medical and Surgical Management
++
++
++
++
In many cases, people facing the threat of AVM may perceive the status quo to be preferable to the risks associated with attempting to surgically correct the malformation (Shin, 1997). Each patient must be uniquely evaluated, balancing the risks of intervention against the likelihood of cure, and the likely consequences of long-term conservative management.
When diagnosed with an AVM after neurological problems from CVA or cerebral bleeding, the patient must be closely monitored to ensure normal blood pressure and intra-abdominal pressure until stable enough for surgery. This may include intravenous feeding and administration of stool softeners (Altschul, 2002).
Surgery can frequently be used to completely correct all but Grade 5 lesions, and seems to be the treatment of choice for achieving permanent obliteration of AVMs (Altschul, 2002). A study on children reported some success with Grade 5 malformations (Levy, 2000). Risks vary according to patient condition and the characteristics of the AVM.
Alternative management may involve embolization of the AVM with glue or special particles, coils, spheres, or balloons applied through radiographic-guided intervention (NINDS, 2001; Altschul, 2002). This technique is often used to increase manageability of larger AVMs prior to surgery.
Stereotactic radiosurgery is becoming an increasingly popular tool (NINDS, 2001; Altschul, 2002; Pollock, 2003; Friedman, 2003; Ross, 2000). This technique delivers focused radiation in high doses that causes a toxic reaction to obliterate the AVM (Ferrara, 2011; Lai, 2017). This procedure is minimally invasive. When compared to repeated resection, radiosurgery should be used as a therapeutic alternative for patients with partially resected AVMs (Ding, 2016).
Combinations of embolization with radiosurgery and/or surgery are frequently used to treat larger malformations. Embolization may either be used before surgery to make the malformation more manageable, or afterwards to clean up any residual bleeding (Chang, 2003).
Experimental treatments being explored include: predictive diagnosis, genetics, and molecular characterization, demonstrating considerable interest in funding applied research and advancing medicine in the area of AVM treatment and management (Harrigan, 2003; Friedman, 2003; Tomak, 2003; Shenkar, 2003; Massoud, 2000; Sadato, 2000; Mast, 1995).
+++
V. Therapeutic Management
++
++
Weight training and impact sports that might increase ICP or blood pressure are absolutely contraindicated. Any form of exercise which increases blood pressure is to be avoided.
Blood pressure increase is most frequently brought about by exercises that increase intra-abdominal pressure such as lifting, straining, bending over, prolonged sitting or standing, chronic or forceful cough, pregnancy, ascites, obesity, congestive heart failure, low-fiber diet, constipation, delayed bowel movement, or vigorous exercise, and should be avoided.
Possibly, the best approach is light aerobic exercise such as walking, stretching, and repetitive, unresisted or gravity-resisted motions.
++
++
Increases in blood pressure must be strictly avoided until the AVM is completely resolved. This is due to the blood vessels being excessively hypotensive beyond the point of the AVM. After surgery, the vessels require adequate time to recover and tolerate normal blood pressure (Ferrara, 2011).
Light aerobic exercise for cardiovascular fitness, stretching, and repetitive motion without straining will help maintain flexibility, tone, and some strength.
++
Most important points for the therapist:
++
Known presence of AVM or recent corrective surgery requires strict maintenance of normal blood pressure. Do not prescribe exercise or activities which increase intra-abdominal pressure. Educate the patient to avoid the Valsalva maneuver, which involves attempted exhalation against a closed airway.
Monitor vital signs carefully, before, during, and after therapy.
Remain alert for the signs of TIA or CVA onset listed above.
+
Akimoto
H, Komatsu
K, Kubota
Y. Symptomatic de novo arteriovenous malformation appearing 17 years after the resection of two other arteriovenous malformations in childhood: case report. Neurosurgery. 2003;52:228–31.
+
Berman
MF, Sciacca
RR, Pile-Spellman
J,
et al. The epidemiology of brain arteriovenous malformations. Neurosurgery. 2000;47:389–96.
+
Chang
SD, Marcellus
ML, Marks
MP, Levy
RP, Do
HM, Steinberg
GK. Multimodality treatment of giant intracranial arteriovenous malformations. Neurosurgery. 2003;53:1–11.
+
Ding
D, Xu
Z, Shih
H-H, Starke
RM, Yen
C-P, Sheehan
JP. Stereotactic radiosurgery for partially resected cerebral arteriovenous malformations. World Neurosurg. 2016;85:263–272.
+
Dursaut
TE, Magro
E, Gentric
J-C,
et al. Treatment of brain AVMs (TOBAS): study protocol for a pragmatic randomized controlled trial. Trails. 2015; 16: 1–12 12p.
+
Ferrara
AR. Brain arteriovenous malformations. Radiologic Technology. 2011: 82(6):543MR–556MR.
+
Friedman
WA, Bova
FJ, Bollampally
S, Bradshaw
P. Analysis of factors predictive of success or complications in arteriovenous malformation radiosurgery. Neurosurg. 2003;52:296–307.
+
Harrigan
MR. Angiogenic factors in the central nervous system. Neurosurgery. 2003;53:639–60.
+
Hofmeister
C, Stapf
C, Hartmann
A,
et al. Demographic, morphological, and clinical characteristics of 1289 patients with brain arteriovenous malformations. Stroke. 2000;31:1307–10.
+
Kim
H, Abla
AA, Nelson
J,
et al. Validation of the Supplemented Spetzler-Martin Grading System for brain arteriovenous malformations in a multicenter cohort of 1009 surgical patients. Neurosurgery. 2015:76(1):25–33.
+
Lai
LF, Chen
M, Chen
JX,
et al. Multidisciplinary Care of Unruptured Brain Arteriovenous Malformations to Improve Symptomatic Headache and the Onset, Progression, and Outcomes of Unruptured Brain Arteriovenous Malformations. Pain Physician. 2017;20:E127–E136.
+
Levy
EI, Niranjan
A, Thompson
TP,
et al. Radiosurgery for childhood intracranial arteriovenous malformations. Neurosurg. 2000;47:834–41.
+
Massoud
TF, Hademenos
GJ, De Salles
AA, Solberg
TD. Experimental radiosurgery simulations using a theoretical model of cerebral arteriovenous malformations. Stroke. 2000;31:2466–77.
+
Mast
H, Mohr
JP, Thompson
JLP,
et al. Transcranial Doppler ultrasonography in cerebral arteriovenous malformations: diagnostic sensitivity and association of flow velocity with spontaneous hemorrhage and focal neurological deficit. Stroke. 1995;26:1024–7.
+
Ogilvy
CS, Stieg
PE, Awad
I,
et al. Recommendations for the Management of Intracranial Arteriovenous Malformations. Stroke. 2001;32:1458–1471.
+
Pik
JH, Morgan
MK. Microsurgery for small arteriovenous malformations of the brain: results in 110 consecutive patients. Neurosurgery. 2000;47:571–5.
+
Pollock
BE, Gorman
DA, Coffey
RJ. Patient outcomes after arteriovenous malformation radiosurgical management: results based on a 5- to 15-year follow-up study. Neurosurgery. 2003;52:1291–6.
+
Rinaldi
M, Mezzano
E, Berra
M, Pares
H, Ploceo
R, Papalini
F. Malformaciones arteriovenosas revision yanalisis descriptivo de 52 mavs tratadas durante el period de 2000-2010. Surgl Neurol Int. 2015;6:1–10.
+
Ross
DA, Sandler
HM, Balter
JM, Hayman
JA, Deveikis
J, Auer
DL. Stereotactic radiosurgery of cerebral arteriovenous malformations with a multileaf collimator and a single isocenter. Neurosurgery. 2000;47:123–30.
+
Sadato
A, Wakhloo
AK, Hopkins
LN. Effects of a mixture of a low concentration of n-butylcyanoacrylate and ethiodol on tissue reactions and the permanence of arterial occlusion after embolization. Neurosurgery. 2000;47:1197–203.
+
Sahlein
DH, Mora
P, Becske
T, Huang
P, Jafar
JJ, Connolly
ES, Nelson
PK. Features Predictive of Brain Arteriovenous Malformation Hemorrhage. Stroke. 2014;45:1964–1970.
+
Shenkar
R, Elliott
JP, Diener
K,
et al. Differential gene expression in human cerebrovascular malformations. Neurosurgery. 2003;52:465–77.
+
Shin
AY, Porter
PJ, Wallace
MC, Naglie
G. Quality of life of stroke in younger individuals: utility assessment in patients with arteriovenous malformations. Stroke. 1997;28:2395–9.
+
Shotar
E, Debarre
M, Sourour
NA,
et al. Retrospective study of long-term outcome after brain arteriovenous malformation rupture: the RAP score. J Neurosurg. 2017. DOI:10.3171/2016.9.JNS161431.
+
Stapf
C, Mohr
JP, Sciacca
RR,
et al. Incident hemorrhage risk of brain arteriovenous malformations located in the arterial borderzones. Stroke. 2000;31:2365–8.
+
Tomak
PR, Cloft
HJ, Kaga
A, Cawley
CM, Dion
J, Barrow
DL. Evolution of the management of tentoral dural arteriovenous malformations. Neurosurgery. 2003;52:750–60.
+
Volpe
NJ, Sharma
MC, Galetta
SL,
et al. Orbital drainage from cerebral arteriovenous malformations. Neurosurgery. 2000;46:820–4.
++
Charcot-Marie Tooth encompasses numerous hereditary sensory and motor neuropathies (El-Abassi, 2013).
Incidence and Prevalence
It is the most frequently inherited peripheral neuropathy (NINDS, 2011)
Current prevalence reported at 1/1214 to 1/2500 (El-Albassi, 2014)
Prevalence up to 150,000 Americans (Bush, 2007); 50,000 in the European Union and more than 2.6 million people worldwide (Bouhy, 2013)
Equal gender inheritance (Chetlin, 2004).
Onset most common in 2nd decade; usually mid-childhood or early adulthood.
Caused by 60 different genes with more than 900 mutations in those genes (Bouhy, 2013)
So far, 50 genes have been identified as CMT linked, but 30-50 other genes remain unidentified (El-Albassi, 2014)
Mutation in a single gene can cause multiple phenotypes or forms; mutations in different genes can cause the same phenotype (Bouhy, 2013)
90% of genetically stratified patients will have a mutation in one of four genes: PMP22, GJB1, MPZ, MFN2 (Bird, 2015)
Among those in UK/north European and US, about 90% of the cases of CMT are either Autosomal Dominant (AD)- or X-linked (Reilly, 2009).
Predicted greater number of genes and mutations in the coming years to be discovered (Bouhy, 2013)
Classifications:
Charcot-Marie-Tooth 1 (CMT1)
Genetic duplication of peripheral myelin protein 22, inherited in autosomal dominant pattern resulting in neuropathy with reduced nerve conduction velocity (NCV) (typically 5-30 m/sec) (Brennan, 2015) and onion bulb formations
CMT1 type A constitutes 80% of CMT1 cases (Tazir, 2014) (Newman, 2007; Shy, 2008) and 50% of all CMT cases (Shy, 2008)
CMT1 involves primary progression of demyelination of a peripheral nerve, leading to decreased nerve conduction velocity in the peripheral nerves (Bouhy, 2013). The most prominent symptom in all forms of CMT1 is progressive distal wasting of peroneal and calf muscles resulting in distal lower limb weakness (Gess, 2015).
The six subtypes of CMT1 are clinically indistinguishable and are designated solely on molecular findings (Bird, 2015)
Charcot-Marie-Tooth 2 (CMT2)
Comprises 1/3 of the CMT population (Tazir, 2014)
Genetic mutation, inherited in autosomal dominant or recessive pattern
CMT2 involves degeneration of the axon, which leads to a decreased nerve action potential amplitude (Bouhy, 2013)
Axonal neuropathy with preserved NCV but decreased sensory and motor action potentials (Fabrizi, 2007)
NCV may be recorded in the mildly abnormal range (35-48 m/sec) (Bird, 2015).
CMT2 type C – vocal cord and diaphragm paralysis (Santoro, 2002)
Charcot-Marie-Tooth 3 (CMT3 or Dejerine-Sottas syndrome) (NINDS, 2007)
Genetic mutation of FGD4/frabin (Fabrizi, 2009), inherited in autosomal dominant or recessive pattern with early onset (< age 3) of severe demyelination
CMT3 is synonymous with Dejerene-Sottas neuropathy, considered today to be the most severe form of demyelinating CMT (Pareyson, 2009)
Charcot-Marie-Tooth 4 (CMT4)
Charcot-Marie-Tooth X (CMTX)
Phenotypic variability exists even in the same CMT types, but for the most part age of onset, disease course, rate of progression, and overall severity depends on the CMT form, causative gene, and type of mutation (Pareyson, 2009)
Pathogenesis:
Different genetic mutations cause different symptoms (Reilly, 2011).
Affects all nerve related structures and functions leading to muscular, sensory and balance impairments (NINDS, 2011).
Focal and Segmental Glomerulosclerosis (FSGS), which affects 1 in 400 patients with CMT, causes proteinuria and glomerular scarring which eventually leads to a high rate of progression to end stage renal disease (De Rechter, 2015)
++
Signs and Symptoms:
Symptoms usually present distally and progress proximally (Kenis-Coskun, 2016)
First sign may be pes cavus, followed by peroneal weakness; frequent ankle inversion injuries, inability to walk on heels
Wasting, weakness, and then sensory loss of distal segments of lower and then upper extremities (Pareyson, 2009)
Symmetrical weakness
Motor function loss more pronounced than sensory (Don, 2007).
Decrease in: manual muscle test of the foot, leg, and hands; grip strength; range of motion for active dorsiflexion and eversion; balance; vibration sensation; deep tendon reflexes (Pareyson, 1999)
Sensory deficits
Loss of proprioception and decreased sensation in distal lower extremities (Zeitler, 2006).
Rarely positive sensory symptoms such as paresthesia (Pareyson, 2009)
Deformities including:
Pes cavus, claw toes, inversion of calcaneus, adduction of the forefoot (Refshauge, 2006); hammer toes (Pareyson, 2009) or pes planus that eventually progresses to pes cavus (Pareyson, 2009)
Overall, a shortening of foot length occurs as a midfoot "plantar folding", with an adducto-varus component. Increased calcaneal pitch follows, with forefoot equinus appearing as an Achilles tendon contracture" (Stilwell, 2016).
Decreased muscle tone and atrophy of distal muscles, predominantly in lower extremities, and includes intrinsic muscles of hands and feet (Zeitler, 2006).
Marked reduction of aerobic capacity during exercise testing (Carter, 2006)
Rarely, patients experience respiratory fatigue due to diaphragm weakness and vocal cord palsy (El-Albassi, 2014)
Sleep abnormalities may also be experienced (El-Albassi, 2014)
Pain
May report tingling and burning (Zeitler, 2006) and mild to severe pain in LEs (MedlinePlus, 2011)
Two out of ten patients with CMT have neuropathic pain that is accompanied by burning or tingling in the lower extremities. (Azzedine, 2012)
Pain is common, especially in the lumbar spine, lower limbs, and feet (Pareyson, 2009)
Others:
Reduced or absent deep tendon reflexes (Pareyson, 2009).
"Steppage gait" pattern – hip hiking and hip and knee flexion during swing phase resulting in a shortened stride length (Bean 2001, Don 2007, Shy 2009); foot drop (Vinci, 2002; Don, 2007); if early onset you may see floppy baby syndrome (hypotonia), delayed motor development, and toe walking (Pareyson, 2009)
Tenderness on palpation over the anterior talofibular ligament; corns over metatarsal heads; hand tremor at rest; areflexia; palpable, enlarged peripheral nerves (Berciano, 2003)
Foot callouses on the bottom of feet (Pareyson, 2009)
Neuropathic ulcers can be common in various plantar or digital high-pressure locations, depending on deformity and progression of weakness and muscle imbalances (Stilwell, 2016).
Acrocyanosis; cold feet (Pareyson, 2009)
Hand tremors (Pareyson, 2009)
"Jambes de coq"- distal lower amyotrophy giving the aspect of an inverted champagne bottle
"Dejerine-Sottas Neuropathy"-minority of patients have severe phenotype with onset in infancy and delayed motor milestones (Bird, 2015)
Underdiagnosed Restless Leg Syndrome and Sleep Apnea with patients with CMT1 especially women were affected in combination with EDS and fatigue (Boentert, 2013)
Intolerance to exercise and fatigue are generally reported (Carter, 2006; El Mhandi, 2008)
Functional Limitations
Difficulty manipulating small objects, carrying a shopping bag, opening jars (Svensson, 2006), buttoning shirts, zippers, utensils, or writing
Frequent tripping over carpet or other objects when foot gets caught during swing phase (foot drop); frequent sprains, history of sprains; difficulty walking and running; frequent twisting of the ankle (Pareyson, 2009)
Parents may notice clumsiness (Shy, 2009) and change in child's gait – knee brought high during swing phase in order to clear the drop foot
++
++
Diagnosis is usually first made in childhood (Kenis-Coskun, 2016)
No set clinical criteria to diagnose CMT. Usually based on NCV studies, electromyogram, family history, genetic testing (no genetic test available for CMT2) (Stadler, 2002) and findings of pes cavus and peripheral weakness; blood tests (Bush, 2007) or NCV studies (MedlinePlus, 2011) help differentiate forms.
Pertinent information from as far back as three generations can be beneficial in diagnosing CMT. It is crucial that information include major health issues for each family member, child death and age, and ethnicity. (Patzko, 2012)
Diagnostic Approach (Pareyson, 2009):
Definition of the clinical phenotype
Identification of the inheritance pattern
Electrophysiological examination- subdivided into 2 main groups on the basis of nerve conduction study (Pareyson, 2009):
Demyelinating form (CMT1 if autosomal dominant; CMT4 if autosomal recessive)
Axonal form (CMT 2)
Also rare intermediate forms between CMT1 and CMT2 such as X-linked CMT (CMTX1) and rare dominant-intermediate types (DI); also a third smaller group represented by pure motor forms (dHMN) (Pareyson 2009)
Molecular analysis
Nerve biopsy for select cases
The epidermal nerve fiber density (ENFD) biopsy can be helpful as an initial screening device when attempting to make the provisional diagnosis of CMT. After performing an ENFD exam, nerve conduction velocity (NCV) and EMG tests will be useful (Stilwell, 2016).
The CMT Neuropathy Score (CMTNS) is modified from the Total neuropathy score (TNS) as a single measure to quantify CMT disability (Shy, 2006) and takes into account that weakness is more prominent in patient's with CMT than sensory deficits (Shy 2006).
The CMTNS can distinguish between patients who are severely, moderately, or mildly disabled; and is a validated measure of length-dependent axonal and demyelinating CMT disability. The intraclass correlation coefficient between the total CMTNS score performed by two examiners was 0.98 (p < 0.01). (Shy, 2006).
Other Diagnostic Texts
Chest Roentgenogram to reveal raised hemidiaphragm bilaterally (Srivastava, 2014)
Pulmonary Function Tests to reveal compromised respiratory function and paralysis (Srivastava, 2014)
++
Slowly progressive disorder that is usually not life threatening (NINDS, 2007)
Symptoms begin in the first or second decade of life and then progress (Pareyson, 2009);
Health related quality of life has been shown to significantly decrease due to sedentary lifestyle (El-Albassi, 2014)
Clinical and pathological presentation varies from mild manifestations to severe disease with poor outcome (Piantino, 2007).
Phenotypic variability exists even in the same CMT types, but for the most part age of onset, disease course, rate of progression, and overall severity depends on the CMT form, causative gene, and type of mutation (Pareyson, 2009)
Expected sequelae:
Contractures if muscle weakness is left unmanaged; respiratory problems (Carter, 2008); scoliosis (Horacek, 2007)
CMT2 type C has shorter life expectancy due to ventilatory problems and aspiration (Lacy, 2001). There is also the possibility of developing bilateral abductor vocal cord paralysis – rare, requires surgical intervention (Lacy, 2001).
Dysfunctions in joint mobility and skeletal structure result from not having medical care. There is no advantage in knowing the type of CMT that a clinician must treat. No treatment has been proven to reverse or slow the disease but quality of life can be improved. (Patzko, 2012)
Diabetes was associated with more severe motor and sensory impairment in patients with CMT1A (Sheth, 2008).
Barthel Index assesses degree of physical disability (Padua, 2006) and CMT Neuropathy Score and the Neuropathy Impairment Score measures severity of peripheral neuropathy and rate of disease progression (Shy, 2008).
Gender differences (Colomban, 2013):
Women reported an earlier onset of symptoms and higher disability of their upper limbs.
Women were significantly more affected than men by difficulties in running, jumping, and falls, with no clear explanation
Women reported higher deterioration of their quality of life (QoL).
Women were more affected by carpal tunnel syndrome (CTS), the upper limb disability assessed by ONLS arm score was higher in women (independently of the CTS)
+++
IV. Medical/Surgical Management
++
There is not a cure (NINDS, 2011)
Only symptomatic or palliative treatment currently available (Pareyson, 2006).
Others:
+++
V. Implications for Therapeutic Management
++
+++
VI. Consumer and Professional Resources
++
+
Azzedine
H, Senderek
J, Rivolta
C, Chrast
R. Molecular Genetics of Charcot-Marie-Tooth Disease: From Genes to Genomes. Molecular Syndromology. 2012;3:204–214.
+
Bean
J, Walsh
A, Frontera
W. Brace Modification Improves Aerobic Performance in Charcot-Marie Tooth Disease. Am J Phys Med Rehabil. 2001;80(8):578–582.
+
Berciano
J, Combarros
O. Hereditary neuropathies. Curr Opin Neurol. 2003;16(5):613–622.
+
Bird
TD. Charcot-Marie-Tooth Hereditary Neuropathy Overview. 1998 Sep 28 [Updated 2015 May 7]. In: Pagon
RA, Adam
MP, Ardinger
HH
et al.. GeneReviews® [Internet]. Seattle (WA): University of Washington, Seattle; 1993-2016. Available from:
http://www.ncbi.nlm.nih.gov/books/NBK1358/?report=classic +
Bouhy
D, Timmerman
V. Animal models and therapeutic prospects for Charcot-Marie-Tooth disease. Ann Neurol. 2013;74(3):391–6.
+
Boentert
M, Knop
K, Schuhmacher
C, Burkhard
G, Okegwo
A, Young
P. Sleep disorders in Charcot-Marie-Tooth disease type 1. J Neurol Neurosurg Psychiatry. 2014;85: 319–325.
+
Boffeli
T, Tabatt
J. Minimally Invasive Early Operative Treatment of Progressive Foot and Ankle Deformity Associated with Charcot-Maria-Tooth Disease. J Foot Ankle Surg. 2014;3:1–8.
+
Brennan
K, Bai
Y, Shy
M. Demyelinating CMT- what's known, what's knew and what's in store? Neurosci Lett. 2015;10:1–13.
+
Bush
BC. Case study: management of aggressive periodontitis and Charcot-Marie-Tooth disorder. Access. 2007:31–34.
+
Carter
GT, Han
JJ, Mayadev
A, Weiss
M. Modafil Reduces Fatigue In Charcot-Marie Tooth Disease Type 1A: A Case Series. Am J of Hospice and Palliative Med. 2006;23(5):412–6.
+
Carter
GT, Weiss
MD, Han
JJ, Chance
PF, England
JD. Charcot-Marie-Tooth disease. Curr Treat Options Neurol. 2008;10:94–102.
+
Chetlin
RD, Gutmann
L, Tarnokpolsky
M,
et al. Resistance Training Effectiveness in Patients With Charcot-Marie-Tooth Disease: Recommendations for Exercise Prescription. Arch Phys Med Rehabil. 2004;85:1217–1223.
+
Colomban
C, Micallef
J. Clinical spectrum and gender differences in a large cohort of Charcot-Marie-Tooth type 1A patients. J Neurol Sci. 2014;336:155–160.
+
Del Bianco
J, Fatone
S. Comparison of silicone and posterior leaf spring ankle-foot orthoses in a subject with Charcot-Marie-Tooth disorder. J Prosthetics Orthotics. 2008;20(4):155–162.
+
Don
R, Serrao
M, Vinci
P,
et al. Foot drop and plantar flexion failure determine different gait strategies in Charcot-Marie-Tooth patients. Clin Biomechanics. 2007;22:905–916.
+
Dreher
T, Wolf
SI, Heitzmann
D, Fremd
C, Klotz
MC, Wenz
W. Tibialis posterior tendon transfer corrects the foot drop component of cavovarus foot deformity in Charcot-Marie-Tooth disease. J Bone Joint Surg Am. 2014;96(6):456–62.
+
El Mhandi
L, Millet
GY, Calmels
P,
et al. Benefits of interval-training on fatigue and functional capacities in Charcot-Marie-Tooth disease. Muscle Nerve. 2008;37(5):601–610.
+
El-Abassi
R, England
J, Carter
G. Charcot-Marie-Tooth Disease: An Overview of Genotypes, Phenotypes, and Clinical Management Strategies. PM&R. 2014;6(4):342–355.
+
Fabrizi
GM, Cavallaro
T, Angiari
C,
et al. Charcot-Marie-Tooth disease type 2E, a disorder of the cytoskeleton. Brain. 2007;130:394–403.
+
Fabrizi
GM. Further evidence that mutations in FGD4/frabin cause Charcot-Marie-Tooth disease type 4H. Neurology. 2009;72(13):1160–4.
+
Ferrarin
M, Lencioni
T, Rabuffetti
M, Moroni
I, Pagliano
E, Pareyson
D. Changes of gait pattern in children with Charcot-Marie-Tooth disease type 1A: a 18 months follow-up study. J Neuroeng Rehabil. 2013;10(1):65.
+
Garcia-Ferreira
J, Hernandez-Palazon
J. Response to cisatracurium in patient with Charcot-Marie-Tooth disease. Euro J of Anesthesiology. 2005;22:154–163.
+
Gess
B, Baets
J, De jonghe
P, Reilly
MM, Pareyson
D, Young
P. Ascorbic acid for the treatment of Charcot-Marie-Tooth disease. Cochrane Database Syst Rev. 2015;12:CD011952
+
Guillebastre
B, Calmels
P, Rougier
PR. Assessment of Appropriate Ankle-Foot Orthoses Models for Patients with Charcot-Marie-Tooth Disease. Am J Phys Med Rehabil. 2011;90(8):619–627.
+
Hoff
J, Gilhus
N, Daltveit
A. Pregnancies and deliveries in patients with Charcot-Marie-Tooth disease. Neurology. 2005; 64: 459–462.
+
Horacek
O, Mazanec
R, Morris
CE, Kobesova
A. Spinal deformities in hereditary motor and sensory neuropathy. Spine. 2007; 32(22): 2502–2508.
+
Ibarra-Luzar
JI
et al.. Our experience in the clinical and electrophysiological study of patients with Type I Charcot-Marie-Tooth disease [Spanish]. Rehabilitacion. 2008 Jan-Feb;42(1):13–9.
+
Jani-Acsadi
A, Krajewski
K. Charcot-Marie-Tooth Neuropathies: Diagnosis and Management. Semin Neurol. 2008;28(2):185–194.
+
Kenis-Coskun
OMatthews D. Rehabilitation issues in Charcot-Marie-Tooth disease. J Pediatr Rehabil Med. 2016;9(1):31–34.
+
Lacy
P, Hartley
B, Rutter
M, Cotton
R. Familial Bilateral Vocal Cord Paralysis and Charcot-Marie-Tooth Disease Type II-C. Arch Otolaryngol-Head Neck Surg. 2001; 127: 322–4.
+
Lafarge
C, Talsania
K, Townshend
J, Fox
P. Living with Charcot-Marie-Tooth Disease: a qualitative analysis. Brit J Neurosci Nursing. October 2014;10(5):226–235.
+
Leeuwesteijn
AE, de Visser
E, Louwerens
JW. Flexible cavovarus feet in Charcot-Marie-Tooth disease treated with first ray proximal dorsiflexion osteotomy combined with soft tissue surgery: A short-term to mid-term outcome study. Foot and Ankle Surgery. 2010;16(3):142–147.
+
Menotti
F, Laudani
L, Damiani
A, Mignogna
T, Macaluso
A. An anterior ankle-foot orthosis improves walking economy in Charcot-Marie-Tooth type 1A patients. Prosthet Orthot Int. 2014;38(5):387–92.
+
Meuleman
J, Timmerman
V, Nelis
E, De Jonghe
P. Molecular genetics of inherited peripheral neuropathies: who are the actors? Acta Neurol Belg. 2000;100(3):171–180.
+
Newman
C, Walsh
M, O'Sullivan
et al.. The characteristics of gait in Charcot-Marie-Tooth disease types I and II. Gait Posture. 2007;26;120–27.
+
Nicholson
GA. The dominantly inherited motor and sensory neuropathies: clinical and molecular advances. Muscle Nerve. 2006;33(5):589–597.
+
Olney
B. Treatment of the cavus foot. Deformity in the pediatric patient with Charcot-Marie-Tooth. Foot Ankle Clin. 2000;5(2):305–315.
+
Padua
L, Aprile
I, Caliandro
P, Pazzaglia
C, Commodari
I, Tonali
P. Reliability and validity of the CMT neuropathy score as a measure of disability. Neurology. 2006; 66: 615–6.
+
Pareyson
D, Marchesi,
C. Diagnosis, natural history, and management of Charcot–Marie–Tooth disease. Lancet Neurol. 2009;8(7):654–667.
+
Pareyson
D, Schenone
A, Fabrizi
GM,
et al. A multicenter, randomized, double blind, placebo-controlled trial of long-term ascorbic acid in Charcot-Marie-Tooth disease type 1A. Pharmalogical Research. 2006; 54: 436–441.
+
Pareyson
D. Charcot-marie-tooth disease and related neuropathies: molecular basis for distinction and diagnosis. Muscle Nerve. 1999;22(11):1498–1509.
+
Passage
E, Norreel
J. Ascorbic acid treatment corrects the phenotype of a mouse model of Charcot-Marie-Tooth disease. Nature Medicine. 2004;10(4):396.
+
Patzko
A, MD, Shy
ME, MD. Charcot-Marie-Tooth Disease and Related Genetic Neuropathies. Continuum Lifelong Learning Neurol. 2012;18(1):39–59.
+
Pfeiffer
G. Disability and quality of life in Charcot-Marie-Tooth disease type 1. J Neurol, Neurosurg Psychiat. 2001;70(4):548–550.
+
Piantino
JA, Torres
A. Monoclinic Seizures in a Patient With Charcot-Marie-Tooth disease. Pediatr Neurol. 2007;36:118–120.
+
Piscosquito,
G, Reilly,
M. M., Schenone,
A., Fabrizi,
G. M., Cavallaro,
T., Santoro,
L., Manganelli,
F., Vita,
G., Quattrone,
A., Padua,
L., Gemignani,
F., Visioli,
F., Laurà,
M., Calabrese,
D., Hughes,
R. A. C., Radice,
D., Solari,
A., Pareyson,
D. and the CMT-TRIAAL and CMT-TRAUK Group. Responsiveness of clinical outcome measures in Charcot–Marie–Tooth disease. Eur J Neurol. 2015;22:1556–1563.
+
Ramdharry
GM, Pollard
AJ, Marsden
JF, Reilly
MM. Comparing Gait Performance of People with Charcot-Marie-Tooth Disease Who Do and Do Not Wear AnkleFoot Orthoses. Physiother Res Int. 2012; 17: 191–199.
+
Rechter
S, Waele
L, Leutchenko
E, Mekahli
D. Charcot-Marie-Tooth: Are you testing for proteinuria. Europ J Paediat Neurol. 2015; 19: 1–5.
+
Refshauge
K, Raymond
J, Nicholson
G, Dolder
P. Night splinting does not increase ankle range of motion in people with Charcot-Marie Tooth disease: A randomized, cross-over trial. Austral J of Physiotherapy. 2006; 52; 193–9.
+
Reilly
MM, Murphy
SM, Laurá
M. Charcot-Marie-Tooth disease. J Periph Nerv Syst. 2011;16(1):1–14.
+
Santoro
L, Manganelli
F, Di Maio
L,
et al. Charcot-Marie-Tooth disease type 2C: a distinct genetic entity. Clinical and Molecular characterization of the first European family. Neuromusc Disord. 2002;12:399–404.
+
Sahenk
Z, Galloway
G, Clark
K
et al.. AAV1.NT-3 Gene Therapy for Charcot–Marie–Tooth Neuropathy. Mol Ther. 2014;22(3):511–521.
+
Sheth
S
et al.. Diabetes mellitus exacerbates motor and sensory impairment in CMT1A. J Peripher Nerv Syst. 2008 Dec; 13(4): 299–304.
+
Shy
ME, Blake
J, Krajewski
K, Fuerst
DR, Laura
M, Hahn
AF, Li
J, Lewis
RA, Reilly
M. Reliability and validity of the CMT neuropathy score as a measure of disability. Neurology. 2005;64(7):1209–14.
+
Shy
ME, Chin
L, Swan
ER,
et al. Neuropathy progression in Charcot-Marie-Tooth disease type 1A. Neurology. 2008; 70: 378–383.
+
Shy
ME. Overview of Charcot-Marie-Tooth disease. "Virtual Grand Rounds": Athena Diagnostic. Available at
www.brainshark.com/athena/CMT. Accessed on February 1, 2009.
+
Siskind
CE, Panchal
S, Smith
CO, Feely
SM, Dalton
JC, Schindler
AB, Krajewski
KM. A review of genetic counseling for Charcot Marie Tooth disease (CMT). J Genet Couns. 2013;22(4):422–36.
+
Srivastava
V, Pasha
T, Knowles
A, Boothman
B, Parahca
M, Kalkat
M, Zacharias
J. Dramatic Improvement After Bilateral Diaphragmatic Plication in Charcot-Marie-Tooth disease. Ann Thorac Surg. 2014; 97: 2177–9.
+
Stadler
T, Ross
D. Charcot-Marie-Tooth Disease in a high school tennis player. The Physician and Sports Med. 2002; 30: 47–51.
+
Stilwell
D, Stilwell
S. Podiatrists' Role in Charcot-Marie-Tooth Disease. Podiatry Management. January 2016:119–123.
+
Svensson
E, Hager-Ross
C. Hand function in Charcot-Marie-Tooth: test- retest reliability of some measurements. Clin Rehabil. 2006; 20: 896–908.
+
Szigeti
K, Lupski
JR. Charcot-Marie-Tooth disease. European Journal of Human Genetics. 2009;17(6):703–710.
+
Tazir
M, Hamadouche
T, Nouioua
S, Mathis
S, Vallet
J. Hereditary motor and sensory neuropathies of Charcot-Maria-Tooth diseases: An update. J Neurol Sci. 2014; 347: 14–22.
+
van Pomeren
M
et al.. The hypothesis of overwork weakness in Charcot-Marie-Tooth: a critical evaluation. J Rehabil Med 2009 Jan; 41(1): 32–4.
+
Vinci
P, Perelli
SL. Foot Drop, Foot Rotation, and Plantar Flexor Failure in Charcot-Marie Tooth Disease. Arch Phys Med Rehabil. 2002; 83: 513–6.
+
Ward
CM, Dolan
LA, Bennett
DL, Morcuende
JA, Cooper
RA. Long-term results of reconstruction for treatment of a flexible cavovar us foot in Charcot-Marie-Tooth disease. J Bone Joint Surg, 2008; 90(12): 2631–42.
+
Young
P, DeJonghe
P, Stogbauer
F, Butterfass-Bahloul
T. Treatment for Charcot-Marie-Tooth disease. Cochrane Database Syst Rev, 2008. doi: 10.1002/14651858.CD006052.
+
Zeitler
H, Ulrich-Merzenich
G, Hoffmann
L,
et al. Long-term effects of a multimodal approach including immunoadsorption for the treatment of myasthenic crisis. Artif Organs. 2006;30(8):597–605.
+++
Complex Regional Pain Syndrome (Reflex Sympathetic Dystrophy)
++
Syndrome characterized by continuing (spontaneous and/or evoked) regional pain (i.e. not in a specific nerve territory or dermatome) that is seemingly disproportionate in time or degree to the usual course of any known trauma or other lesion (Harden, 2013)
Etiology of CRPS:
Type I- (Formerly Reflex Sympathetic Dystrophy - RSD)
Occurs post injury/illness without direct damage to the nerve in the affected extremity , emotional stress can play a role
Pain descriptors usually include allodynia and hyperalgesia (Casale, 2015)
Type II- (formerly causalgia)
Involves specific nerve injury
No evidence that they differ in terms of pathophysiology or treatment responsiveness (Bruehl, 2010)
Most common initiating events: surgery, fractures, crush injuries, and sprains (Harden, 1999)
CRPS NOS (Not Otherwise Specified)
Knowledge of the incidence and prevalence of CRPS is incomplete (RSDSA, 2012)
Pathogenesis
++
Characterized by disproportionate pain and variable combinations of the following changes (Kuttikat, 2016):
Sensory: allodynia, hyperalgesia
Vasomotor: temperature and skin color changes or asymmetry
Sudomotor: sweating changes or asymmetry
Edema
Trophic: thin glossy skin, abnormal hair growth, coarse nails
Motor: weakness, decreased range of motion, tremor, dystonia
Early diagnosis is difficult because the initial pain experienced with CRPS may be believed to be associated with the traumatic event (Glajchen, 1998)
Distribution:
Commonly occurs in the body part affected by the traumatic event or in the side affected by cerebral pathology
Upper limbs are observed to be affected more with a ratio of 3:2 compared to lower limbs (Goh, 2017).
Other Distributions
Continuity - symptoms may start in hand then spread to entire quadrant
Mirror image - may spread from one extremity to opposite extremity
Independent - may jump to a distant part of body
There is not a diagnostic "gold standard" due to unknown pathophysiology of this syndrome (Harden, 2013). The absence of a "gold standard" diagnosis test makes the validation of diagnostic criteria difficult (Borchers, 2014)
Diagnosis is made by excluding other diagnoses and finding no other cause for signs and symptoms (Palmer, 2015)
Diagnostic criteria of CRPS I and II depend on history and physical examination without confirmation by specific test or gold standard (Su, 2009). There is minimal clinical distinction between type I & II, with similar signs/symptoms (Bruehl, 2010).
Regional pain and sensory findings may be accompanied by abnormalities including edema, changes in skin color, temperature, and sudomotor activity (Pappagallo, 2001).
The quality of pain may change during the course of the syndrome, but intensity tends to stay the same. (Casale, 2015)
Diagnostic Criteria:
Other Diagnostic Criteria
Bruehl criteria: pain/sensation, vasomotor, sudomotor/edema, motor/trophic - from the International Association for the Study of Pain (IASP)
Noxious event/cause of immobilization
Continuing pain disproportionate to known occurrence
Edema, changes in skin blood flow or abnormal sudomotor activity (signs OR symptoms)
Exclude present condition(s) accounting for pain/dysfunction. (Aronoff, 2002)
High sensitivity (0.87) and high specificity (0.94) (McBride, 2008)
Low sensitivity (0.70) and high specificity (0.94) (Aronoff, 2002; Niehof, 2006)
Miscellaneous Diagnostic Testing
Psychological factors (Glajchen, 1998)
Radiograph, bone scan, quantitative sensory testing, temperature differences during sympathetic stimulation, MRI, magnetoencephalogram or functional MRI (Baron, 2004)
Measurement of pro-inflammatory cytokines in blister fluid (Groeneweg, 2006)
Screening for antinuclear antibodies in patients with CRPS symptoms shows high prevalence when compared to healthy patient (Dirckx, 2015). To help in the diagnosis of CRPS II, electrodiagnostic testing can evaluate the peripheral nervous system (Freedman, 2014)
++
Varies from person to person (NINDS, 2013)
Very little reliable information on the overall outcome of CRPS due to the use of varied diagnosis criteria and non-standardized outcome measures (Borchers, 2014)
More research is needed to understand the causes of CRPS, how it progresses, and the role of early treatment (NINDS, 2013)
Almost all children and teenagers have good recovery (NINDS, 2013)
Poorer prognosis in smokers compared to non-smokers (Goh, 2017).
Symptoms can resolve within weeks or months spontaneously in some patients, while persistent pain and allodynia can be experienced by other patients (Palmer, 2015)
Anecdotal evidence suggests early treatment, particularly rehabilitation, is helpful in limiting the disorder, but this benefit has not yet been proven in clinical studies (NINDS, 2013)
Progression
Show variable progression over time (Harden, 2013)
Non-progressive; often chronic; rapid onset of pain
Early – usually pain and swelling
Late – often "movement disorder, trophic changes, and autoimmune abnormalities" (Glajchen 1998)
Expected sequelae – depends on severity and location, limitations vary.
The severity of CRPS can be evaluated on the presence or absence of 17 signs and symptoms that have been clinically assessed by utilizing the CRPS Severity Score (CSS). (Freedman, 2014)
In CRPS- Type I, warm skin temperature at onset is classified as having a better prognosis than cold skin temperature. (Gatti, 2016)
Outcomes measures are concentrated on impairments and measure of disability. (Smith, 2009)
++
+++
V. Implications for therapeutic management
++
Guide to Physical Therapist Practice preferred practice pattern (APTA, 2017)
4D: Impaired joint mobility, motor function, muscle performance, and range of motion associated with connective tissue dysfunction
Others: 4A, 5G, 7B (APTA, 2017)
The array of possible patient presentations and the fact that the presentation often changes over time complicates successful identification and treatment (Bruehl, 2002)
The only treatment methodology that can possibly successfully span these gaps in medical science is a systematic and orderly interdisciplinary approach (Harden, 2013)
Role of Physical Therapy
Physical therapy is the cornerstone and first line treatment for CRPS (Rho, 2002)
Treatment of kinesiophobia (Goh, 2017)
Use of modalities catered to patient-specific symptoms (Goh, 2017)
Elevation, massage, contrast baths, transcutaneous electrical nerve stimulation, gentle range of motion, isometric strengthening exercise, and stress loading can all be used to reduce presenting symptoms (Goh, 2017)
Establish plan focused on maintenance and restoration of function by decreasing pain, controlling edema, and increasing ROM, strength, and endurance
Include proprioceptive exercises
Avoid overtreatment
Casting and splinting not typically recommended but may be appropriate with deformities of resting position (Thomas, 2005)
Attention needs to be paid throughout the entire course of treatment to maintain as normal a posture and movement pattern as possible and to prevent changes to adjacent joints and muscles, e.g. changes brought about by contraction (Beems, 2006)
Educate patient and refer to other health care members as needed (Smith, 2009)
Education should support self-management of CRPS and can involve fear-avoidance models by gradually increasing exposure to situations the patient identifies as dangerous and threatening under therapist supervision (Ganty, 2014; Freedman, 2014)
Hydrotherapy can be effective for lower extremity CRPS (Stanton-Hicks, 1998)
A whirlpool bath treatment can be used to decrease edema and pain in CRPS (Devrimsel, 2015)
Neuromuscular electrical stimulation can also improve edema and pain, however improvements with the whirlpool bath treatment exceed NMES (Devrimsel, 2015)
Virtually all patients with advanced CRPS will present with myofascial pain syndrome of the associated joint. Aggressive treatment of this myofascial pain is a critical component of successful treatment and is principally the responsibility of the PT (Harden, 2013)
Neurological therapy: Graded Motor Imagery GMI may be effective and is in essence a sequential set of brain exercises, comprising laterality training, imagined hand movements, and mirror feedback therapy (Moseley, 2006)
Occupational therapy focuses functionality for activities of daily living (Goh, 2017).
Specialized garments or wrappings may reduce edema and sensory overload (Goh, 2017).
Mirror box therapy is used to attempt reductions of neuropathic pain and improve two-point sensation in affected limb (Goh, 2017).
+++
VI. Consumer and professional resources
++
Reflex Sympathetic Dystrophy Syndrome Associate
National Institute of Neurological Disorders and Stroke
International Research Foundation for RSD/CRPS
American RSDHope
+
Aronoff
G, Harden
N, Stanton-Hicks
M, Dorto
A, Ensalada
L, Klimek
E
et al.. American Academy of Disability Evaluating Physicians (AADEP) position paper: complex regional pain syndrome I (RSD): impairment and disability issues. Pain Med. 2002;3:274–288.
+
Bandyk
D, Johnson
B, Kirkpatrick
A, Novotney
M, Back
M, Schmacht
D. Surgical sympathectomy for reflex sympathetic dystrophy syndromes. J Vasc Surg. 2002;35:269–77.
+
Baron
R, Schattschneider
J, Binder
A, Siebrecht
D, Wasner
G. Relation between sympathetic vasoconstrictor activity and pain and hyperalgesia in complex regional pain syndromes: a case-control study. Lancet. 2002;359:1655–1660.
+
Baron
R, Jänig
W. Complex regional pain syndromes—how do we escape the diagnostic trap? Lancet. 2004;364(9447):1739–1741.
+
Beydoun
A. Neuropathic pain: from mechanisms to treatment strategies. J Pain Symptom Manage. 2003;25(5 Suppl):S1–3.
+
Borchers
AT, Gershwin
ME. Complex regional pain syndrome: A comprehensive and critical review. Autoimmunity Reviews. 2014;13(3):242–265.
+
Bruehl
S, Harden
RN, Galer
BS, Saltz
S, Backonja
M, Stanton-Hicks
M. Complex regional pain syndrome: are there distinct subtypes and sequential stages of the syndrome? Pain. 2002;95(1-2):119–124.
+
Bruehl
S. An update on the pathophysiology of complex regional pain syndrome. Anesthesiology. 2010;113(3):713–725.
+
Bussa
M, Guttilla
D, Lucia
M, Mascaro
A, Rinaldi
S. Complex regional pain syndrome type I: a comprehensive review. Acta Anaesthesiol Scand. 2015;59(6):685–97.
+
Casale
R, Atzeni
F, Masala
IF, Sarzi-Puttini
P. The words of pain in complex regional pain syndrome. Best Pract Res Clin Rheumatol. 2015;29(1):71–76.
+
Choi
YS, Lee
MG, Lee
HM, Lee
JL, Jo
JY, Jeon
SY, Lee
SC, Kim
YC. Epidemiology of complex regional pain syndrome: a retrospective chart review of 150 Korean patients. J Korean Med Sci. 2008;23(5):772–775.
+
Correll
GE, Maleki
J, Gracely
EJ, Muir
JJ, Harbut
RE. Subanesthetic Ketamine Infusion Therapy: A Retrospective Analysis of a Novel Therapeutic Approach to Complex Regional Pain Syndrome. Pain Medicine. 2004;5(3):263–275.
+
de Rooij
AM, Mos
M, Sturkenboom
MC, Marinus
J, Maagdenberg
AM, Hilten
JJ. Familial occurrence of complex regional pain syndrome. Eur J Pain. 2009 Feb 1;13(2):171–7.
+
Devrimsel
G, Turkyilmaz
AK, Yildirim
M, Beyazal
MS. The effects of whirlpool bath and neuromuscular electrical stimulation on complex regional pain syndrome. J Phys Ther Sci. 2015;27(1):27–30.
+
Dirckx
M, Schreurs
MWJ, de Mos
M, Stronks
DL, Huygen
FJPM. The prevalence of autoantibodies in complex regional pain syndrome type I. Mediators Inflamm. 2015;2015:718201. doi:10.1155/2015/718201
+
Drucker
W, Hubay
C, Holden
W, Bukovnic
J. Pathogenesis of post-traumatic sympathetic dystrophy. Am J Surg. 1959;97(4):454–465.
+
Eberle
T
et al. Warm and cold complex regional pain syndromes: differences beyond skin temperature? Neurology. 2009;72(6):505–12
+
Freedman
M, Greis
AC, Marino
L, Sinha
AN, Henstenburg
J. Complex regional pain syndrome: diagnosis and treatment. Phys Med Rehabil Clin N Am. 2014;25(2):291–303.
+
Ganty
P, Chawla
R. Complex regional pain syndrome: recent updates. Contin Educ Anaesth Crit Care Pain. 2014;14(2):79–84.
+
Gaspar
M, Bovaira
M, Carrera-Hueso
FJ,
et al. [Efficacy of a topical treatment protocol with dimethyl sulfoxide 50% in type 1 complex regional pain syndrome]. Farm Hosp. 2012;36(5):385–391.
+
Glajchen
M. Reflex sympathetic dystrophy syndrome. APS Bulletin. 1998:8:6. Available from: American Pain Society. Accessed February 14, 2007.
+
Gatti
D, Rossini
M, Adami
S. Management of patients with complex regional pain syndrome type I. Osteoporos Int. 2016. doi: 10.1007/s00198-016-3531-9.
+
Goebel
A, Barker
CH, Turner-Stokes
L, Cameron
H, Lewis
J, Shenker
N. Complex regional pain syndrome in adults: UK guidelines for diagnosis, referral and management in primary and secondary care. London: RCP, 2012.
+
Goh
EL, Chidambaram
S, Ma
D. Complex regional pain syndrome: a recent update. Burns Trauma. 2017;5. doi:10.1186/s41038-016-0066-4.
+
Groeneweg
JG, Huygen
FJPM, Heijmans-Antonissen
C, Niehof
S, Zijlstra
FJ. Increased endothelin-1 and diminished nitric oxide levels in blister fluids of patients with intermediate cold type complex regional pain syndrome type 1. BMC Musculoskelet Disord. 2006;7:91. doi:10.1186/1471-2474-7-91.
+
Haddox
JD, Van Alstine
D. Pharmacologic therapy for reflex sympathetic dystrophy. Arch Phys Med Rehabil. 1996;10:297–307.
+
Harden
RN, Bruehl
S, Galer
BS, Saltz
S, Bertram
M, Backonja
M, Gayles
R, Rudin
N, Bhugra
MK, Stanton-Hicks
M. Complex regional pain syndrome: are the IASP diagnostic criteria valid and sufficiently comprehensive? Pain. 1999;83(2):211–219.
+
Harden
RN, Bruehl
S, Stanton-Hicks
M, Wilson
PR. Proposed New Diagnostic Criteria for Complex Regional Pain Syndrome. Pain Medicine. 2007;8(4):326–331.
+
Harden
RN, Bruehl
S, Perez
RSGM, Birklein
F, Marinus
J, Maihofner
C, Lubenow
T, Buvanendran
A, Mackey
S, Graciosa
J, Mogilevski
M, Ramsden
C, Chont
M, Vatine
JJ. Validation of proposed diagnostic criteria (the "Budapest Criteria") for Complex Regional Pain Syndrome. Pain. 2010;150(2):268–274.
+
Harden
RN. Diagnosis of Complex Regional Pain Syndrome: Signs, Symptoms, and New Empirically Derived Diagnostic Criteria. Clin J Pain. 2010;22:415–419.
+
Harden
RN, Oaklander
AL, Burton
AW,
et al. Complex regional pain syndrome: practical diagnostic and treatment guidelines, 4th edition. Pain Med. 2013;14(2):180–229.
+
International Research Foundation for RSD/CRPS. International Research Foundation for RSD/CRPS. Available at:
http://www.rsdfoundation.org/. Accessed January 18, 2014.
+
Kastler
A, Aubry
S, Sailley
N,
et al. CT-guided stellate ganglion blockade vs. radiofrequency neurolysis in the management of refractory type I complex regional pain syndrome of the upper limb. Eur Radiol. 2012. doi:10.1007/s00330-012-2704-y.
+
Kingery
WS. A critical review of controlled clinical trials for peripheral neuropathic pain and complex regional pain syndromes. Pain. 1997;73(2):123–139.
+
Kuttikat
A, Noreika
V, Shenker
N, Chennu
S, Bekinschtein
T, Brown
CA. Neurocognitive and Neuroplastic Mechanisms of Novel Clinical Signs in CRPS. Front Hum Neurosci. 2016;10. doi:10.3389/fnhum.2016.00016.
+
Lee
SK, Yang
DS, Lee
JW, Choy
WS. Four treatment strategies for complex regional pain syndrome type 1. Orthopedics. 2012;35(6):e834–842.
+
Lewis
J. What is complex regional pain syndrome (CPRS)? Brit J Hand Ther. 2006;11(3):92.
+
Mackey
S, Feinberg
S. Pharmacologic therapies for complex regional pain syndrome. Curr Pain Headache Rep. 2007;11:8–43.
+
Mailis-Gagnon
A, Lakha
SF, Allen
MD, Deshpande
A, Harden
RN. Characteristics of Complex Regional Pain Syndrome in Patients Referred to a Tertiary Pain Clinic by Community Physicians, Assessed by the Budapest Clinical Diagnostic Criteria. Pain Med. 2014;15(11):1965–1974.
+
McBride
ART, Barnett
AJ, Livingstone
JA, Atkins
RM. Complex regional Pain Syndrome (Type 1): A comparison of 2 diagnostic criteria methods. Clin J Pain. 2008;24(7):637–640.
+
McCabe
C, Haigh
R, Ring
P, Halligan
P, Wall
P, Blake,
R. A controlled pilot study of the utility of mirror visual feedback in the treatment of complex regional pain syndrome (type 1). Rheumatology. 2003; 42:97–101.
+
Moseley
GL. Graded motor imagery for pathologic pain: a randomized controlled trial. Neurology. 2006;67(12):2129–2134.
+
Niehof
S, Huygen
F, van der Weerd
R, Westra,
M, Zijlstra
F. Thermography imaging during static and controlled thermoregulation in complex regional pain syndrome type 1: diagnostic value and involvement of the central sympathetic system. BioMedical Engineering Online. 2006:5.
+
Palmer
G. Complex regional pain syndrome. Aust Prescr. 2015;38(3):82–6.
+
Pappagallo
M, Rosenberg
AD. Epidemiology, pathophysiology, and management of complex regional pain syndrome. Pain Pract. 2001;1(1):11–20.
+
Pons
T, Shipton
EA, Williman
J, Mulder
RT. Potential risk factors for the onset of complex regional pain syndrome type 1: a systematic literature review. Anesthesiol Res Pract. 2015;2015:956539. doi:10.1155/2015/956539.
+
Reuben,
S,
et al. Surgery on the affected upper extremity of patients with a history of complex regional pain syndrome: A retrospective study of 100 patients. J Hand Surg. 2000;25:1147–1151.
+
Rho
RH, Brewer
RP, Lamer
TJ, Wilson
PR. Complex regional pain syndrome. Mayo Clin Proc. 2002;77(2):174–180.
+
Sinis
N, Birbaumer
N, Gustin
S
et al.. Memantine treatment of complex regional pain syndrome: a preliminary report of six cases. Clin J Pain. 2007;23:237–243.
+
Smith,
M. The Peripheral System. In: Pathology Implications for the Physical Therapist. 3rd edition. St Louis, MO. Saunders Elsevier. 2009, 1634–1636.
+
Stanton-Hicks
M, Baron
R, Boas
R,
et al. Complex Regional Pain Syndromes: guidelines for therapy. Clin J Pain. 1998;14(2):155–166.
+
Su
ES. Practical management of complex regional pain syndrome. Am J Ther. 2009;16(2):147–54.
+
Thomas
RJ, Degnan
GG. Early diagnosis and treatment are critical to achieving a good outcome—Managing complex regional pain syndrome. J Musculoskel Med. 2005;22(10):514–26.
+
Van de Meent
H, Oerlemans
M, Bruggeman
A,
et al. Safety of "pain exposure" physical therapy in patients with complex regional pain syndrome type 1. Pain. 2011;152(6):1431–1438.
+++
Congenital Limb Deficiency
++
Definition: the congenital absence or hypoplasia of a long bone and/or digits (Gold, 2011)
Childhood amputations in the US: 60% congenital, 40% acquired (Dillingham, 2002)
Demographics: primarily male children (1.15:1) over female with an average age of 6.2 years; the upper extremity (UE) more involved than the lower extremity (LE) (Al-Worikat, 2008).
Genetic component: autosomal recessive disorder (Chen, 2007)
Limb deficiency occurs primarily in the 1st trimester; mesodermal formation of the limb occurs at day 26 of gestation and continues until week 8 (Rossi, 2004). Part or all of a limb fails to form between weeks 4 and 8 of gestation; most cases: cause unknown – most result from sporadic genetic mutation (Ephraim, 2003)
Many causes have been identified including: Fanconi anemia, trisomy 18, exposure in the womb to medications like Thalidomide and Misoprostal. (Gold, 2011)
Teratogenic factors include: Thalidomide, Warfarin, contraceptives, and irradiation (Holmes, 2002)
Hypoxia in fetuses during the middle of the 1st trimester
Maternal anemia, anticonvulsant Phenytoin, and Misoprostol (Webster, 2007)
Smoking during pregnancy is associated with terminal transverse deficiencies (Czeizel, 1994, Man, 2006)
Sequence of events that lead to deformities: hypoxia, endothelial cell damage, hemorrhage, tissue loss, and reperfusion (McQuirk, 2001).
Amniotic band syndrome, split hand/foot syndrome, Nager's acrofacial dystosis, trisomy 18, Brachman-de Lange syndrome, acardiac twin (Gold, 2011)
Children born to mothers with a limb deficiency have a relative risk of 5.6 of having the same defect (Skjaerven, 1999)
++
++
More common in the upper limbs (58%) leading to unilateral, transverse below-elbow limb deficiency; in the lower limbs (18%), proximal femoral focal deficiency (PFFD) is most common leading to leg length discrepancy, hip & knee flexion contractures, & knee instability; and both UE and LE deficiencies (12%) (Makhoul, 2003)
Dependent on the mechanism and severity of deficiency
Damage in utero by thalidomide = symmetrical polydactyly on pre-axial side of both arms and legs (McQuirk, 2001).
Exposed to misoprostol early in pregnancy = asymmetrical digit loss, constriction rings, and syndactyly (McQuirk, 2001).
There is inconsistent use of terminology and classification of congenital limb deficiencies and a common language for diagnostic purposes is needed. (Lowry, 2016)
Congenital limb deficiencies are classified as either transverse or longitudinal (Al-Worikat, 2008).
Transverse deficiency = no distal portion of the limb; named after the segment beyond with which these is no skeletal formation.
Longitudinal deficiency = distal portions remain; named by the bones that are affected (Rossi, 2004).
Classification: ISO/ISPO system - 7 categories based on embryologic failure
Pediatric Orthopaedic Society of North America lists seven categories of congenital deficiencies: "failure of formation, failure of differentiation, duplication, overgrowth, undergrowth, constriction bands, and generalized skeletal abnormalities" (POSNA 2012)
Types: Upper limb or Lower limb
Common Congenital UE deficiencies: (Rossi, 2004)
Transradial deficiency = most common limb deficiency is left terminal transradial deficiency.
Both transradial and trans humeral deficiencies usually require a prosthetic that should meet the child's individual needs at various stages of development.
Transhumeral prosthetics can be large and cumbersome to infants, inhibiting mobility (i.e. rolling to the side), however each child should be evaluated individually for optimal introduction of prosthetics.
Common Congenital LE deficiencies: (Birch, 2011)
Fibular longitudinal deficiency (fibular hemimelia) is the most common congenital lower limb deficiency.
Transtibial deficiency = more common than transfemoral
Transfemoral deficiency = PFFD is the absence of development of the proximal femur, including shortening of the entire femur. Femur held in flexion, abduction, and ER.
++
++
Prenata l genetic tests, metabolic tests, and ultrasounds identify deficiencies (Holder Espinasse, 2004; Makhoul, 2003)
Prenatal karyotyping (chorionic villus sampling and amniocentesis) before 14 weeks gestation (Cederholm, 2005)
Radiographs to confirm bony/joint involvement
MRI to evaluate tissue and ligament integrity
++
Non-progressive, however, musculoskeletal complications can arise as the child grows.
Depending on the severity of deficiency, amputation of the affected limb may be necessary
Motor development may be delayed/impaired while ROM & strength often maintained through developmental activities
Can experience phantom limb pain following revision amputation
Level of deficiency and timing of initial prescription predicts prosthesis rejection/acceptance: the longer the residual limb, the more likely to reject prosthesis; more likely to accept prosthesis if initially fitted before the age of 2 (Scotland, 1983)
Upper extremity prosthesis are found to be worn only during specific activities, not during daily activities (Buffart, 2007)
Children/adolescents with limb deficiencies show greater behavioral and emotional problems and lower social competence than those without limb deficiencies (Varni, 1992)
+++
IV. Medical/Surgical Management
++
++
Medical and pharmaceutical management will be dependent on the individual child's needs based on the type and extent of deficiency.
Post-operative pain management
Pain management for phantom limb sensations
++
++
General principles of childhood amputation surgery (Krajbich, 1998): Preserve limb length; preserve growth plates; choose disarticulation rather than transosseous amputation whenever possible; preserve knee joint whenever possible; stabilize and normalize the proximal portion of the limb.
Upper extremity amputation: Rarely necessary
Limb-lengthening procedures: Ilizarov technique (Orhun, 2003)
Knee arthrodesis and foot amputation: Syme or Boyd amputation
Rotationplasty: Typically for PFFD; requires functioning hip and ankle joint
Excision of distal femur and proximal tibia, 180° rotation of residual limb, and reattachment to proximal femur (Sakkers, 2016)
Nonvascular phalangeal transfer (Unglaub, 2006)
Osseointegration (Jonsson, 2011)
Epiphysiodesis – epiphyseal plate is removed, or a portion is reinserted and rotated 90o (Birch, 2011).
+++
V. Key Aspects of therapeutic management
++
Rehab team: patient, child, caregivers, rehabilitation physician, orthopedist, prosthetist, physical therapist, occupational therapist and social worker
Role of PT: Parent/child education, post-op rehabilitation programs, mobility and self-care training, input regarding prosthetic options (Jain, 1996) encourage individuality and self-management despite use of prosthesis/ability status (Murray, 2008)
General therapy goals:
Optimize participation at various life stages and maintain quality of life (Michielsen, 2010).
Prevent joint contractures and minimize muscle strength imbalances • Minimize muscle strength imbalances
Reduce pain and maintain skin integrity
Attainment of mobility and transitional skills
Independent ambulation
Prosthetic/orthotic education: patient/caregiver
Donning/doffing, skin inspection, prosthesis inspection, and wearing schedule (Krebs, 1991, Nelson, 2006)
Develop independence with mobility and self-care
Prevent skin breakdown/scar maintenance (Watson, 2000)
Psychosocial functioning is generally comparable to healthy peers, although it has been described as at risk (Michielson, 2010).
Prosthetic management: Initial fitting timing, adjustments to growth, and rehabilitation (Krebs, 1991, Nelson, 2006)
Prosthetics are indicated if they will improve movement or function.
Infancy/Toddler: monitor developmental progress. Assess range of motion, strength, anthropometric measures at 1 mo. intervals; consider fitting LE prosthetic (8 -10 mos. old) and UE prosthetic (3-6 mos. old) if indicated for the individual child's functional needs.
Preschool/School: emphasize independence in self-care skills, mobility, acquisition of skills necessary to navigate the educational environment and participate with peers.
Adolescence/Adulthood: Aid in transition to adulthood (college, driving, career) by assessing prosthetic and functional needs as they change.
Improvements in function with the prosthesis can depend on the daily prosthesis usage time and the child's individual daily activity experiences with the prosthesis. (Korkmaz, 2012)
Outcome measures
Functional assessment: Assisting Hand Assessment (AHA) and Unilateral Below Elbow Test (UBET) (Buffart, 2006)
Outcome tool: Child Amputee Prosthetics Project-Functional Status Inventory (CAPP- FSI) (Pruitt, 1996)
Prosthetic Upper Extremity Functional Index (PUFI) (Pruitt, 1997)
Functional Mobility Assessment (FMA) (Pierce, 2011)
+
Al-Worikat
AF, Dameh
W. Children with limb deficiencies: demographic characteristics. Prosthet Orthot Int. 2008;32(1):32–28.
+
Birch
JG, Lincoln
TL, Mack
PW, Birch
CM. Congenital fibular deficiency: a review of thirty years' experience at one institution and a proposed classification system based on clinical deformity. J Bone Joint Surg Am. 2011;93:1144–1151.
+
Buffart
L, Roebroeck
ME, Peschbatenburg,
J, Janssen
W, Stam
H. Assessment of arm/hand functioning in children with a congenital transverse or longitudinal reduction deficiency of the upper limb. Disabil Rehabil. 2006;28(2):85–95.
+
Buffart
L, Roebroeck
ME, Van Heijningen
VG, Pesch-Batenburg
JM, Stam
HJ. Evaluation of arm and prosthetic functioning in children with a congenital transverse reduction deficiency of the upper limb. J Rehabil Med. 2007;39(5):379–386.
+
Cederholm
M, Haglund
B, Axelsson
O. Infant morbidity following amniocentesis and chorionic villus sampling for prenatal karyotyping. BJOG: Int J Gynaecol Obstet. 2005;112:394–402.
+
Chen
C-P. Syndromes and disorders associated with omphalocele (III): single gene disorders, neural tube defects, diaphragmatic defects and others. Taiwan J Obstet Gynecol. 2007:46(2):111–120.
+
Courvoisier
A, Sailhan
F, Thevenin-Lemoine
C, Vialle
R, Damsin
JP. Congenital tibial deficiencies: Treatment using the Ilizarov's external fixator. Orthpo Trauma: Surg Res. 2009;95:431–436.
+
Czeizel
AE, Kodaj
I, Lenz
W. Smoking during pregnancy and congenital limb deficiency. Biomed J. 1994;308:1473–1476.
+
Dillingham
TR, Pezzin
LE, MacKenzie
EJ. Limb amputation and limb deficiency: epidemiology and recent trends in the United States. South Med J. 2002;95(8):875–883.
+
Ephraim
PL, Dillingham
TR, Sector
M, Pezzin
LE, MacKenzie
EJ. Epidemiology of limb loss and congenital limb deficiency: a review of the literature. Arch Phys Med Rehabil. 2003;84:747–761.
+
Gold
NB, Westgate
M-N, Holmes
LB. Anatomic and etiological classification of congenital limb deficiencies. Am J Med Genet. 2011;155:1225–1235.
+
Holder-Espinasse
M, Devisme
L, Thomas
D,
et al. Pre- and postnatal diagnosis of limb anomalies: a series of 107 cases. Am J Med Genet. 2004;124:417–422.
+
Holmes
L. Teratogen-induced limb defects. Am J Med Genet. 2002;112:297–303.
+
Jain
S. Rehabilitation in limb deficiency. 2. The pediatric amputee. Arch Phys Med Rehabil.1996;77(3SuPPL.):S9–S13.
+
Jonsson
S, Caine-Winterberger
K, Branemark
R. Osseointegration amputation prostheses on the upper limbs: methods, prosthetics and rehabilitation. Prosthet Orthot Int. 2011;35(2):190–200.
+
Korkmaz
M, Erbahceci
F, Ulger
O, Topuz
S. Evaluation of functionality in acquired and congenital upper extremity child amputees. Acta Orthop Traumatol Turc. 2012;46(4):262–268.
+
Krajbich
JI. Lower-limb deficiencies and amputations in children. J Am Acad Orthop Surg. 1998;6(6):358–367.
+
Krebs
DE, Edelstein
JE, Thornby
MA. Prosthetic management of children with limb deficiencies. Phys Ther. 1991;71(12):920–934.
+
Lowry
RB, Bedard
T. Congenital limb deficiency classification and nomenclature: the need for a consensus. Am J Med Genet Part A. 2016;170A:1400–1404.
+
Man
LM, Chang
B. Maternal cigarette smoking during pregnancy increases the risk of having a child with a congenital digital anomaly. Plast Reconstr Surg. 2006;117(1):301–308.
+
Makhoul
IR, Goldstein
I, Smolkin
T, Avrahami
R, Sujov
P. Congenital limb deficiencies in newborn infants: prevalence, characteristics and prenatal diagnosis. Prenat Diagn. 2003;23:198–200.
+
McGuirk
CK, Westgate
MN, Homles
LB. Limb Deficiencies in Newborn Infants. Pediatrics. 2001; 108(4): 1–7.
+
Michielsen
A, Van Wijk
I, Ketelaar
M. Participation and quality of life in children and adolescents with congenital limb deficiencies: a narrative review. Prosthet Orthot Int. 2010;34(4):351–361.
+
Murray
CD, Being like everybody else: the personal meanings of being a prosthesis user. Disabil Rehabil. 2008;22:1–9.
+
Nelson
VS, Flood
KM, Bryant
PR, Huang
ME, Pasquina
PF, Roberts
TL. Limb deficiencies and prosthetic management. 1. Decision making in prosthetic prescription and management. Arch Phys Med Rehabil. 2006;87(3SUPPL.1):S3–S9.
+
Orhun
H, Saka
G, Bilgic
E, Kavakh
B. Lengthening of short stumps for functional use of prostheses. Prosthet Orthot Int. 2003;27(2):153–157.
+
Pierce
S, Fergus
A, Brady
B, Wolff-Burke
M. Examination of the functional mobility assessment tool for children and adolescents with lower extremity amputations. Pediatr Phys Ther. 2011;23(2):171–177.
+
Pruitt
SD, Varni
JW, Setoguchi
Y. Functional status in children with limb deficiency: development and initial validation of an outcome measure. Arch Phys Med Rehabil. 1996;77(12):1233–1238.
+
Pruitt
SD, Varni
JW, Seid
M, Setoguchi
Y. Prosthesis satisfaction outcome measurement in pediatric limb deficiency. Arch Phys Med Rehabil. 1997;78:750–4.
+
Rossi
R, Alexander
M, Cuccurullo
S. Pediatric Limb Deficiencies. Physical Medicine and Rehabilitation Board Review. New York: Demos Medical Publishing; 2004.
+
Sakkers
R, van Wijk
I. Amputation and rotationplasty in children with limb deficiencies: current concepts. J Child Orthop. 2016;10(6):619–626.
+
Scotland
TR, Galway
HR. A long-term review of children with congenital and acquired upper limb deficiency. Bone Joint J. 1983;65-B(3):346–349.
+
Skjaerven
R, Wilcos
AJ, Lie
TR. A population-based study of survival and childbearing among female subjects with birth defects and the risk of recurrence in their children. N Engl J Med. 1999;340:1057.
+
Smith
DG. Congenital limb deficiencies and acquired amputations in childhood. inMotion. 2006: 16(1):41–45.
+
Unglaub
F, Lanz
U, Hahn
P. Outcome analysis, including patient and parental satisfaction, regarding nonvascularized free toe phalanx transfer in congenital hand deformities. Ann Plast Surg. 2006;56(1):87–92.
+
Varni
JW, Setoguchi
Y. Screening for behavioral and emotional problems in children and adolescents with congenital or acquired limb deficiencies. Am J Dis Chil. 1992;146:103–107.
+
Watson
S. Current Topic: The principles of management of congenital anomalies of the upper limb. Arch Dis Child. 2000;83:10–17.
+
Webster
WS, Abela
D. The effect of hypoxia in development. Birth Defects Res. 2007;81:215.
++
++
CD is difficult to diagnose and may require several clinical examinations, several experts, and additional tests including fMRI and EEG (Cottencin, 2014).
"The diagnosis of conversion disorder is unusual in that psychiatrists typically have to rely on the skills of other doctors (most often neurologists) to be fully confident in its application".(Stone, 2011)
There are some diagnoses, such as panic disorder, that need to be tested by excluding the internal inconsistencies; but to be diagnosed with conversion disorder one needs to have evidence that there are internal inconsistencies with the disease the patient is believed to have. (Stone, 2011)
Signs and Symptoms:
The classic symptoms of conversion disorder include inconsistencies in repeated testing of sensation and muscle strength, manual muscle strength testing that does not correspond with the patient's functional abilities, and sensory impairments that do not follow anatomical patterns". (Ness, 2007)
Signs may include
"Jerky, exaggerated movements; unexplained tremors; bizarre gait patterns; and simultaneous contraction of agonist and antagonist muscles" (Ness, 2007)
Ataxia, tremors, tics, or paralysis of multiple limbs, and may be severely limiting (Ness, 2007)
Bilateral quadriparesis, dystonia, blindness, and dysphasia. (Heruti, 2002)
Pain or disturbed sexual functioning.
'La belle indifference' (Kaur, 2012)-
Patient may also appear detached from physical symptoms that would usually cause stress. (Heruti, 2002)
Patient will show a lack of concern regarding the severe symptom and the underlying psychological problem will get better. (MedlinePlus, 2014)
PNES can present in numerous ways.
Some individuals may report an aura prior to seizure while others do not. Onset is normally gradual, occurs when individual is awake and often includes motor manifestations. The most common observable manifestations of PNES are physical collapse or altered responsiveness. (Walker, 2009)
Positive neurological signs are the most reliable measures for diagnosing CD (Sonoo, 2014).
Characteristics of hysterical paresis: giving-way weakness, peculiar distributions of weakness.
Tests utilizing synergy to uncover nonorganic paresis: Hoover's test, Sonoo abductor test (Sonoo, 2014).
Psychological factors appear related as symptom initiation or exacerbation is preceded by conflict or stressor.
Patients with CD have higher scores on trauma scales and measures of dissociation and show hypervigilance to threat (Kaplan, 2013).
Specific clusters of positive signs that are indicative (highly reliable to suggestive) of the presence of CD (Daum, 2015):
Highly reliable:
Giveway weakness, drift without pronation, co-contraction, splitting the midline, splitting of vibration sense, and Hoover's sign. (Daum, 2015)
Reliable:
Spinal injury test, SCM-test, collapsing weakness, non-concavity of the palm, inconsistence of direction, non-anatomical sensory loss, psychogenic Romberg, and leg dragging. (Daum, 2015)
Suggestive:
Irregular drift, non-digiti quinti sign, falls always towards support, non-economic posture, knee buckling, and tremulousness. (Daum, 2015)
Medical Diagnostic testing
Imaging studies
Sensory and nervous tracts both remain intact regardless of clinical symptoms being present, even in a patient with pseudoseizures
Imaging studies should be avoided because this could prolong the disease course and reinforce their symptoms (Sonoo, 2014).
The neural structures of motor inhibition in CD patients and in the controls who simulate the inhibitions are corresponding uniformly in the inferior frontal gyri with some variation expected (Hassa, 2016)
DSM-IV-TR diagnostic criteria (American Psychiatric Association, 2000):
One or more symptoms or deficits affecting voluntary motor or sensory function that suggest a neurological or other general medical condition.
Psychological factors are judged to be associated with the symptoms or deficit because the initiation or exacerbation of the symptom or deficit is preceded by conflicts or other stressors.
The symptom or deficit is not intentionally produced or feigned (as in Factitious Disorder or Malingering).
The symptom or deficit cannot, after appropriate investigation, be fully explained by a general medical condition, or by the direct effects of a substance, or as culturally sanctioned behavior or experience.
The symptom or deficit causes clinically significant distress or impairment in social, occupational, or other important areas of functioning or warrants medical evaluation.
The symptom or deficit is not limited to pain or sexual dysfunction, does not occur exclusively during the course of Somatization Disorder, and is not better accounted for by another mental disorder.
The DSM-V has a diagnostic criteria (B) requiring that "clinical findings provide evidence for incompatibility between the symptom and recognized neurological or medical conditions" (American Psychiatric Association, 2013)
According to DSM-5 conversion disorder may be observed as the patient modifies a voluntary motor or sensory movement/feeling that does not match medical findings. (Nelson, 2017)
The DSM-V emphasizes the role of neurologists in making a positive diagnosis of CD (Daum, 2015).
The incompatibility between the symptom and recognized neurological or medical condition is established by negative findings and exclusion, but relies on presence of positive signs such as Hoover's sign in motor weakness, the ictal eye closure sign in seizure or an entrainment phenomenon in tremor (Daum, 2015).
Diagnosis of PNES
May be classified as psychogenic nonepileptic seizures (PNES) by using video EEG recordings, history, and psychological testing (Stonnington, 2006; Gedzelman, 2014)
Video EEG recording is currently the standard for diagnosis of PNES. This is due to the accuracy and reliability of EEG to monitor the electrical discharges set off during PNES. (Gedzelman, 2014)
The best indication of PNES: EEG background appears normal during waking state prior to the ictus, during the ictus, and immediately following the ictus. (Gedzelman, 2014)
Tremor Entrainment Test
Used for patients with a unilateral arm tremor
It involves "asking them to make a voluntary rhythmical movement with their unaffected arm. If the patient is either unable to perform this voluntary movement or the rhythm of the affected hand "entrains" to the rhythm of the voluntary movement this is strong evidence of a "conversion" tremor.' (Hallet, 2010)
Differential Diagnosis
++
Acute symptoms usually resolve within weeks (Feinstein, 2011)
Tends to be recurrent, separated by asymptomatic periods.
20-25% of patients may have symptoms recur within a year. (Feinstein, 2011)
"As many as 25% of these responders relapse or develop new conversion symptoms over time" (Stonnington, 2006)
If conversion syndrome becomes chronic, it may result in contractures or other complications. (Owens, 2001)
Evidence from a retrospective study suggests that patients with severe, long-standing motor CD can achieve significant improvements in functioning after admission to a neuropsychiatry unit (McCormack, 2014).
Good prognosis associated with:
Psychological conflict centering on Oedipal sexuality
Stable relationships, stable work history
Ability to develop trusting working doctor-patient relationship
Ability to feel and express emotion without developing incapacitating anxiety or depression
Ability to have psychological distance from consciously experienced emotion
Capacity for introspection
Symptoms related to environmental stress
Short duration of symptoms, and early diagnosis (Gelauff, 2014).
High satisfaction with care (Gelauff, 2014).
"Sudden onset, a definite stressor, good premorbid functioning, lack of comorbid psychiatric disorders, and absence of litigation proceedings related to the illness" (Blitztein, 2008)
Poor prognosis:
Delayed diagnosis, and co-existent personality disorder (Gelauff, 2014)
Longer duration of conversion symptoms (Blitztein, 2008)
Prognostic factors that vary:
Age, comorbid anxiety and depression, IQ, educational status, marital status, and pending litigation (Gelauff, 2014).
Gender had no association with prognosis (Gelauff, 2014).
+++
IV. Medical/Surgical Management
++
Medications:
Psychotropic drugs are used to treat comorbidities, such as depression and PTSD (Scevola, 2014).
Antidepressants (including selective serotonin reuptake inhibitors, tricyclic antidepressants, venlafaxine, and bupropion) may be used to treat underlying mood disorder (Hurwitz, 2003)
Neuroleptics may be used to treat somatic delusions (Hurwitz, 2003)
Electroconvulsive therapy may recover neurologic function while treating underlying disorder (Hurwitz, 2003)
The current method of treating CD is to use appropriate medication for comorbid symptoms. (Stonnington, 2006)
Cognitive Behavioral Therapy (CBT)
Cognitive Analytical Therapy (CAT)
+++
V. Therapeutic management
++
Practice Patterns (APTA, 2017)
ICD-10 Code: F44 Conversion disorder; "Dissociative and Conversion disorders" Psychotherapy
CD is stigmatized and associated with malingering; however, "it is not malingering nor a factitious disorder" (Cottencin, 2014).
Although no specific treatment exists, there is a consensus in favor of psychotherapy (Cottencin, 2014).
Explain to patients their physical complaint has a psychological cause (Cottencin, 2014; Vermeulen, 2014).
First explain what the diagnosis actually means, and then explain what they do not have (Vermeulen, 2014).
A diagnosis of CD may be very upsetting to a patient. A physical therapist or other health care provider must be sympathetic and understanding when discussing conversion disorder with a patient.
In order to deliver the diagnosis effectively and respectfully, clinicians have to first convince themselves before trying to convince patients (Cottencin, 2014).
When talking to a patient, explain that CD may be related to prior trauma or stress. Even though the traumatic event may have happened in the past, the physical symptoms usually begin at a later time following a new triggering event. (Stonnington, 2006)
"Combined consultation (medicine and psychiatry) is a useful tool to help patients" (Cottencin, 2014).
Because this disorder has such a large psychiatric component, using a well-rounded health care team would be most beneficial and is an important part of this treatment, which usually requires long-term intervention (Stonnington, 2006; Cottencin, 2014).
Working with the patient's family may be necessary when family and sociocultural factors are at the root of the disorder, especially in pediatric patient populations. Recognizing and treating comorbid psychiatric conditions, which would fall outside the scope of a physical therapist, are typically required (Stonnington, 2006).
Physical Therapy
Often used in combination with an effective behavioral therapy program in order to assist patients both physically and mentally. (Speed, 1996)
Suggested progression of physical interventions:
Develop patient rapport, maintain flexibility and early weight-bearing activities, gait training in parallel bars (provides success and confidence), and finally gait in open environment/community." (Kaur, 2012)
"A positive rehabilitative approach is adopted with graded steps and positive reinforcement" (McCormack, 2014).
Twice weekly interventions; techniques may include stretching and massage (if dystonia), exercises targeting posture, stability, balance, and strength, as well as specific functional training for any activities with limitations (Ness, 2007; McCormack, 2014).
PTs are encouraged to help the patient recognize inconsistences in their presentation and work to 'relearn' control over the body, 'teach' muscles how to reconnect with nerves and how to 'overcome mental blocks'. Once the patient can identify these 'triggers', they can become autonomous with their intervention. (McCormack, 2014).
Treatment should
Be symptom-based in order to regain functional capacity in the affected areas (Heruti, 2002).
Focus on prevention of secondary disabilities such as muscle weakness due to disuse atrophy. (Heruti, 2002)
Focus on reducing undesirable behaviors and strengthening desirable ones using positive reinforcement. (Heruti, 2002).
FES and biofeedback techniques have been shown to be effective interventions when combined with physical exercise. (Heruti, 2002)
"Mnemonic for remembering critical components for the successful management of patients with conversion disorder (Neese, 2007):
C Confrontation is avoided
O Organic illness is ruled out
N Not allowed to progress to next step, without mastery of previous step
V Vulnerability requires "face saving" options
E Establish concrete measures of progress/set goals
R Reinforcement through positive feedback
S Stress management techniques
I Ignore abnormal behavior
O Open communication with rehab team and family
N Need for consistency in care providers
Outcome Measures:
A 2014 systematic review determined that the efficacy of physical therapy management, specific to the pediatric population, was limited and had poor evidence due to the lack of valid, sensitive and objection functional outcome measures. (FitzGerald, 2014)
The modified gait abnormality rating scale (GARS-M) has been found to have excellent inter- and intra-rater reliability, using an objective scale to assess gait disturbances in individuals with CD, to quantify gait disturbances and monitor improvements throughout rehabilitation. (Vandenberg, 2015)
Contraindications will change from patient to patient depending on symptoms. Precautions will mainly include loss of balance and detrained neuromuscular system. A gait belt should be used while the patient is still regaining strength and balance, particularly if unpredictable in their behavior.
+++
VI. Consumer and Professional Resources
++
Websites
ExperienceProject.com:
MayoClinic.com:
UpToDate.com:
NYU Langone Medical Center:
http://www.med.nyu.edu/content?ChunkIID=96743
Provides an easy to read patient oriented overview of conversion disorder which can be used to supplement the information given by the primary caregiver. It includes basic information as well as current treatment options.
The Drug Information Online Health Guide:
Medscape:
The Generic and Rare Diseases (GARD):
National Alliance on Mental Illness:
Functional and Dissociative Neurological Symptoms: A Patient's Guide:
http://neurosymptoms.org
Provides information on symptoms, causes, misdiagnosis, possible treatments and an educational explanation of what occurs in the brain when symptoms are felt.
+
Allpsyc. AllPsych Online. 29 November 2011. Heffner Media Group. 14 February 2013.
+
Allin
M, Streeuwitz
A, Cuttis
V. Progress in understanding conversion disorder. Neuropsychiatr Dis Treat. 2005;1(3):205–298.
+
American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. 5th ed., Washington: American Psychiatric Assoc., 2013.
+
American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. 4th ed., text rev. Washington: American Psychiatric Assoc., 2000.
+
Aybek
S, Nicholson
TR, Zelaya
F, O'Daly
OG, Craig
TJ, David
AS, Kanaan
RA. Neural correlates of recall of life events in conversion disorder. JAMA Psychiatry. 2014;71(1):52–60.
+
Blitzstein
SM. Recognizing and Treating Conversion Disorder. Virtual Mentor. 2008;10(3):158.
+
Cottencin
O. Conversion disorders: psychiatric and psychotherapeutic aspects. Neurophysiol Clin. 2014;44(4):405–10.
+
Daum
C, Gheorghita
F, Spatola
M,
et al. Interobserver agreement and validity of bedside 'positive signs' for functional weakness, sensory and gait disorders in conversion disorder: a pilot study. J Neurol Neurosurg Psychiatry. 2015;86:425–430.
+
Feinstein,
A. Conversion disorder: advances in our understanding. Can Med Assoc J. 2011;183(8):915–920.
+
FitzGerald
TL, Southby
AK, Haines
TP, Hough
JP, Skinner
EH. Is physiotherapy effective in the management of child and adolescent conversion disorder? A systematic review. J Paediatr Child Health. 2015;51(2):159–167.
+
Gedzelman
E, LaRoche
S. Long-term video EEG monitoring for diagnosis of psychogenic nonepileptic seizures. Neuropsychiatr Dis Treat. 2014;10:1979–86.
+
Gelauff
J, Stone
J, Edwards
M, Carson
A. The prognosis of functional (psychogenic) motor symptoms: a systematic review. J Neurol Neurosurg Psychiatry. 2014;85(2):220–6.
+
Hallet
M. Physiology of Psychogenic Movement Disorders. J Clin Neurosci. 2010;17(8):959–965.
+
Hassa
T, deJel
E, Tuescher
O, Schmidt
R, Schoenfeld
M. Functional networks of motor inhibition in conversion disorder patients and feigning subjects. NeuroImage Clin. 2016;11:719–727.
+
Heruti
RJ, Levy
A, Adunski
A, Ohry
A. Conversion motor paralysis disorder: overview and rehabilitation model. Spinal Cord. 2002;40:327–334.
+
Hurwitz,
T. Somatization and Conversion Disorder. Can J Psychiatry. 2003;49:172–178
+
Kaplan
MJ, Dwivedi
AK, Privitera
MD, Isaacs
K, Hughes
C, Bowman
M. Comparisons of childhood trauma, alexithymia, and defensive styles in patients with psychogenic non-epileptic seizures vs. epilepsy: implications for the etiology of conversion disorder. J Psychosom Res. 2013;75(2):142–6.
+
Kaur
J, Garnawat
D, Ghimiray
D, Sachdev
M. Conversion disorder and physical therapy. Delhi Psychiatry J. 2012;15(2):394–397.
+
McCormack
R, Moriarty
K, Mellers
JD, Shotbolt
P, Pastena
R, Landes
N, Goldstein
L, Fleminger
S, David
AS. Specialist inpatient treatment for severe motor conversion disorder: a retrospective comparative study. J Neurol Neurosurg Psychiatry. 2014;85(8):895–900.
+
Nasiri
H, Ebrahimi
A, Zahed
A, Arab
M, Samouei
R. Cognitive-analytical therapy for a patient with functional neurological symptom disorder-conversion disorder (psychogenic myopia): A case study. J Res Med Sci. 2015;20(5):522–524.
+
Neer
S, Trachik
B, Munyan
B, Beidel
D. Comprehensive Treatment: Intensive Exposure Therapy for Combat-Related PTSD and Comorbid Conversion Disorder. Clin Case Stud. 2016;15(5):343–359.
+
Nelson
E, Wu
J. Postoperative Conversion Disorder Presenting as Inspiratory Stridor and Hemiparesis in a Pediatric Patient. Am J Case Rep. 2017;18:60–63.
+
Ness
D. Physical Therapy Management for Conversion Disorder: Case Series. J Neurol Phys Ther. 2007;31:30–39.
+
Nicholson
TR, Aybek
S, Kempton
MJ, Daly
EM, Murphy
DG, David
AS, Kanaan
RA. A structural MRI study of motor conversion disorder: evidence of reduction in thalamic volume. J Neurol Neurosurg Psychiatry. 2014;85(2):227–9.
+
Owens,
C, Dein
S. Conversion Disorder: the modern hysteria. Adv Psychiatr Treat. 2001;3:1755–1776.
+
Scevola
L, Korman
G, Oddo
S, Kochen
S, D'Alessio
L. Psychogenic non-epileptic sizures. Differential diagnosis with epilepsy, clinical presentation and therapeutic approach. Vertex. 2014;25(116):266–73.
+
Sonoo
M. Neurology of hysteria (conversion disorder). Brain Nerve. 2014;66(7):873–71.
+
Speed
J. Behavioral management of conversion disorder: retrospective study. Arch Phys Med Rehabil. 1996;77(2):147–54.
+
Stone
J, Carson
A, Duncan
R, Roberts
R, Warlow
C, Hibberd
C, Coleman
R, Cull
R, Murray
G, Pelosi
A, Cavanagh
J, Matthews
K, Goldbeck
R, Smyth
R, Walker
J, Sharpe
M. Who is referred to neurology clinics? – the diagnoses made in 3781 new patients. Clin Neurol Neurosurg. 2010;112(9):747–51.
+
Stone
J, LaFrance
Jr C, Brown
R, Spiegel
D, Levenson
J, Sharpe
M. Conversion Disorder: Current Problems and Potential Solutions for DSM-5. J Psychosom Res. 2011;71:369–376
+
Stonnington
C, Barry
JJ, Fisher
RS. Conversion Disorder. Am J Psychiatry. 2006;163:1510–1517.
+
Vandenberg
JM, George
DR, O'Leary
AJ, Olson
LC, Strassburg
KR, Hollman
JH. The modified gait abnormality rating scale in patients with a conversion disorder: A reliability and responsiveness study. Gait Posture. 2015;41(1):125–129.
+
Vermeulen
M, Hoekstra
J, Kuipers-van
Kooten MJ, van der Linden
EA. Management of patients with conversion disorder. Ned Tijdschr Geneeskd. 2014;158(3):A6997.
+
Vuilleumier
P. Functional neuroanatomical correlates of hysterical sensorimotor loss. Brain. 2001;124(6):1077–1090.
++
Delirium is a common and seriously under-recognized problem
It may be missed in up to 2/3 of cases (Collins, 2009)
Delirium is known as "the Great Imitator" as it is often mistaken for depression, mania, anxiety, psychosis
Incidence and Prevalence:
Delirium is the most frequent cause of acute onset cognitive impairment in the inpatient setting (Caplan, 2010).
The prevalence of detected delirium in ICU patients is 32.3% (Salluh, 2010).
Following hip fracture surgery, incidence of delirium is higher if age >80, admitted from an institution, or having a previous diagnosis of dementia (Smith, 2017)
Delirium presents in 35 to 85% of critically ill hospitalized patients, but only 1 to 2% of healthy individuals (Ali, 2011).
Multiple Causes
Medications such as narcotics, sedatives, anticholinergic drugs, and antipsychotic drugs.
Diseases or illnesses including anemia, infections (e.g. UTI, respiratory), shock, and stroke (Ali, 2011)
Leukocytosis and hyponatremia can be significant causes of delirium. (Rai, 2014)
Surgical procedures (with a higher incidence occurring after cardiac and orthopedic surgeries); in 15 to 53% of older adults postoperatively (Ali, 2011). Post-surgery delirium causes an increased risk for prolonged hospital stays, discharge to an institution versus to home, and 30-day readmission after discharge (Zenilman, 2017).
Environmental factors including emotional stress, pain, and catheters (Greer, 2011)
Electrolyte or other body chemical disturbances (Board, 2011)
Alcohol withdrawal (Board, 2011)
Wernicke encephalopathy-underlying cause is thiamine deficiency (Osiezagha, 2013)
Hypoxia, hypertensive crisis, meningitis, poisons. (Caplan, 2010)
Although the pathophysiological mechanisms that cause delirium are not understood at this time, hypotheses that exist include:
Increasing dopamine and decreasing acetylcholine levels are responsible for delirium
Inflammation, toxic or metabolic derangements resulting in encephalopathy, hypoxia, and neurotransmitter imbalance (Hipp, 2012)
++
++
Delirium is associated with increased length of stay at a hospital, increased mortality upon discharge, institutionalization, and a 10 to 26% mortality rate of older adults hospitalized over a 12-month period (Siddiqi, 2006; Gower, 2012)
Effects of comorbidities:
Identifying the cause of delirium lead to better prognosis in terms of decreased hospital stay, mortality and disability (Rai, 2014).
May have later development of PTSD if delirium remains undiagnosed (Caplan, 2010)
Delirium typically resolves within a few hours or days, but can have lingering effects.
In patients experiencing delirium after undergoing a CABG, the long term risk of death and stroke was increased compared to the patients undergoing a CABG who did not experience delirium (Martin, 2011).
Recovery is poor in 69% of patients with delirium, at mean follow-up at 104 days (Dasgupta, 2014).
Functional dependence at discharge after stroke correlates with a delirium diagnosis during the hospital stay (Alvarez-Perez, 2016).
+++
IV. Medical / Surgical Management
++
Early recognition and treating the underlying cause of delirium is key.
Prevention:
Re-orientation strategies, visual and hearing aids, sleep protocols, and nursing education (Farina, 2015).
No FDA-approved drug for treatment available at this time (Vanderbilt, 2011)
Dexmedetomidine has been shown to decrease the duration of delirium in patients instead of using benzodiazepine however, the levels of dexmedetomidine were at levels unapproved by the FDA (Pandharipande, 2007)
Pharmacological interventions for delirium are aimed at decreasing dopaminergic activity and increasing cholinergic activity (Somes, 2010).
Haloperidol prophylaxis treatment in the emergency department is being studied to prevent development of delirium during hospital stay (Schrijver, 2014).
Haloperidol lactate has calming effect and can be administered parenterally, but does not decrease length of delirium. However, antipsychotics (olanzapine and quetiapine furmate) can reduce the length of delirium (Farina, 2015).
The ABCDE approach (Awakening and Breathing Coordination, Delirium Monitoring, Early Mobility and Exercise) is a non-pharmacological, interdisciplinary approach used in the ICU to treat delirium. It encompasses early mobility, screening, and coordinated care (Hipp, 2012).
+++
V. Implications for Therapeutic Management
++
Neuromuscular Practice Patterns (APTA, 2017):
Because immobility is a risk factor for delirium early mobility is key.
Early active and passive physical therapy decreases length of stay and duration of delirium (Somes, 2010).
PT in the earliest days of illness result in better functional outcomes, shorter duration of delirium, and more ventilator-free days in patients receiving mechanical ventilation in the ICU (Kalabalik, 2013).
Prevention:
Physical therapists should be involved as part of a multidisciplinary team to assess risk factors for delirium, watch for signs of pain, and mobilize at risk patients. Both bed mobility (ROM) exercises and ambulation are appropriate (NICE delirium guideline, 2010).
Physical therapists spend more time with patients than their physicians- this puts them in a better position to recognize the mental changes that may signal early delirium (Neufeld, 2011).
Educate patient and family about the progression of delirium with possible underlying issues (Board, 2011).
Speak in a soothing tone, give simple one step instructions, and generally take care not to agitate the patient
+++
VI. Consumer and Professional Resources
++
Helping caregiver:
Beneficial organizations:
American Delirium Society
National Family Caregivers Association
Alzheimer's Association
National Institute on Aging
American Geriatric Association
The Hospital Elder Life Program (HELP)
The Mayo clinic
Vanderbilt University provides an overview of delirium at
The ICU Delirium and Cognitive Impairment Study Group
HealthinAging.org –
+
Ali
S, Milapkumar
P, Shagufta
J, Rahn
KB, Tejas
P, Marwah
S, Wayne
JR, Amir
A. Insight into Delirium. Innovations in Clinical Neuroscience. 2011; 8(11): 25–34.
+
Alvarez-Perez
F, Paiva
F. Prevalence and Risk Factors for Delirium in Acute Stroke Patients. A Retrospective 5-Years Clinical Series. Journal of Stroke and Cerebrovascular Diseases. 2016.
+
American Association of Critical Care Nurses. Delirium Assessment and Management. Crit Care Nurse. 2012; 32(1); 79–84.
+
Bhattacharya
B, Maung
A, Barre
K, Maerz
L, Rodriguez-Davalos
M, Schilsky
M, Mulligan
D, Davis
K. Postoperative delirium is associated with increased intensive care unit and hospital length of stays after liver transplantation. Journal of Surgical Research. 2017; 207: 223–228.
+
Collins
N, Blanchard
M, Tookman
A, Sampson
E. Detection of delirium in the acute hospital. Oxford Journals. 2009;201: 131–135.
+
Caplan
J, Rabinowitz
T. An Approach to the Patient with Cognitive Impairment: Delirium and Dementia. Med Clin N Am. 2010;94:1103–1116.
+
Dasgupta
M, Brymer
C. Prognosis of delirium in hospitalized elderly: worse than we thought. Int J Geriatr Psychiatry. 2014;29:497–505.
+
Farina
N, Smithburger
P, Kane-Gill
S. Screening and Management of Delirium in Critically Ill Patients. Hosp Pharm. 2015;50(8):667–671.
+
Gower
LE, Gatewood
MO, Kang
CS. Emergency Department Management of Delirium in the Elderly. West J Emerg Med. 2012;13(2):194–201.
+
Grover
S. Assessment scales for delirium: A review. World J Psychiatry. 2012;2(4):58. doi:10.5498/wjp.v2.i4.58
+
Hipp
DM, Ely
WE. Pharmacological and Nonpharmacological Management of Delirium in Critically Ill Patients. Neurotherapeutics. DOI 10.1007/s13311-011-0102-9
+
Hyo
JL, Deuk
SH, Seong
KW, Ik
SC, Sengmi
B, Jeong
LK. Early assessment of Delirium in Elderly Patients after Hip Surgery. Psychiatry Investig. 2011;8:340–347.
+
Jang
S, Jung
K, Yoo
W, Jung
M, Ohn
S. Risk Factors for Delirium During Acute and Subacute Stages of Various Disorders in Patients Admitted to Rehabilitation Units. Ann Rehabil Med. 2016: 40(6): 1082–1091.
+
Kalabalik
J, Brunetti
L, El-Srougy
R. Intensive Care Unit Delirium A Review of the Literature. J Pharm Pract. 2013:0897190013513804. doi:10.1177/0897190013513804
+
Martin
B, Buth
K, Rakesh
A, Baskett
R. Delirium: A cause for Concern Beyond the Immediate Postoperative Period. Ann Thorac Surg. 2011 Dec 24;93:1114–1120.
+
Michaud
CJ, Thomas
WL, McAllen
KJ. Early Pharmacological Treatment of Delirium May Reduce Physical Restraint Use A Retrospective Study. Ann Pharmacother. 2013:1060028013513559. doi:10.1177/1060028013513559.
+
Mittal
V, Muralee
S, Williamson
D, McEnerney
N, Thomas
J, Cash
M, Tampi
R. Delirium in the Elderly: A Comprehensive Review. Am J Alzheimers Dis Other Demen. 2011;26(2):97–109.
+
Neufeld
KJ, Joseph
BO, Rosenberg
PB, Mears
SC, Lee
HB, Kamdar
BB, Sieber
FE, Krumm
SK, Walston
JD, Hager
DN, Touradji
P, Needham
DM. The Johns Hopkins Delirium Consortium: A Model for Collaborating Across Disciplines and Departments for Delirium Prevention and Treatment. J AM Geriatr Soc. 2011;59:S244–S248. doi: 10.1111/j.1532-5415.2011.03672.x
+
Ohta
T, Murao
K, Miyake
K, Takemoto
K. Melatonin Receptor Agonists for Treating Delirium in Elderly Patients with Acute Stroke. J Stroke Cerebrovasc Dis. 2013;22(7):1107–10.
+
Osiezagha
K, Ali
S, Freeman
C, Barker
C, Jabeen
S, Maitra
S, Olagbemiro
Y, Richie
W, Bailey
RK. Thiamine Deficiency and Delirium. Innov Clin Neurosci. 2013;10(4):26–32.
+
Pandharipande
PP, Pun
BT, Herr
DL,
et al. Effect of Sedation With Dexmedetomidine vs Lorazepam on Acute Brain Dysfunction in Mechanically Ventilated Patients: The MENDS Randomized Controlled Trial.
JAMA.[JAMA and JAMA Network Journals Full Text] 2007;298(22):2644–2653. doi:10.1001/jama.298.22.2644.
+
Pisani
MA, Araujo
KLB, Ness
P, Zhang
Y, Ely
EW, Inouye
SK. A Research Algorithm to Improve Detection of Delirium in the Intensive Care Unit. Crit Care. 2006; 10(4): R121. doi:10.1186/cc5027.
+
Rai
D, Garg
RK, Malhotra
HS. Acute confusional state/delirium: An etiological and prognostic evaluation. Ann Indian Acad Neurol. 2014;17:30–4.
+
Salluh
JI, Soares
M, Teles
JM,
et al. Delirium epidemiology in critical care (DECCA): an international study. Crit Care. 2010;14(6):R210. doi:10.1186/cc9333
+
Schrijver
EJ, de Vries
OJ, Verburg
A, de Graff
K, Bet
PM, van de Ven
PM, Kamper
AM, Dieppeveen
SHA, Anten
S, Siegel
A, Kuiperi
E, Lagaay
AM, van Marum
RJ, van Strien
AM, Boelaarts
L, Pons
D, Kramer
MHH, Nanayakkara
PWB. Efficacy and safety of haloperidol prophylaxis for delirium prevention in older medical and surgical at-risk patients acutely admitted to hospital through the emergency department: study protocol of a multicenter, randomised, double-blind, placebo-controlled clinical trial. BMC Geriatrics 2014;14:96. doi:10.1186/1471-2318-14-96.
+
Siddiqi
N, House
A, Holms
J. Occurrence and outcome of delirium in medical in-patients: a systemic literature review. Age and Aging. 2006;35:350–364.
+
Smith
T, Cooper
A, Peryer
G, Griffiths
R, Fox
C, Cross
J. Factors predicting incidence of post-operative delirium in older people following hip fracture surgery: a systematic review and meta-analysis. Int J Geriatr Psychiatry. 2017. Reference number: R21697.
+
Smulter
N, Lingehall
HC, Gustafson
Y, Oloffson
B, Engstrom
G. Validation of Confusion Assessment Method in Detecting Postoperative Delirium in Cardiac Surgery Patients. Am J Crit Care. 2015;24(6):480–7.
+
Somes
J, Donatelli
N, Barrett
J. Sudden Confusion and Agitation: Causes to Investigate! Delirium, Dementia, Depression. J Emerg Nurs. 2010;36:486–8.
+
Treloar
A, MacDonald
A. Outcome of Delirium: Part I. Int. J. Geriatr. 1997;12:609–613.
+
Yang
F, Marcantonio
E, Inouye
S, Kiely
D, Rudolph
J, Fearing
M, Jones
R. Phenomenological Subtypes of Delirium in Older Persons: Patterns, Prevalence, and Prognosis. Psychosomatics. 2009;50(3):248–254.
+++
Dementia / Alzheimer's disease
++
– Dementia is defined as "a clinical syndrome caused by neurodegeneration and characterized by inexorably progressive deterioration in cognitive ability and capacity for independent living." (Prince, 2013) and is public health priority due to its large social and financial effects (Groot, 2016).
– Findings of dementia vary among patients depending on which area of the brain is involved.
– Dementia diagnosed in individuals before the age of 65 is termed "early-onset dementia." (Fadil, 2009)
– Prevalence
Dementia is estimated to effect 35 million people worldwide. It is expected to increase to about 135 million by the year 2050 (Wilkins, 2016).
Approximately 3.8 million Americans suffer from dementia and 60–80% of these cases are believed to be due to Alzheimer's disease (McLaren, 2013).
Estimates of prevalence of dementia range from 5% at the age of 65 to 40% at the age of 80 (Luzny, 2014).
Patients with schizophrenia are more likely to develop dementia than those without (Ku, 2016).
– Vascular dementia is the second most common cause of dementia behind Alzheimer's disease. (O'Brien, 2015)
– Causes of dementia include brain tumors, alcohol abuse, low B12 levels, central nervous system infections, lack of thyroid hormone, and depression.
– Dementia can be classified into four categories:
Loss of recent memory
Loss of recent memory in patients without attention deficits
Unilateral or bilateral lesions in the hippocampi or other related structures (fornices, mammillary bodies, dorsomedial thalamus) (Whitehouse, 1993)
Lesions to the above mentioned areas cause verbal memory deficits if it occurs in the patient's dominant hemisphere
Cortical dementia
Subcortical dementia
Lesions that cause subcortical dementia occur predominately in the basal ganglia, the brainstem, and the cerebellum. (Turner, 2002)
Disorders/Diseases that cause it are:
➢ Steele-Richardson- Olchewsky disease: caused by deterioration of brain volume
➢ Parkinson disease (PD): results from abnormal microscopic deposits ("Lewy bodies") that damage the brain cells over time
➢ Huntington's disease: progressive brain disorder with cognitive impairment caused by a single defective gene on chromosome 4
➢ HIV-associated dementia (HAD): There is little bases to confidently determine the etiology of cognitive impairment in a patient with HIV infection. Existing data suggests that there may be an overlap in neuropathology that is reasonable to consider. (High, 2006) More recently, a study done in Uganda found that subtypes of HIV may have a different biological impact on neurological complications of HIV infection. (Sacktor, 2009)
Vascular dementia (VaD)
++
– In order for an individual to be diagnosed with dementia, the patient must show presence of the signs and symptoms for at least six months. (Frances, 2000)
– Signs and symptoms of dementia may be reversible depending on the cause of the disease. (Frances, 2000)
– Symptoms of dementia must include a decline in memory and decline in one of the following cognitive functions: (Alzheimer's Association, 2012)
Ability to generate coherent speech or understand written or spoken language
Ability to recognize or identify objects (if their sensory system is intact)
Ability to perform motor tasks (if motor and sensory systems intact, as well as understanding of task)
Ability to think conceptually, plan and carry out a difficult task, and make judgments
– Recent studies have shown that a diagnosis of dementia in Huntington's disease should be at the point where there is demonstrable evidence of impairment in at least two areas of cognition (e.g. attention, speed of processing, executive functions, visuospatial abilities, memory) but without a requirement of memory impairment. (Peavy, 2010)
– Signs and Symptoms:
Loss of recent memory
Difficulty with visuospatial tasks
It should be noted that "absent-mindedness" is not a legitimate characteristic of this type of dementia. (Reeves, 2004)
➢ Absent-mindedness is more commonly linked to age-related changes in the hippocampal-temporal lobe function and is a common complaint of individuals over sixty years of age
➢ Absent-mindedness is considered a "benign memory difficulty" and can be differentiated from pathological loss of learning capacity.
➢ These patients are cognizant that they have problems remembering things and are able to bring themselves in to see a physician. This shows that they have intact insight.
➢ A patient with pathological memory loss will have problems with imprinting new memories and will not be aware that they have a memory problem. A concerned family member must bring these patients to the physician.
Cortical dementia
Characterized by cognitive dysfunction of language, praxis, and visuospatial functions
Stroke
Alzheimer's disease
➢ Difficulty remembering newly learned information
➢ Disorientation
➢ Mood/personality changes
➢ Confusion about events, time, and place
➢ Suspicions about family and friends
➢ Difficulty speaking, swallowing, and walking
Frontotemporal dementia
➢ Loss of semantic memory
➢ Non-fluent aphasia
➢ Initial personality changes such as disinhibition and impulsivity
➢ Loss of empathy
➢ May display obsessive-compulsive behavior (Pressman, 2014)
Frontal Lobe Dementia
➢ Intellectual function may be intact upon routine examination, but the patient's behavior may be inappropriate (disinhibited)
➢ In later stages, calculation, proverb, and similarity tests are poorly done
➢ Presence of regressive reflexes that are more common in infants (grasp reflex, gait apraxia, suck reflex) (Reeves, 2004)
Lewy body disease
➢ Visual hallucinations
➢ Parkinson's symptoms (hunched posture, rigid muscles, etc.)
➢ Delusions
➢ Trouble interpreting visual information
➢ Depression
➢ Olfactory dysfunction
➢ Mood and sleep disorders (Fujishiro, 2013)
Creutzfieldt-Jakob disease
➢ Depression
➢ Agitation, apathy, and mood swings
➢ Muscle stiffness, twitches and involuntary jerky movements
Subcortical dementia
Characterized by motor dysfunction, attention and concentration deficits, and improvements of memory with prompting (in some cases)
These disorders and diseases that cause dementia often affect memory and motivation in the individual.
Steele-Richardson-Olchewsky disease
Parkinson's disease
➢ Motor symptoms must be present for 1 year prior to the onset of dementia to be classified as PD dementia (Goldman, 2014)
➢ Irritability and anxiety
➢ Muffled speech
➢ Shuffling gait
➢ Sleep disturbances
➢ Poor visuospatial function (Goldman, 2014)
➢ Bradyphrenia-slow thinking (Turner, 2002)
Huntington's disease (cognitive deficits occur later in the progression)
➢ Memory performance is affected by slowed retrieval largely independent of the ability to store information and motor slowing.
➢ Bradyphrenia
➢ Attention deficits
➢ Impaired planning and problem solving
➢ Visuoperceptual and construction deficits (Peavy, 2010)
➢ Mood disorders
➢ Personality changes
➢ Irritable and explosive behavior (Hamdy, 2001)
HIV-associated dementia
➢ Progressive cognitive decline, especially in attention and concentration
➢ Difficulty understanding written material
➢ Executive functions impaired
➢ Depression
➢ Anxiety
➢ Motor dysfunction: tremors, ataxia, and frequent falls
➢ Apathy (Hamdy, 2001)
Vascular Dementia
– Other signs and symptoms of dementia
Neuropsychiatric symptoms (NPS), also known as behavioral and psychological symptoms of dementia (BPSD), can affect dementia patients ubiquitously across all stages and etiologies, and include: aggression, agitation, depression, anxiety, delusions, hallucinations, apathy, disinhibition, sleep disturbances, and psychosis (Fraker, 2014; Wilkins, 2016).
Paranoia
Personality changes
Depression in 20-30% of people with dementia (Geddes, 2005)
Anxiety in 20% of people with dementia (Geddes, 2005)
Motor perseveration (repeating the same movement because of inability to inhibit ongoing activity) (Reeves, 2004)
– Diagnostic Testing
Neuropathological confirmation of a diagnosis of dementia is rarely possible during life (Koepsell, 2013).
Diagnosis is primarily made based on a clinical assessment (Koepsell, 2013).
Brain scans such as CT, MRI, EEG, PET, and SPECT
➢ MRI
Can be used as a predictor of Alzheimer's disease in those with early onset dementia by evaluating changes in whole brain volume, hippocampal and entorhinal cortical volume (Jagust, 2006)
It has been found that patients with smaller values in whole brain volume, and hippocampal and entorhinal cortical volume are at risk for Alzheimer's disease.
➢ PET Scans
Fluorodeoxyglucose PET imaging can reveal a patient's pathology and provide markers of clinical status and disease progression.
PET scans have been found to be most useful in differentiating between Alzheimer's disease and frontotemporal lobe degeneration.
➢ Single-photon emission computed tomography (SPECT)
➢ Diagnosis of Alzheimer's disease involves the identification of two biomarker categories: (Alzheimer's Association, 2012)
Cognitive and neuropsychological tests
➢ It is important to understand that no single test will be able to diagnose all forms of dementia. Multiple tests may need to be utilized in order to determine the type of dementia. (Jacova, 2007)
➢ Tests include but are not limited to:
Abbreviated Mental Test Score (AMTS)
Mini Mental State Examination (MMSE)
Modified Mini-Mental State Examination (3MS)
Cognitive Abilities Screening Instrument (CASI)
Montreal Cognitive Assessment (MoCA) (Korczyn, 2012)
Vascular Dementia Assessment Scale (VADAS-cog) (O'Brien, 2015)
Hopkins Verbal Learning Test (HVLT): This test is less influenced by demographic factors, such as age and education, compared to MMSE. It has been translated in multiple languages and it has been proven feasible to administer in different countries with minimal impact from culture/ethnicity (Xu, 2015)
Neurological evaluation
Psychiatric evaluation
Genetic testing (Pressman, 2014)
++
– In general, dementia is a progressive condition; however, the rate of progression varies between patients. (Gauthier, 2006)
– The prognosis of dementia depends on how rapidly the disease process progresses.
While some patients may decline in cognitive function gradually, others may have a more rapid decline.
Patients with a rapid decline rate will have a poorer prognosis than those who decline at a slower rate.
– As the dementia progressively worsens, quality of life and lifespan will decrease. (Covinsky, 2004)
– Dementia incidence increases with age, specifically ages 85-89 and 90 and older (Tom, 2015).
– There is a smaller chance of finding a reversible cause of dementia with chronic degenerative diseases as compared to acute dementias.
– Expected Complications
– Life Expectancy
Greater educational attainment has a positive correlation with life expectancy in people with dementia, increasing it by 0.5 years (Tom, 2015).
Women with Alzheimer's disease were found to have a life expectancy of 5.7 years, as compared to 4.2 years for men after diagnosis (Tom, 2015).
Mortality is higher with vascular dementia with a mean survival rate of 3-5 years mainly because of the cardiovascular and cerebrovascular causes. (O'Brien, 2015)
+++
IV. Medical/Surgical Management
++
– Suggested primary prevention of dementia:
"Decrease all known risk factors closely linked to etiopathogenesis of neurodegenerative dementia – smoking habits, early detection and treatment of hyperlipidemia, arterial hypertension, hyperglycemia and hyperuricemia" (Luzny, 2014).
– Multiple medications will be used to treat the varying symptoms of dementia because there currently is no FDA approved medication that will treat all of the neuropsychiatric symptoms (NPS) of dementia (Wilkins, 2016).
– Medications are selected based on the specific etiologic type of dementia (Koepsell, 2013)
Cholinesterease inhibitors are first-line treatment for PD and AD dementia (Goldman, 2014)
SSRI medication can be an effective pharmacological intervention when a symptom of the dementia is agitation. (Wilkins, 2016)
– Common medications used:
Antipsychotics: Haloperidol, Risperidone, Olanzapine
Mood stabilizers: Fluoxetine, Imipramine, Citalopram, Trazodone
Stimulants: Methylphenidate
Symptom-slowing and memory-sustaining drugs: Donepezil (Aricept), Rivastigmine (Exelon), Galantamine (Reminyl), Memantine (Namenda)
+++
V. Implications for Therapeutic Management
++
– Preferred Practice Patterns (APTA, 2017)
Pattern 5A: Primary Prevention/Risk Reduction for Loss of Balance and Falling
Pattern 5E: Impaired Motor Function and Sensory Integrity Associated With Progressive Disorders of the Central Nervous System
– Role of the Physical Therapist
"Exercise has been shown to benefit mood, cognition, and overall health in patients with dementia." (Pressman, 2014) and should be introduced as a main intervention, along with medications (Groot, 2016).
Exercise as a type of therapy improved symptoms in all types of dementia, and should include aerobic exercise, which had a positive impact on dementia symptoms (Groot, 2016).
Interventions shown to have positive results on at least one functional outcome measure: "aerobic exercise (usually walking), resistance training or weightlifting, and balance and flexibility training" (McLaren, 2013).
A one-year physical therapy exercise program that includes strengthening, flexibility, and balance training can slow the progression of Alzheimer's disease. (Buchner, 2007)
A consistent exercise program can also potentially slow the decline of ADLs in people with dementia (Toots, 2016).
➢ Gait training can increase gait velocity, step length, etc.; Patients with mild dementia frequently exhibit "cautious gait" (Wilson, 2000).
Interventions need to be individualized to the patient and their stage of disease progression, and include activities the patient enjoys in order to increase compliance.
Exercise and modifications in the clinic to help with AD:
➢ Increased lighting in the early evening can decrease agitation sometimes called "sundowning".
➢ A structured plan appears to help with restlessness.
➢ Programs done during the day help to control nighttime pacing which leads to daytime drowsiness the following day.
➢ A multimodal exercise program is associated with a reduction in NPS in patients with AD and contributes to attenuation of the impairment in the performance of IADLs in elderly women living with AD. (Nascimento, 2012)
Because people with dementia tend to have problems with memory and communication, the therapist may find it helpful to:
➢ Wear a name badge to identify yourself
➢ Use alternative wording to aid in understanding
➢ Minimize background noise and distractions during classes
➢ Demonstrate the exercises
➢ For patients with dyspraxia, copying the movement (mirroring) may be easier than following verbal instructions.
➢ Hands on guidance may be appropriate for some patients (Brown, 2015)
– Physical Therapy Contraindications
– Other Therapeutic Interventions
Non-pharmacological therapies such as light therapy, music therapy, and preferred music listening have shown promising results for managing behavioral problems that can be associated with dementia (Eggert, 2015).
Music listening has been associated with decreased levels of agitation and improved mood (Eggert, 2015).
Disruptive behavior, anxiety, cognition, depression and quality of life can all be improved through music therapy (Zhang 2017).
Music therapy, inexpensive and non-invasive, is a good choice for patients with dementia suffering from anxiety and disruptive behaviors (Zhang, 2017).
Nature images and music
➢ In a study done on the use of nature images and music, the authors hypothesized that they improve engagement and provide environmental positive distractions that alleviated some of the boredom, sensory deprivation, and disengagement found to be underlying determinants of disordered behaviors commonly seen in patients with dementia. (Eggert, 2015)
+++
VI. Consumer and Professional Resources
++
– Pubmed Health
– Community Resource Finder
– Alzheimer's Caregiver Corner
– Geriatric Examination Tool Kit
– Dementia Support Groups
– Alzheimer's Disease Website
– The Alzheimer Association
http://www.alz.org
This website offers information, a 24/7 helpline, caregiver resources, educational classes, and many other resources for patients, caregivers, and medical professionals.
– The Association for Frontotemporal Degeneration
– National Institute on Aging: Alzheimer's Disease Education and Referral Center
https://www.nia.nih.gov/alzheimers
This website offers great information about Alzheimer's disease and other dementias to patients, caregivers, and medical professionals. There is also a section that can help you find clinical trials that are near you.
– Dementia.org Healthcare Brands
+
Alzheimer's Association. "2011 Alzheimer's disease facts and figures". Alzheimer's & Dementia. 2012;7: 208–244.
+
Brown
D, Spanjers
K, Atherton
N, Lowe
J, Stonehewer
L, Bridle
C, Sheehan
B, Lamb
SE. Development of an exercise intervention to improve cognition in people with mild to moderate dementia: Dementia And Physical Activity (DAPA) Trial, registration ISRCTN32612072. Physiotherapy. 2015;101(2):126–134.
+
Buchner
DM. Exercise slows functional decline in nursing home residents with Alzheimer's disease. Aust J Physiother. 2007;53(3):204.
+
Covinsky
KE, Yaffe
K. Dementia, Prognosis, and the Needs of Patients and Caregivers. Ann Intern Med. 2004;140(7):573–574.
+
Eggert
J, Dye
CJ, Vincent
E, Parker
V, Daily
SB, Pham
H, Watson
AT, Summey
H, Roy
T. Effects of viewing a preferred nature image and hearing preferred music on engagement, agitation, and mental status in persons with dementia. SAGE Open Med. 2015;3:doi#2050312115602579.
+
Fadil
H, Borazanci
A, Haddou
EAB, Yahyaoui
M, Korniychuk
E, Jaffe
SL, Minagar
A. Early Onset Dementia. Int Rev Neurobiol. 2009;84:245–262.
+
Fraker
J, Kales
HC, Blazek
M, Kavanagh
J, Gitlin
LN. The role of the occupational therapist in the management of neuropsychiatric symptoms of dementia in clinical settings. Occup Ther Health Care. 2014;28(1):4–20.
+
Frances,
A. Diagnostic and Statistical Manual of Mental Disorders (DSM-IV-TR). 4th ed. Washington D.C.: American Psychiatric Association. 2000.
+
Fujishiro
H, Iseki
E, Nakamura
S, Kasanuki
K, Chiba
Y, Ota
K, Sato
K. Dementia with Lewy bodies: early diagnostic challenges. Psychogeriatrics. 2013;13(2):128–138.
+
Gauthier
S, Vellas
B, Farlow
M, Burn
D. Aggressive course of disease in dementia. Alzheimer's & Dementia. 2006; 2:210–217.
+
Geddes
J, Gelder
MG, Mayou
R. Psychiatry. Oxford [Oxfordshire]: Oxford University Press. 2005; pp.141.
+
Goldman
J, Holden
S. Treatment of psychosis and dementia in Parkinson's disease. Curr Treat Options Neurol. 2014;16(3):281.
+
Groot
C, Hooghiemstra
AM, Raijmakers
PGHM,
et al. The effect of physical activity on cognitive function in patients with dementia: A meta-analysis of randomized control trials. Ageing Res Rev. 2016;25:13–23.
+
Hamdy,
RC. Fact Sheets. South Med J. 2001;94(7):673–673.
+
High
KP, Valcour
V, Paul
R. HIV infection and dementia in older adults. Clin Infect Dis. 2006;42(10):1449–1454.
+
Jacova
C, Kertesz
A, Blair
M, Fisk
J, Feldman
H. Neuropsychological testing and assessment for dementia. Alzheimer's Dementia. 2007;3:299–317.
+
Jagust
W. Positron emission tomography and magnetic resonance imaging in the diagnosis and prediction of dementia. Alzheimer's Dementia. 2006;2(1):36–42.
+
Koepsell
T, Gill
D, Chen
B. Stability of clinical etiologic diagnosis in dementia and mild cognitive impairment: results from a multicenter longitudinal database. Am J Alzheimers Dis Other Demen. 2013;28(8):750–758.
+
Korczyn
AD, Vakhapova
V, Grinberg
LT. Vascular dementia. J Neurol Sci. 2012;322(1-2):2–10.
+
Ku
H, Lee
EK, Lee
KU, Lee
MY, Kwon
JW. Higher prevalence of dementia in patients with schizophrenia: A nationwide population-based study. Asia-Pacific Psychiatry. 2016;8(2):145–153.
+
Luzny
J, Holmerova
I, Wija
P, Ondrejka
I. Dementia Still Diagnosed Too Late - Data from the Czech Republic. Iranian Journal of Public Health. 2014;43(10):1436–1443.
+
McLaren
A, LaMantia
M, Callahan
C. Systematic review of non-pharmacologic interventions to delay functional decline in community-dwelling patients with dementia. Aging Ment Health. 2013;17(6):655–666.
+
Nascimento
C, Teixeira
CV, Gobbi
LT, Gobbi
S, Stella
F. A controlled clinical trial on the effects of exercise on neuropsychiatric disorders and instrumental activities in women with Alzheimer's disease. Braz J Phys Ther. 2012;16(3):197–204.
+
O'Brien
J, Thomas
A. Vascular dementia. The Lancet. 2015;386(10004):1698–1706.
+
Peavy
GM, Jacobson
MW, Goldstein
JL,
et al. Cognitive and functional decline in Huntington's disease: Dementia criteria revisited. Mov Disord. 2010;25(9):1163–1169.
+
Pressman
P, Miller
B. Diagnosis and Management of Behavioral Variant Frontotemporal Dementia. Biological psychiatry. 2014;75(7):574–81.
+
Prince
M, Bryce
R, Albenese
E, Wimo
A, Ribeiro
W, Ferri
CP. The global prevalence of dementia: a systematic review and metaanalysis. Alzheimer's Dementia; 2013;9(1):63–75.
+
Sacktor
N, Nakasujja
N, Skolasky
RL,
et al. HIV Subtype D Is Associated with Dementia, Compared with Subtype A, in Immunosuppressed Individuals at Risk of Cognitive Impairment in Kampala, Uganda. Clin Infect Dis. 2009;49(5):780–786.
+
Thal,
DR, Grinberg,
LT, Attems
J. Vascular dementia: Different forms of vessel disorders contribute to the development of dementia in the elderly brain. Exp Gerontol. 2012;47(11):816–824.
+
Tom
S, Hubbard
R, Crane
P, Haneuse
S, Bowen
J, McCormick
W, McCurry
S, Larson
E. Characterization of Dementia and Alzheimer's Disease in an Older Population: Updated Incidence and Life Expectancy With and Without Dementia. Am J Public Health. 2015;105(2):408–413.
+
Toots
A, Littbrand
H, Lindelöf
N,
et al. Effects of a High-Intensity Functional Exercise Program on Dependence in Activities of Daily Living and Balance in Older Adults with Dementia. J Am Geriatr Soc. 2016;64(1):55–64.
+
Turner,
MA, Moran,
NF, Kopelman,
MD. Subcortical dementia. Br J Psychiatry. 2002;180(2):148–151.
+
Wilkins
JM, Forester
BP. Update on SSRI Treatment for Neuropsychiatric Symptoms of Dementia. Curr Psychiatry Rep. 2016;18(2):1–7.
+
Wilson
RS, Bennett
DA, Gilley
DW, Beckett
LA, Schneider
JA, Evans
DA. Progression of parkinsonism and loss of cognitive function in Alzheimer disease. Arch Neurology. 2000;57(6):855–860.
+
Xu
X, Xiao
S, Rahardjo
T, Hogervorst
E. Risk Factors for Possible Dementia Using the Hopkins Verbal Learning Test and the Mini-Mental State Examination in Shanghai. Diagnostics. 2015;5(4):487–496.
+
Zhang
Y, Cai
J, An
L,
et al. Does music therapy enhance behavioral and cognitive function in elderly dementia patients? A systematic review and meta-analysis. Ageing Research Reviews. 2017;35:1–11.
+++
Developmental Coordination Disorder
++
Criteria that describe developmental coordination disorder (DCD) - from Diagnostic and Statistical Manual of Mental Disorders, 5th Edition (DSM-5) (American Psychiatric Association, 2013):
The acquisition and execution of coordinated motor skills is substantially below that expected given the individual's chronological age and opportunity for skill learning and use. Difficulties are manifested as clumsiness (e.g., dropping or bumping into objects) as well as slowness and inaccuracy of performance of motor skills (e.g., catching an object, using scissors or cutlery, handwriting, riding a bike, or participating in sports).
The motor skills deficit in Criterion A significantly and persistently interferes with activities of daily living appropriate to chronological age (e.g., self-care and self-maintenance) and impacts academic/school productivity, prevocational and vocational activities, leisure, and play.
Onset of symptoms is in the early developmental period.
The motor skills deficits are not better explained by intellectual disability (intellectual developmental disorders) or visual impairment and are not attributable to a neurological condition affecting movement (e.g., cerebral palsy, muscular dystrophy, degenerative disorder).
[Note: DSM-4 excluded children with diagnosis of ASD but DSM-5 omitted that exclusion so ASD may now be legitimate co-occurring condition.]
Co-occurring conditions: attention deficit hyperactivity disorder (ADHD), autism spectrum disorder (ASD), learning disabilities, anxiety and depression, obesity, and hypermobility of joints (Harris, 2015; Zoia, 2006;Zwicker, 2012; Zwicker, 2013a).
A comprehensive systematic review (1598 articles) determined more differences than similarities in behavioral profiles of individuals with ASD and DCD (Cacola, 2017)
Etiology not well understood; possibilities include:
Prenatal, perinatal or neonatal insult (Miyahara, 1995)
Appears to be more prevalent in children who were born before 37 weeks gestation, or who had low birth weight (<2500 grams) or who were born to parents of lower socioeconomic backgrounds (Goyen, 2009; Zwicker, 2013b; Zwicker, 2012; Lingam, 2009)
Incidence: effects approximately 5-6% of school-aged children (American Psychiatric Association, 2013); conflicting data reported on increased incidence in males
Pathogenesis
Possible areas of nervous system involvement are sub-cortical areas including cerebellum, basal ganglia, thalamus and posterior parietal cortex; likely diffuse damage (Cantin, 2007)
fMRI studies showed differences, including differences in activation and timing, in motor areas of the brain of children with DCD and those of age-matched peers (Zwicker, 2011; Debrabant, 2013)
Neuronal group selection theory (NGST) may explain symptoms of DCD and influence therapeutic intervention; NGST characterized by two phases of variability; both phases influenced by deficits in processing sensory information; limited primary (sub) cortical neuronal networks (Hadders-Algra, 2001).
++
Criteria established by DSM-5 definition of DCD (above) must be addressed/evaluated for diagnosis to be made.
Interdisciplinary or transdisciplinary team may include: teachers, occupational therapists, physical therapists, speech and language pathologists, psychologists, and physicians working in collaboration with child/parents.
Diagnosis based on patient history including achievement of motor milestones, motor coordination, sensory function and physical as well as neurological evaluation (Polatojko, 2005).
Motor ability a multi-faceted process; no one "best" evaluative measure exists; multiple sources recommended including standardized tests of motor development/proficiency and functional assessments that incorporate motor tasks performed in child's natural environment (Barnett, 2008).
Screening and evaluation measures commonly used:
Developmental Coordination Disorder Questionnaire (DCDQ2007) (Wilson, 2009; Wilson, 2012)
Little Developmental Coordination Disorder Questionnaire – version of DCDQ2007 parent questionnaire designed to screen younger children (ages 3 or 4 years) for coordination disorders; available on website (www.dcdq) for minimum fee
Movement Assessment Battery for Children, 2nd edition (MABC-2) (Henderson, 2007)
Three sections: (1) manual dexterity, (2) aiming and catching ball, (3) static and dynamic balance; each section contains items for each of 3 age bands (3-6 years, 7-10 years and 11-16 years)
Includes qualitative descriptors and behavioral checklist; does not assess handwriting
Test of Motor Impairment (Henderson, 1992) - to identify reaction times
Peabody Developmental Motor Scales 2 (Folio, 2000) – norm-referenced test to examine gross and fine motor development in children ages birth to 5 years
Bruinincks-Oseretsky Test of Motor Proficiency 2 (Bruinincks, 2007) – norm-referenced test (including a "short-form" screening component) for children ages 4-21 years; includes motor composites for (1) fine motor control, (2) manual coordination, (3) body coordination, and (4) strength and agility; does not assess quality of movement or handwriting.
++
Signs and Symptoms
Various aspects of the movement process may be involved: "…difficulty analyzing sensory information from the environment, using this information to choose a desired plan of action, sequencing the individual motor movements of the task, sending the right message to produce a coordinated action, or integrating all of these things in order to control the movement while it is happening" (Missiuna, 2003b).
Physical and emotional/behavioral characteristics adapted from summary by Missiuna (2003b)
Clumsy or awkward movements; may bump into, spill, or knock things over.
Delay acquiring motor skills such as tricycle or bike riding, ball catching, handling a knife and fork, and writing.
Discrepancy between motor abilities and abilities in other areas (e.g., intellectual and language skills strong while motor skills delayed).
Difficulty learning new motor skills; once learned, certain motor skills may be performed quite well while others may continue to be performed poorly.
More difficulty with activities that require constant changes in body position or adaptation to changes in the environment (e.g., baseball, tennis or jumping rope).
Difficulty with activities that require the coordinated use of both sides of the body (e.g., cutting with scissors, stride jumps, or swinging a bat).
Difficulty with handwriting; challenge is continually interpreting feedback about the movements of the hand while planning new movements; grip on pencil may be either too light or too firm.
Avoid, or give appearance of being disinterested in, activities which require a physical component.
Secondary emotional problems (e.g., frustration tolerance, low self-esteem, and lack of motivation)
Avoid socializing with peers, particularly on the playground; may seek out younger children to play with or go off on their own.
Dissatisfaction with performance (e.g., erases written work, complains of performance in motor activities, shows frustration with work product).
Resistant to changes in routine or in environment.
Additional symptoms:
Persistent influence of early tonic reflexes (e.g., asymmetrical tonic neck reflex or tonic labyrinthine reflex)
Hyper or hypo-responsiveness to sensory experiences
Soft neurologic signs
Difficulty learning feedforward control or using past movement experiences to predict movement requirements; greater end-point variability (Menz, 2013)
Slow, awkward, jerky movements and poor eye-hand or eye-foot coordination (Rivard, 2017)
Deficits in balance, postural stability, proprioception, and planning, timing, and sequencing movements (Barnhart, 2003; Rivard, 2017)
Difficulty controlling anterior-posterior sway in stance (Chen, 2015)
Use of abnormal strategies to accomplish tasks (Rivard, 2017)
Low muscle tone and weakness (Rivard, 2017)
Increased reaction time (Henderson, 1992; Rivard, 2017)
Frequent falls and difficulty maintaining balance (Barnhart, 2003; Rivard, 2017)
Impaired fine motor skills (writing, gripping, dressing, using utensils) (Rivard, 2012)
Problems learning a new task that requires coordination (Rivard, 2012)
Impaired predictive motor timing (Debrabant, 2013)
Difficulty performing two tasks at once; lack of automatization of motor actions when attentional demands are increased (Rivard, 2017)
Visual Problems - decreased ability to estimate an object's size and location in space; decreased visual attention and visual-motor integration (Visser, 2003)
Learning difficulties, especially motor-based academic tasks (Rivard, 2017)
Increased anxiety and depression; lower self-efficacy and competence in physical and social domains (Zwicker, 2013a)
Tend to avoid physical activity, both individual and organized sports (Caimey, 2005)
+++
III. Prognosis and Sequelae
++
The symptoms of DCD persist throughout adolescence and adulthood and behavioral/emotional symptoms (including depression and anxiety) may increase with time and negatively impact success in home, school and work environments, participation in everyday life, and perception of quality of life and life satisfaction (Barnhart, 2003; Pearsall Jones, 2008; Rivard, 2017; Zwicker, 2013a; Harris, 2015)
Difficulties with coordination may lead to decreased physical activity, participation in sports, weight gain and overall poor physical fitness (Caimey, 2005; Rivard, 2017).
+++
IV. Medical/Surgical Management
++
No surgical management; medical management for symptoms (e.g., ADHD), not underlying pathology
Methylphenidate can contribute to improvement in fine motor skills in children with DCD who also have a co-morbidity of ADHD, but further research is needed for how best to treat functional difficulties (Flapper, 2006; Smits-Engelsman, 2013).
Omega-3 and omega-6 fatty acids correlated with improvement in reading and spelling levels, as well as symptoms of ADHD, but not motor function (Richardson, 2005; Smits-Engelsman, 2013).
+++
V. Key Aspects of Therapeutic Management
++
In general, since children with DCD have difficulty transferring and generalizing skills it is important to provide feedback at appropriate time; provide opportunities to solve movement tasks; use varied instructional strategies (e.g., verbal, visual and imitation); and allow frequent practice in variable settings.
Two basic approaches to treating a child with DCD (Barnhart, 2003):
Bottom-up approaches focus on underlying deficits or impairments; examples include:
Sensory integration – therapeutic approach developed by AJ Ayres; involves child in variety of sensory experiences with emphasis on vestibular
Process-oriented treatment – based on specific kinesthetic training activities with inherent reward system
Perceptual motor training – eclectic approach with wide range of motor experiences and opportunities for practice
Neurodevelopmental treatment (NDT)
Top down approaches focus on having child develop cognitive or problem-solving skills to select and implement the most appropriate strategies for successful task performance; contextually based; intervention in every day situations; examples include:
Aquatic therapy (Vonder Hulls, 2006; Hillier, 2010)
Treatments such as "weight bearing exercises, writing, cognitive orientation to daily occupational performance (COOP), contemporary treatment approach (CTA), process oriented, traditional individual tutoring and motor imagery" showed limited evidence to determine effectiveness (Hillier, 2007)
Core stability exercise and task-oriented motor training both effective in improving motor proficiency in children ages 6-9 (face to face, once per week for eight weeks with home exercise encouraged); core stability exercise did not improve equilibrium (Au, 2014).
Neuromotor task training (NTT)- a child-centered, task-oriented program that focuses on the functional exercises that address the child's involved motor control processes including action planning and initiation (Niemeijer, 2007).
Teaching strategies used are important in improving movement performance and gross/fine motor skills; specifically providing clues on how to perform a task, asking children to cognitively process aspects of task, and providing children with explanation (Niemeijer, 2006).
Greater resemblance between functional skills of daily life and functional exercises implemented during therapy allows for greater transfer of the learned skills (Niemeijer, 2007).
Positive trends have been reported regarding the effect of participation in group therapy on self-esteem (McWilliams, 2005).
Strong relationship between decreased physical activity and low self-efficacy (Carney, 2005).
Investigation of 13-week group-based task-oriented invention for children aged 5-9 years offered in 3 delivery modes: (1) school environment by a school assistant (supported by a physical therapist), (2) school environment by a physical therapist, or (3) clinic environment by a physical therapist. Improvement noted in motor skills and self-perception in all 3 delivery modes (Ward, 2017).
Blocked practice of isolated, simple joint movements during high repetition low load strength training in a single case study with a 6 year 11 month old girl with ataxia, hypotonia and motor delays (Menz, 2013)
Consistent with premise that etiology may be explained by NGST, suggestion that early intervention aim at enlargement of primary neuronal networks with emphasis gradually shifting, as child ages, to provision of varied opportunities for active practice (Hadders-Algra, 2001).
Children with DCD may limit physical activity, influenced by generalized self-efficacy, and may benefit from structured encouragement to be physically active (Caimey, 2005); physical activity important as children have increased risk of being overweight or obese (Harris, 2015)
Clinical and laboratory measures to assess effectiveness of intervention; MABC-2 detected individual and group change; motion analysis laboratory data did not detect individual change (Larke, 2015)
Contraindications/Precautions
+
American Psychiatric Association (APA). Diagnostic and Statistical Manual of Mental Disorders. 5th edition. Washington, DC: American Psychiatric Association; 2013.
+
Au
MK, Chan
WM, Lee
L, Chen
TM, Chau
RM, Pang
MY. Core stability exercise is as effective as task-oriented motor training in improving motor proficiency in children with developmental coordination disorder: a randomized controlled pilot study. Clin Rehabil. 2014;28(10):992–1003.
+
Barnhart
RC, Davenport
MJ, Epps
SB, Nordquist
VM. Developmental coordination disorder. Phys Ther. 2003;83(8):722–731.
+
Barnett
AL. Motor assessment in developmental coordination disorder: from identification to intervention. Int J Disabil Dev Educ. 2008;55(2):113–129.
+
Bruininks
RH, Bruininks
BD. BOT 2: Bruininks-Oseretsky Test of Motor Proficiency, 2nd edition. San Antonio, TX: Pearson; 2005.
+
Cacola
P, Miller
HL, Williamson
PO. Behavioral comparisons in autism spectrum disorder and developmental coordination disorder: a systematic literature review. Res Autism Spect Disord. 2017;38:6–18.
+
Caimey
J, Hay
JA, Faught
BE, Wade
TJ, Corna
L, Flouris
A. Developmental coordination disorder, generalized self-efficacy toward physical activity, and participation in organized and free play activities. J Pediatr. 2005;147(4):515–520.
+
Cantin
N, Polatajko
H, Thach
W, Jaglal
S. Developmental coordination disorder: exploration of a cerebellar hypothesis. Hum Move Sci. 2007; 26:491–509.
+
Carney
J, Hay
J, Faught
B, Wace
T, Corna
L, Flouris
A. Developmental coordination disorder: generalized self-efficacy toward physical activity, and participation in organized and free play activities. J Pediatr. 2005;147(4):515–520.
+
Chen
FC, Tsai
CL, Stoffregen
TA, Wade
MG. Postural responses to a suprapostural visual task among children with and without developmental coordination disorder. Res Dev Disabil. 2011;32:1948–1956.
+
Chen
FC, Tsai
CL, Chang
WD, Li
YC, Chou
CL, Wu
SK. Postural control of anteroposterior and mediolateral sway in children with probable developmental coordination disorder. Pediatr Phys Ther. 2015;27(4):328–335.
+
Takon
I. Clinical use of a modified release methylphenidate in the treatment of childhood attention deficit hyperactivity disorder. Annals Gen Psych [serial online]. 2011;10(1):25–33.
+
Debrabant
J, Gheysen
F, Caeyenberghs
K, Van Waelvelde
H, Vingerhoets
G. Neural underpinnings of impaired predictive motor timing in children with developmental coordination disorder. Res Dev Disabil. 2013:34(5);1478–1487.
+
Flapper
BCT, Houwen
S, Schoemaker
MM. Fine motor skills and effects of methylphenidate in children with attention-deficit-disorder and developmental coordination disorder. Dev Med Child Neurol. 2006;48:165–169.
+
Folio
MR, Fewell
RR. Peabody Developmental Motor Scales: Examiner's Manual, 2nd edition. Austin, TX: Pro-Ed; 2000.
+
Goyen
T-A, Lui
K. Developmental coordination disorder in "apparently normal" school children born extremely preterm. Arch Dis Child. 2009;94:298–302.
+
Hadders-Algra
M. Early brain damage and the development of motor behavior in children: clues for therapeutic intervention? Neural Plastic. 2001; 8(1-2):31–49.
+
Harris
SR, Mickelson
E, Zwicker
JG. Diagnosis and management of developmental coordination disorder. Can Med Assoc J. 2015;187(9):659–665.
+
Henderson
L, Rose
P, Henderson
S. Reaction time and movement time in children with a developmental coordination disorder. J Child Psychol Psychiatry.1992;33(5):895–905.
+
Henderson
SE, Sugden
DA, Barnett
A. Movement Assessment Battery for Children:Manual. 2nd edition. San Antonio, TX: Pearson Assessment; 2007.
+
Hillier
S, McIntyre
A, Plummer
L. Aquatic physical therapy for children with developmental coordination disorder: a pilot randomized controlled trial. Phys Occup Ther Pediatr. 2010;30(2):111–124.
+
Hillier
S. Intervention for children with developmental coordination disorder. A systematic review. Internet J Allied Health Sci Pract. 2007;5 (3):1–11.
+
Larke
D, Campbell
A, Jensen
L, Straker
L. Responsiveness of clinical and laboratory measures to intervention effects in children with developmental coordination disorder. Pediatr Phys Ther. 2015;27(1):44–51.
+
Lingam
R, Hunt
L, Golding
J, Jongmans
M, Emond
A. Prevalence of developmental coordination disorder using DSM-IV at 7 years of Age: a UK population-based study. Pediatr. 2009;123:693–700.
+
McWilliams
S. Developmental coordination disorder and self-esteem: do occupational therapy groups have a positive effect? Brit J Occup Ther. 2005;68:393–400.
+
Menz
S, Hatten
K, Grant-Beuttle
M. Strength training for a child with suspected developmental coordination disorder. Pediatr Phys Ther. (2013):214–222.
+
Miyahara
M, Mobs
I. Developmental dyspraxia and developmental coordination disorder. Neuropsychol Rev. 1995; 5:245–268.
+
Missiuna
C, Rivard
L, Bartlett
D. Early identification and risk management of children with developmental coordination disorder. Pediatr Phys Ther, 2003a;15(1):32–38.
+
Niemeijer
AS, Schoemaker
MM, Smits-Engelsman
BCM. Are teaching principles associated with improved motor performance in children with developmental coordination disorder? A pilot study. Phys Ther. 2006;86(9):1221–30.
+
Niemeijer
AS, Smits-Engelsman
BCM, Schoemaker
MM. Neuromotor task training for children with developmental coordination disorder: a controlled trial. Dev Med Child Neurol. 2007;49:406–411.
+
Pearsall-Jones
JG, Piek
JP, Martin
NC, Rigoli
D, Levy
F, Hay
DA. A monozygotic twin design to investigate etiological factors for DCD and ADHD. J Pediatr Neurol. 2008;6(3):209–219.
+
Polatajko
HJ, Cantin
N. Developmental coordination disorder (dyspraxia): an overview of the state of the art. Semin Pediatr Neurol. 2005;12(4):250–8.
+
Richardson
AJ, Montgomery
P. The Oxford-Durham study: a randomized, controlled trial of dietary supplementation with fatty acids in children with developmental coordination disorder. Pediatr. 2005;115(5):1360–1366.
+
Rivard
L, Pollock
N, Sieman
J, Missiuna
C. Development coordination disorder. In: Palisano
RJ, Orlin
MN, Schreiber
J. Campbell's Physical Therapy for Children. 5th ed. St. Louis, MO: Elsevier; 2017: 398–422.
+
Smits-Engelsman
BCM, Blank
R, van der Kaay
AC,
et al. Efficacy of interventions to improve motor performance in children with developmental coordination disorder: a combined systematic review and meta-analysis. Dev Med Child Neurol. 2013;55(3):229–237.
+
Visser,
J. Developmental coordination disorder: a review of research on subtypes and co morbidities. Human Move Sci. 2003;22:479–493.
+
Vonder
Hulls DS, Walker
LK, Powell
JM. Clinicians' perceptions of the benefits of aquatic therapy for young children with autism: a preliminary study. Phys Occup Ther Pediatr. 2006;26(1-2):13–22.
+
Ward
EJ, Hillier
S, Raynor
A, Petkov
J. A range of service delivery modes for children with developmental coordination disorder are effective: a randomized controlled trial. Pediatr Phys Ther. 2017;29(3):230–236.
+
Wilson
BN, Crawford
SG, Green
D, Roberts
G, Aylott
A, Kaplan
BJ. Psychometric properties of the revised Developmental Coordination Disorder Questionnaire. Phys Occup Ther Pediatr. 2009;29:182–202.
+
Wilson
BN, Kaplan
BJ, Crawford
SG, Campbell
A, Dewey
D. Reliability and validity of a parent questionnaire on childhood motor skills. Am J Occup Ther. 2000;54:484–493.
+
Zoia
S, Barnett
A, Wilson
P, Hills
E. Developmental coordination disorder: current issues. Child Care Health Dev. 2006;32(6):613–618.
+
Zwicker
JG, Missiuna
C, Harris
SR, Boyd
LA. Brain activation associated with motor skill practice in children with developmental coordination disorder: an fMRI study. Int J Dev Neurosci. 2011;29(2):145–152.
+
Zwicker
JG, Harris
SR, Klassen
AF. Quality of life domains affected in children with developmental coordination disorder: a systematic review. Child Care Health Dev. 2013a;39:562–580.
+
Zwicker
JG, Yoon
SW, MacKay
M, Petrie-Thomas
J, Rogers
M, Synnes
AR. Perinatal and neonatal predictors of developmental coordination disorder in very low birthweight children. Arch Dis Child. 2013b;98(2):118–122.
+
Zwicker
JG, Missiuna
C, Harris
SR, Boyd
LA. Developmental coordination disorder: a review and update. Eur J Paediatr Neurol. 2012;16(6):573–581.
++
Down syndrome is a genetic condition occurring when an individual has a full or partial extra copy of chromosome 21.
Overall prevalence of Down syndrome is approximately 1 out of every 800-1200 live births. (Agarwal, 2014)
++
++
Having a child with Down syndrome has also been shown to have a positive correlation with an increase in maternal age. (Parker, 2010)
90% of the time the chromosomal error occurs at random during the formation of an egg and come from the mother. (Parker, 2010)
No behavioral activity of the parents or environmental factor is known to cause Down syndrome. (Parker, 2010)
++
++
++
++
Common physical features include brachycephaly with flat occiput; wide open fontanel; flat facial profile; flat nasal bridge; protruding tongue with small mouth; dysplastic, small, low set ears; upward slant of palpebral fissures, epicanthic folds; short and broad neck with excessive neck skin; short and broad hands; short and broad fingers with small middle phalanx of 5th finger (clinodactyly); simian crease; increased space between 1 and 2 toes (sandal gap); hypotonia; and hyper-extensibility/hyper-flexibility. (Agarwal, 2014)
Clinical features can be different across ethnicities (Africans, Asians, and Latin Americans), including brachycephaly, ear anomalies, clinodactyly, sandal gap, and abundant neck skin, which are all significantly less frequent in Africans. The angles at medial canthus and ala of the nose were the only common significant findings amongst different ethnicities. (Kruszka, 2017)
++
++
Antenatal Screening: In the first trimester of pregnancy, ultrasound to test for nuchal fold thickening, maternal age risk, maternal serum human chorionic gonadotropin (β-hCG) levels and pregnancy- associated plasma protein A (PAPP-A) levels are used for screening with an 80-82% detection rate. If the risk is greater than 1:250, prenatal diagnosis either by chorionic villi sampling at 11–12 weeks or amniocentesis at 16–18 weeks may be used to examine the fetal chromosomes. (Benn, 2013)
Postnatal Diagnosis: Presence of clinical features consistent with Down syndrome lead to a suspected diagnosis, however, the gold standard is chromosomal analysis. (Caine, 2005)
Key clinical diagnostic features are a distinctive physical appearance, poor growth and developmental delay. (Agarwal, 2014)
+++
III. Prognosis & Sequelae
++
++
++
++
All individuals with Down syndrome have some degree of intellectual disability with intelligence quotients that can range from mild to severe; the mean IQ is ~50. (Chapman, 2000)
Central nervous system abnormalities arise from a combination of abnormal development and functional changes that result from overexpression of genes in trisomic cells. Cerebellar development is also delayed and appears to terminate prematurely leading to a distinctly smaller cerebellum. (Haydar, 2012)
The early use of echocardiography has been shown to be effective in detecting congenital heart diseases including atrial septal defect, atrioventricular septal defect, ventricular septal defects, patent ductus arteriosus, and tetralogy of Fallot. (Ferencz, 1989) (Malec, 1999)
Children born with Down syndrome are more susceptible to structural gastrointestinal problems including intestinal atresias, stenosis and annular pancreas leading to small bowel obstructions. (Buchin, 1986)
Ophthalmological impairment including refractive errors (50 %), strabismus (20–47 %), cataracts (15 %), nystagmus (10 %), blepharitis (30 %), and retinal anomalies are common. (Ljubic, 2011)
Hearing impairment and otologic problems are common in children with DS and correlate with developmental delays. (Raut, 2011)
10–30 % of children with Down syndrome have atlantoaxial instability, which may be symptomatic in 1–2 %. (Hankinson, 2010) There is no evidence of benefit for routine cervical spine x-rays in asymptomatic children, however, in symptomatic children (neck pain, torticollis, radicular pain, frequent falls, change in bowel or bladder function, change in tone), a detailed neurological evaluation and cervical spine x-rays in neutral, flexion and extension are warranted. (Bull, 2011) Sports such as football, tumbling, or high impact sports should be avoided because of the risk of spinal cord injury.
There is an increased frequency of childhood leukemia, however, a reduced prevalence of adult onset cancers. (Wechsler, 2002)
Hypotonia and ligamentous laxity may contribute to musculoskeletal complications including patellar dislocation or instability, pes planus, scoliosis, and hip dislocation. (Merick, 2000; Caird, 2006)
Infants and children with Down syndrome display delayed motor development. (Kloze, 2016; Mazzone, 2004)
Adults with Down syndrome may develop secondary diagnoses including diabetes mellitus, hypothyroidism, obesity, cardiovascular disease, or Alzheimer's disease. (Henderson, 2007; McCarron, 2014)
+++
IV. Medical/Surgical Management
++
There is no known cure for Down syndrome.
Availability of appropriate, timely and specialized medical care from birth to adulthood is essential for effective diagnosis and management of the many potential sequelae. (Ivan, 2014; Ivan, 2014)
Regular monitoring of thyroid function, growth, vision, hearing, cardiac function, sleep related breathing disorders, and hematology is recommended from birth to school age. (Charlton, 2014)
+++
V. Key Aspects of therapeutic management
++
Possible Preferred Practice Pattern: Pattern 5B: Impaired Neuromotor Development
Standardized tests: Gross Motor Function Measure-88 (GMFM-88) (Palisano, 2001); Pediatric Balance Scale (PBS) (Franjoine, 2003); Developmental Behavior Checklist Child (DBC) (Einfeld, 1995); Functional Independence Measure for Children (WeeFIM) (Msall, 1994; Leonard, 2002)
Body structure and function impairments: psychomotor dysfunction (Pinter, 2001), hypotonicity, hyper flexibility of joints, underlying muscle weakness, delayed postural reactions, delayed reaction time, oppositional and defiant behaviors leading to depression in adolescence. (Tonge, 2003)
Activity and participation restrictions: delayed motor development in antigravity skills including sitting, standing, and walking. (Tudella, 2011)
When considering orthotics for children with Down syndrome, weight, height, leg length and hypermobility should be assessed and factor into the choice of orthotic. (Looper, 2012)
Regular exercise and fitness routines should be incorporated into the child and adult's daily life to reduce the risks of obesity and secondary complications. (Pitetti, 2013)
Initiation of early stimulation or intervention to improve overall developmental skills
When developing an intervention plan, balance and motor functions should be considered together. (Malak, 2015)
A comprehensive approach, including all disciplines, the family, and the child is essential to effective management across the lifespan.
+
Agarwal
Gupta N, Kabra
M. Diagnosis and management of Down syndrome. Indian J Pediatr. 2014;81(6):560–567.
+
Benn
P, Borell
A, Chiu
R, Cuckle
H, Dugoff
L, Faas
B,
et al. Position statement from the Aneuploidy Screening Committee on behalf of the Board of the International Society for Prenatal Diagnosis. Prenat Diagn. 2013;33:622–9.
+
Buchin
PJ, Levy
JS, Schullinger
JN. Down syndrome and the gastrointestinal tract. J Clin Gastroenterol. 1986;8(2):111–114.
+
Bull
MJ. Committee on genetics: health supervision for children with Down syndrome. Pediatrics. 2011;128:393–406.
+
Caine
A, Maltby
AE, Parkin
CA, Waters
JJ, Crolla
JA. Prenatal detection of Down syndrome by rapid aneuploidy testing for chromosomes 13, 18, and 21 by FISH or PCR without a full karyotype: a cytogenetic risk assessment. Lancet. 2005;366:123–128.
+
Caird
MS, Wills
BP, Dormans
JP. Down syndrome in children: the role of the orthopaedic surgeon. J Am Acad Orthop Surg. 2006;Oct;14(11):610–9.
+
Chapman
RS, Hesketh
LJ. Behavioral phenotype of individuals with Down syndrome. Ment Retard Dev Disabil Res Rev. 2000;6(2):84–95.
+
Charleton
PM, Dennis
J, Marder
E. Medical management of children with Down syndrome. Paediatr Child Health. 2014;24(8):362–369.
+
Ferencz
C, Neill
CA, Boughman
JA, Rubin
JD, Brenner
JI, Perry
LW. Congenital cardiovascular malformations associated with chromosome abnormalities: an epidemiologic study. J Pediatr. 1989;114(1):79–86.
+
Franjoine
MR, Gunther
JS, Taylor
MJ. Pediatric Balance Scale: a modified version of the Berg Balance Scale for the school-age child with mild to moderate motor impairment. Pediatr Phys Ther. 2003;15(2):114–28.
+
Einfeld
SL, Tonge
BT. The Developmental Behavior Checklist: the development and validation of an instrument to assess behavioral and emotional disturbance in children and adolescents with mental retardation. J Autism Dev Disord. 1995; 25:81–104.
+
Hankinson
TC, Anderson
RC. Craniovertebral junction abnormalities in Down syndrome. Neurosurgery. 2010;66:A32–8.
+
Haydar
TF, Reeves
RH. Trisomy and early brain development. Trends Neurosci. 2012;35(2):81–91.
+
Henderson
A, Lynch
SA, Wilkinson
S, Hunter
M. Adults with Down's syndrome: the prevalence of complications and health care in the community. Br J Gen Pract. 2007;57(534):50–55.
+
Ivan,
DL, Cromwell,
PF. Clinical practice guideline for management of children with Down syndrome: an update, Part one. J Pediatr Health Care. 2014;28(1):105–110.
+
Ivan,
DL, Cromwell,
PF. Clinical practice guideline for management of children with Down syndrome: an update, Part two. J Pediatr Health Care. 2014;28(3):280–284.
+
Kloze
A, Brzuszkiewicz-Kuzmicka
G, Czyzewski
P. Use of the TIMP in assessment of motor development of infants with Down syndrome. Pediatr Phys Ther. 2016;28(1):40–45.
+
Kruszka
P, Porras
AR, Sobering
AK,
et al. Down syndrome in diverse populations. Am J Med Genet Part A. 2017;173A:42–53.
+
Lautarescu
BA, Holland
AJ, Zaman
SH. The early presentation of dementia in people with Down syndrome: a systematic review of longitudinal studies. Neuropsychol Rev. 2017;27(1):31–45.
+
Leonard
S, Msall
M, Bower
C, Tremont
M, Leonard
H. Functional status of school-aged children with Down syndrome. J Paediatr Child Health. 2002;38:160–165.
+
Ljubic
A1, Trajkovski
V, Stankovic
B. Strabismus, refractive errors and nystagmus in children and young adults with Down syndrome. Ophthalmic Genet. 2011;32(4):204–211.
+
Looper
J, Benjamin
D, Nolan
M, Schumm
L. What to measure when determining orthotic needs in children with Down syndrome: a pilot study. Pediatr Phys Ther. 2012;24(4):313–319.
+
Malak
R, Kostiukow
A, Krawczyk-Wasielewska
A, Mojs
E, Samborski
W. Delays in motor development in children with Down syndrome. Med Sci Monit. 2015;21:1904–1910.
+
Malec
E, Mroczek
T, Pajak
J, Januszewska
K, Zdebska
E. Results of surgical treatment of congenital heart defects in children with Down's syndrome. Pediatr Cardiol. 1999;20:351–4.
+
Mazzone
L, Mugno
D, Mazzone
D. The general movements in children with Down syndrome. Early Hum Dev. 2004;79(2):119–130.
+
McCarron
M, McCallion
P, Reilly
E, Mulryan
N. A prospective 14-year longitudinal follow-up of dementia in persons with Down syndrome. J Intellect Disabil Res. 2014;Jan; 58(1):61–70.
+
Merrick
J, Ezra
E, Josef
B, Hendel
D, Steinberg
DM, Wientroub
S. Musculoskeletal problems in Down Syndrome European Paediatric Orthopaedic Society Survey: The Israeli sample. J Pediatr Orthop. 2000;9:185–92.
+
Msall
ME, DiGaudio
K, Rogers
BT,
et al. The Functional Independence Measure for Children (WeeFIM): conceptual basis and pilot use in children with developmental disabilities. Clin Pediatr. 1994;33:421–430.
+
Palisano
RJ, Walter
SD, Russell
DJ,
et al. Gross motor function of children with Down syndrome: creation of motor growth curves. Arch Phys Med Rehabil. 2001;82(4):494–500.
+
Parker
SE, Mai
CT, Canfield
MA,
et al. Updated national birth prevalence estimates for selected birth defects in the United States, 2004–2006. Birth Defects Res. Part A Clin Mol Teratol. 2010;88:1008–1016.
+
Pinter
JD, Eliez
S, Schmitt
JE, Capone
GT, Reiss
AL. Neuroanatomy of Down's syndrome: a high-resolution MRI study. Am J Psychiatry. 2001;158(10):1659–1665.
+
Pitetti
K, Baynard
T, Agiovlasitis
S. Children and adolescents with Down syndrome, physical fitness, and physical activity. J Sport Health Sci. 2013;2(1):47–57.
+
Raut
P, Sriram
B, Yeoh
A, Hee
KY, Lim
SB, Daniel
ML. High prevalence of hearing loss in Down syndrome at first year of life. Ann Acad Med Singapore. 2011;40(11):493–498.
+
Tonge
BJ, Einfeld
SL, Glidden
LM. Psychopathology and intellectual disability: the Australian child to adult longitudinal study. Int Rev Res Ment Retard. Vol 26. San Diego: Academic Press; 2003:64–66.
+
Tudella
E, Pereira
K, Basso
RP, Savelsbergh
GJ. Description of the motor development of 3-12 month old infants with Down syndrome: the influence of the postural body position. Res Dev Disabil. 2011;32(5):1514–1520.
+
Wechsler
J, Greene
M, McDevitt
MA, Anastasi
J, Karp
JE, Le Beau
MM, Crispino
JD. Acquired mutations in GATA1 in the megakaryoblastic leukemia of Down syndrome. Nat Genet. 2002;32(1):148–152.
++
Incidence and Prevalence:
Dystonia is the third most common movement disorder, after Parkinson's disease and tremor (Kernich, 2003)
15 to 30 per 100,000 population (Phukan, 2011)
Etiology:
Primary dystonia (most common; 2/3 of all cases)
No signs of structural abnormality in CNS (Berardelli, 1998)
Cause is idiopathic; genetic basis is suspected with at least 6 different genes involved (most common mutation is of the DYT1 gene and more recently discovered the CIZ1, G protein, GNAL, and ANO3 genes) (Schwarz, 2009; Jinnah, 2013; Dufke, 2014)
Average age of onset: 8 yrs
In children, symptoms often begin in foot, but in adults, the lower extremity is infrequently affected (Schneider, 2006)
Secondary dystonia
The result of injury or other brain illness with small areas of the brain (putamen and thalamus) damaged, scarring of the CNS, or abnormality in the hand area of the sensorimotor cortex (limb dystonia) (Jinnah, 2013)
While mainly associated with basal ganglia lesions, cerebellar lesions, alone and in combination with basal ganglia lesions, are also thought to be associated with secondary dystonias, based on the eye-blink conditioning paradigm and the cerebello-cortical interaction. (Avanzino, 2012; Antelmi, 2016)
Causes: drugs, medications (i.e. neuroleptics), infections, demyelinating lesions, degenerative disease, encephalitis, focal brain lesions, and acute injuries (traumatic brain injury) (Tarsy, 2006)
In children, secondary dystonic symptoms are often seen in conditions such as cerebral palsy, autism, neurometabolic, neuroinflammatory, and neurogenetic disorders (Lin, 2016)
Pathogenesis
Primary dystonia:
Descending pathways involving reciprocal inhibition of motor neurons is a possible site for idiopathic dystonia
Nerve conduction velocities show failure of neural activities preparing for movement
Secondary dystonia:
Chemical dysfunction or scarring within the striatum (caudate nucleus and putamen) results in overflow of motor cortex activity creating dystonic movements
Defects in ability to process GABA, dopamine, acetylcholine, norepinephrine, and serotonin may contribute to poor inhibition of motor control, and somatosensory may function abnormally (Hallett, 1995)
Pathophysiology
Distribution based classifications:
Focal dystonia: occurs in one specific body part
Often from repetitive use—musicians, writer's cramp.
Includes Cervical dystonia (most common), cranial dystonia, oromandibular dystonia, limb dystonia, spasmodic dysphonia, blepharospasm (Pont-Sunyer, 2010)
Primarily in adult women (Tarsy, 2006) and in Caucasians (Camargo, 2014)
Adult-onset focal dystonias (AOFDs) with age of onset: between 30 and 50 yrs) (Jinnah, 2013)
Earlier onset of symptoms typically occur in men (Camargo, 2014)
Sensory system abnormalities can occur before or after onset in involved and uninvolved body parts. (Conte, 2016)
Segmental dystonia: adjacent body parts or one body segment
Multifocal dystonia: affects non-adjacent body regions, i.e. Meige syndrome (Kernich, 2003)
Hemidystonia: one side of the body; associated with structural lesion (i.e. CVA) or degenerative disorders (i.e. MS) (Chuang, 2002)
Generalized dystonia: multiple regions of the body affected, onset is typically seen in a younger population (Berardelli, 1998; Kartha, 2006; Kernich, 2003)
Those with a family history of the disease have a higher tendency of earlier onset with faster spreading of dystonia compared to sporadic cases (Camargo, 2014)
++
Definition: Dystonia is a movement disorder that causes involuntary sustained muscle contractions, repetitive twisting movements, and abnormal postures of the trunk, neck, face, or arms and legs which may not be present when not involved in specific tasks. (Tarsy, 2006; Albanese, 2006; Phukan, 2011; Dufke, 2014)
Signs and Symptoms
ii. Symptoms can cause functional limitations and balance problems in many patients (Phukan, 2011)
iii. Early symptoms: deterioration in handwriting, foot cramps, neck may turn or pull involuntarily with fatigue; eyes blink rapidly and uncontrollably--"functionally blind"; tremor and speech trouble. (NINDS, 2013)
iv. Movements and Postures
"Action dystonia" or "task-specific dystonia": abnormal postures that occur during voluntary activity, sometimes task-specific (e.g. focal hand dystonia) and may disappear with other unrelated tasks (Tarsy, 2006; Phukan, 2011).
Movements may be rapid or slow, rhythmic and jerky, dystonic tremor (Kartha, 2006).
Sometimes involuntary movements may be present at rest. (Berardelli, 1998)
Overflow dystonia: movement in other parts of the body can cause episodes of dystonia in the primary location (Phukan, 2011)
v. Can be confused with spasticity or rigidity or may be mistakenly attributed to psychogenic causes. (Tarsy, 2006)
vi. Pain
Patients with two or three types of dystonic movements typically have higher severity and pain scores than those with a single presentation (Camargo, 2014)
Patients with a family history of cervical dystonia usually have lower pain scores than those with sporadic cases (Camargo, 2014)
vii. Dystonia usually worsens with stress, anxiety, fatigue, and overuse and is often improved with rest or relaxation. (Kartha, 2006; Phukan, 2011)
viii. Geste antagoniste ("sensory tricks") is a characteristic feature of dystonia in which light touch applied to the affected area may reduce the dystonic contractions/posture; bracing may also act as a "sensory trick" (Phukan, 2011; Schwarz, 2009)
ix. Weakness, atrophy, sensory loss, impaired perception, sphincter disturbances, significant psychological deformities, or alteration of electrical excitability are usually not associated with the disease; assists with differential diagnosis (Camargo, 2014)
Typically not associated with problems thinking or understanding, but anxiety and depression may be present (NINDS, 2013)
x. Higher rates of certain psychiatric disorders have been seen in patients with dystonia
Many studies have shown that the psychiatric disorders in dystonia often occur before any motor symptoms
Up to 91.4% of patients with cervical dystonia may have a current or lifetime psychiatric disorder as opposed to only 35% in the general population (Conte, 2016)
There is an increased prevalence rate of obsessive compulsive disorder (22.1%) and avoidant personality disorder (16.3%) (Lencer, 2009)
Patients with cervical dystonia have been shown to have up to a 53.4% increased risk of developing depression and up to an 83.3% increased risk of developing an anxiety disorder when compared to healthy controls
The severity of depression does not correlate with the severity of dystonia, which suggests that psychiatric disorders are not the patients' reaction to having a disability (Conte, 2016)
Specific Types (Jinnah, 2013):
Diagnostic testing
The diagnosis is strictly clinical (there is no diagnostic medical test) and is based primarily on abnormal postures and recognition of specific features. Expert observation and the use of a structured flow chart are recommended when diagnosing dystonia (Albanese, 2011)
History and examination rule out secondary dystonia: history of trauma, illness, and medications; onset and behavior of symptoms (Kernich, 2003).
EMG studies show sustained simultaneous contractions of agonist and antagonist muscles
MRI used when other neurological findings are present (decreased activity of the internal segment of the globus pallidus is common) (Berardelli, 1998).
Lab tests look for an underlying structural, degenerative, or metabolic disorder (Tarsy, 2006).
Differential diagnoses: Cerebral palsy, dry eyes, myoclonus, and cervical muscle strain, Wilson's disease, and Dopa-responsive dystonia (Tarsy, 2006; Kartha, 2006).
Scales used to classify severity:
Frequency of abnormal movements (FAM) scale: used to evaluate musician's dystonia. (Spector, 2005), Unified Dystonia Rating Scale, Fahn-Marsden Scale, Global Dystonia Rating Scale, Cervical Dystonia Impact Profile (Cano, 2004)
Diagnosis is still mainly clinical but genetic analysis and counseling is being requested more.
Only available for forms of early-onset primary torsional dystonias (PTD) in primary dystonias
Recommended for any PTD case with onset before 26 years and in later onset for those who have an affected relative with early-onset. (Petuccti, 2013)
Genetic testing (blood sample): test for DYT-1 gene which has been identified for primary dystonias (Albanese, 2006; Kernich, 2003)
Sequencing of other known PTD genes should be considered in patients with predominant cranial-cervical and laryngeal involvement. (Petrucci, 2013)
++
Age of onset is best predictor of prognosis
If dystonia starts early in childhood and is present in family members, it will tend to get progressively worse with age
If dystonia starts in childhood and is secondary to cerebral palsy or other brain injury at birth, dystonia tends to remain static
Dystonia is often described as early or childhood onset versus adult onset (NINDS, 2013)
Spontaneous remission occurs in 30% of cases within the 1st year; majority show steady progression with maximum disability occurring after 5 yrs.
Symptoms can progress, eventually leading to sustained fixed skeletal deformities (FSDs) such as scoliosis (Camargo, 2014) and lateral neck shift (Esposito, 2016).
Progression: Initially, dystonic movements are mild and intermittent appearing with volitional movements or stress. Later, may show dystonic postures and movements while walking and later progressing to when they are relaxed. (NINDS, 2013)
The time that it takes for initial symptoms to progress in severity or spread location can vary from days to years (Schwarz, 2009)
Blepharospasm has been proven to progress the furthest and fastest; patients experienced a spread of symptoms to a second location in an average of 1.2 years (Svetel, 2007)
Expected sequlae: May eventually lead to abnormal posturing, shortened tendons (physical deformation), fatigue, weakness, and increased risk of lower urinary tract infection. (NINDS, 2013; Batla, 2016)
+++
IV. Medical/surgical Management
++
Pharmaceuticals
Except for dopa-responsive dystonia, there is no specific pharmacologic treatment for dystonia. (Tarsy, 2006)
Some medications may improve symptoms: Botulinum toxin, anticholinergic drugs, antiepileptic drugs, muscle relaxants (benzodiazepines), dopaminergic drugs (used for early-onset patients) (Albanese, 2006; Kartha, 2006)
Botulinum toxin: improved effects have been found when patients receive physical therapy with the injections (Jankovic, 2006). Botulinum toxin is the first-line treatment of focal dystonia. Only four approved types of botulinum toxins: three BoNT/A and one BoNT/B (Truong, 2012).
Improvement in symptoms after botulinum toxin injections is usually temporary (average of 14.5 weeks) so repeated treatments are necessary and a need for higher doses over time. Long-term, approved botulinum toxins are safe and effective. (Jankovic, 2006; Jinnah, 2013; Truong, 2012)
To reduce the chance of lateral shift in cervical dystonia, avoid excessive doses of Botulinum toxin in one side of the neck as opposed to the other side (Esposito, 2016)
Intrathecal baclofen is also effective in treating dystonia but has adverse events that sometimes occur (Motta, 2014)
25% of patients experienced complications: 9.3% infection, 4.9% CSF leak, 15.1% problem with catheter, and 1% problem related to pump (Motta, 2014)
Surgical techniques-only used in severe cases after failure of other forms of medical management (Kartha, 2006)
Selective peripheral denervation of muscle (inconsistent benefit)
Brain lesioning (pallidotomy has shown best results)
Deep-brain stimulation (DBS) of the globus pallidus for treatment of medically refractory and primary dystonia. (Tarsy, 2006; Dashtipour, 2007; Cersosimo, 2008)
Chronic DBS of the globus pallidus internus is a safe and effective treatment for advanced, disabling dystonia (Isaias, 2008) with the possible exception of axial FSDs (Isaias, 2008)
Primary dystonias respond better to DBS (Vercueil, 2002) with the possible exception of axial FSDs (Isaias, 2008)
DBS of GPi is considered the initial treatment for children with primary generalized dystonia to reduce joint deformity and loss of mobility. (Bragg, 2014)
DBS can cause improvements in the following symptoms: reduction of dystonic movements, abnormal posturing, and pain, which can improve the patient's quality of life, functional capacity, and reduce the need for medications (Schwarz, 2009)
Patients with disease duration shorter than 15 years may expect to improve faster and achieve a better general outcome than those with longer disease duration (Isaias, 2008)
Deep-brain stimulation (DBS) of the subthalamic nucleus to treat medically refractory isolated dystonia (Ostrem, 2017)
Long term DBS of the subthalamic nucleus is a safe and effective alternative to DBS of the globus pallidus
Some patients receiving DBS of the globus pallidus experience minimal results or negative postoperative gait and fine motor changes due to the stimulation
DBS of the subthalamic nucleus can cause very similar improvements as DBS of the globus pallidus with the same surgical risks.
+++
V. Implications for Therapeutic Management
++
Preferred practice patterns (APTA, 2017)
5C: Impaired Motor Function and Sensory Integrity Associated with Nonprogressive Disorders of the Central Nervous System – Congenital Origin or Acquired in Infancy or Childhood
5D: Impaired Motor Function and Sensory Integrity Associated with Nonprogressive Disorders of the Central Nervous System – Acquired in Adolescence or Adulthood
5E: Impaired Motor Function and Sensory Integrity Associated with Progressive Disorders of the Central Nervous System
Role of the PT:
Regaining (and/or maintaining) the patient's level of function.
Treat symptoms of dystonia. Maintain ROM, posture, strength, enhance motor control, assess for AD and/or splinting, and prevent contractures. (Bragg, 2014)
Teach patients how to improve their ability to perform ADLs in their own environment.(van den Dool, 2013)
Patient education and health promotion
Explain general information regarding dystonia (signs/symptoms, expected progression according to each patient's current status, treatment options).
Instruct patient regarding the importance of adhering to the HEP and activity modifications: recognize and reduce compensations, make the pt aware of aggravating activities (Jabusch, 2005)
Relaxation (Feldenkrais) and stress management techniques.
Teach patient self-stretches/strengthening exercises. Modified pen grip in combination with handwriting training (Bauer, 2009)
General PT options
Treatment to reduce spasticity and contractures:
Muscle stretching and strengthening, postural training (precaution: overstretching or excessive exercise can cause exacerbation of symptoms). (Hakkinen, 2004)
Manual therapy techniques to improve soft tissue extensibility, promote muscle relaxation, and decrease pain; precaution: do not overstretch the tissues (Tassorelli, 2006; Crowner, 2007)
Modalities: EMG biofeedback, electrical stimulation to break up the pain-spasm-pain cycle
Casting, splinting, mechanical assistive devices, and orthoses to reduce joint deformity and disability; precaution: long term immobilization can cause muscle weakness and skin integrity may be compromised in braces (Okun, 2002; Bragg, 2014).
Treatment to improve gait
Educate patient about proper use of assistive device, balance exercises to increase stability, fall prevention, and safety techniques.
Sensory training and limb-immobilization techniques have been the treatment of choice for limb dystonia but have not always proven beneficial. (Tarsy, 2006).
Practice with alternative sensory feedback and novel sensory experience (e.g. watching mirror reflection of movement of contralateral limb, variety of sensory experience from terry cloth to sandpaper, feeling Braille or alphabet noodles).
Mental visualization of hand movement has been used to treat focal hand dystonia. (Byl, 2003)
Health care team contributions: Work with physician and pharmacist for medical management; occupational therapists, speech therapists (swallowing, language skills), and psychologists (coping with the emotional aspect of the disease) are all possible contributors.
+++
VI. Consumer and Personal Resources
++
Organizations
Dystonia Medical Research Foundation: publications, i.e. Dystonia Dialogue magazine, events are planned in order to raise funds for future dystonia research; http://www.dystonia-foundation.org/
Dystonia Advocacy Network (DAN): organization that advocates for new legislation for the dystonia community; http://dystonia-advocacy.org/
American Dystonia Society (ADS): promotes awareness of the disorder and raises funds for research, online peer support and outreach forum; http://www.dystonia.us/index.htm
The Bachmann-Strauss Dystonia and Parkinson Foundation, Inc.: foundation that promotes new research and awareness, events and publications (i.e. the Outlook newsletter); http://www.dystonia-parkinsons.org/
Action for Dystonia, Diagnosis, Education and Research (ADDER): foundation that advances the education of the public, promotes research and treatment of dystonia, offer support and welfare to patients, their care givers and families, [email protected] www.actionfordystonia.co.uk
Dystonia Coalition: associated with the Rare Clinical Diseases Research Network; consists of medical researchers and patient advocacy groups that work to advance research in dystonias to find better treatments and a cure; www.rarediseasesnetwork.org/Dystonia
National Institute of Neurological Disorders and Stroke: provides an overview of dystonia and resources for publications, organizations, news, and research literature. http://www.ninds.nih.gov/disorders/dystonias/dystonias.htm
National Spasmodic Torticollis Association: provides resources for and supports the needs of patients with cervical dystonia and their families, promotes awareness and education to the public, and aims to promote research for new treatments for cervical dystonia; http://www.torticollis.org/
Resources
WE MOVE: educational resources, information about movement disorders, current treatment options and studies; http://www.wemove.org/dys/
Support groups
Focal Dystonia Exchange: online community network for people with dystonia, as well as for caregivers, family, and friends; patient stories, patient tools, and networking; http://www.focaldystoniaexchange.com/
+
Albanese
A, Asmus
F, Bhatia
KP,
et al. EFNS guidelines on diagnosis and treatment of primary dystonias. Eur J Neurol. 2011;18(1):5–18.
+
Albanese
A, Barnes
MP, Bhatia
KP. A systematic review on the diagnosis and treatment of primary (idiopathic) dystonia and dystonia plus syndromes: report of an EFNS/MDS-ES Task Force. Eur J Neurol. 2006;13:433–444.
+
Antelmi
E, Di Stasio
F, Rocchi
L,
et al. Impaired eye blink classical conditioning distinguishes dystonic patients with and without tremor. Parkinsonism Relat Disord. 2016;31:23–27.
+
Avanzino
L, Abbruzzese
G. How does the cerebellum contribute to the pathophysiology of dystonia? Basal Ganglia. 2012;2(4):231–235.
+
Batla
A, Pareés
I, Edwards
MJ, Stamelou
M, Bhatia
KP, Panicker
JN. Lower urinary tract dysfunction in patients with functional movement disorders. J Neurol Sci. 2016;361:192–194.
+
Bauer
B, Furholzer
W, Jasper
I, Marquardt
C, Hermsdorfer
J. Effects of modified pen grip and handwriting training on writer's cramp. Arch Phys Med Rehabil. 2009;90(5):867–75.
+
Berardelli
A, Rothwell
J, Hallett
M, Thompson
P, Manfredi
M, Marsden
D. The pathophysiology of primary dystonia. Brain. 1998;121:1195–1212.
+
Bragg
DC, Sharma
N. Update on Treatments for Dystonia. Curr Neurol Neurosci Rep. 2014;14(6):454.
+
Byl
NN, Nagajaran
S, McKenzie
AL. Effects of sensory discrimination training on structure and function with focal hand dystonia. Arch Phys Med Rehabil. 2003;84:1505–1514.
+
Camargo
CHF, Camargos
ST, Becker
N, Munhoz
RP, Raskin
S, Cardoso
FEC, Teive
HAG. Cervical dystonia: about familial and sporadic cases in 88 patients. Arquivos de Neuro-Psiquiatria. 2014;72(2):107–113.
+
Camargo
CHF, Teive
HAG. Evolution of the concept of dystonia. Arquivos de Neuro-Psiquiatria. 2014;72(7):559–561.
+
Cano
S, Hobart
J, Fitzpatrick
R, Bhatia
K, Thompson
A, Warner
T. Patient-based outcomes of cervical dystonia: a review of rating scales. Mov Disord. 2004;19(9):1054–1059.
+
Cersosimo
MG, Raina
GB, Piedimonte
F, Antico
J, Graff
P, Micheli
FE. Pallidal surgery for the treatment of primary generalized dystonia: long-term follow-up. Clin Neurol Neurosurg. 2008;110:145–150.
+
Chuang
C, Fahn
S, Frucht
S. The natural history and treatment of acquired hemidystonia: report of 33 cases and review of the literature. J Neurol Neurosurg Psychiatry. 2002;72(1):59–67.
+
Conte
A, Berardelli
I, Ferrazzano
G, Pasquini
M, Berardelli
A, Fabbrini
G. Non-motor symptoms in patients with adult-onset focal dystonia: Sensory and psychiatric disturbances. Parkinsonism Relat Disord. 2016;22:S111–S114.
+
Crowner
B. Cervical dystonia: disease profile and clinical management. Phys Ther. 2007;87(11):1511–1526.
+
Dashtipour
K, Barahimi
M, Karkar
S. Cervical dystonia. J Pharm Pract. 2007;20:449–457.
+
Dufke
C, Strurm
M, Schroeder
C, Moll
S, Ott
T, Riess
O, Bauer
P, Grundmann
K. Screening of Mutations in GNAL in Sproadic Dystonia Patients. Mov Disord. 2014;29(9):1193–6.
+
Esposito
M, Peluso
S, Dubbioso
R,
et al. The occurrence of lateral shift in cervical dystonia. Neurol Sci. January 2017;88(1):1–4.
+
Häkkinen
A, Ylinen
J, Rinta-Keturi
M, Talvitie
U, Kautiainen
H, Rissanen
A. Decreased neck muscle strength is highly associated with pain in cervical dystonia patients treated with botulinum toxin injections. Arch Phys Med Rehabil. 2004;85(10):1684–1688.
+
Hallett
M. Is dystonia a sensory disorder? Ann Neurol. 1995;38(2):139–140.
+
Isaias
IU, Alterman
RL, Tagliati
M. Outcome predictors of pallidal stimulation in patients with primary dystonia: the role of disease duration. Brain. 2008;131(7):1895–1902.
+
Jabusch
H, Zschucke
D, Schmidt
A, Schuele
S, Altenmüller
E. Focal dystonia in musicians: treatment strategies and long-term outcome in 144 patients. Mov Disord. 2005;20(12):1623–1626.
+
Jankovic
J. Treatment of dystonia. Lancet Neurol. 2006;5(10):864–872.
+
Jinnah
H. A, Berardelli
A, Comella
C, DeFazio
G, DeLong
M, Factor
S, Galpern
W, Hallett
M, Ludlow
C, Perlmutter
J, Rosen
A. The Focal Dystonias: Current View and Challenges for Future Research. Mov Disord. 2013;28:926–943.
+
Kartha
N. Dystonia. Clin Geriatr Med. 2006;22:899–914.
+
Kernich
C. Patient family fact sheet. Dystonia. Neurologist. 2003;9(2):121–122.
+
Lencer
R, Steinlechner
S, Stahlberg
J,
et al. Primary focal dystonia: evidence for distinct neuropsychiatric and personality profiles. J Neurol Neurosurg Psychiatry. 2009;80(10):1176–1179.
+
Lin
J-P, Nardocci
N. Recognizing the Common Origins of Dystonia and the Development of Human Movement: A Manifesto of Unmet Needs in Isolated Childhood Dystonias. Front Neurol. 2016;7:226. doi:10.3389/fneur.2016.00226.
+
Motta
F, Antonello
C. Analysis of complication in 430 consecutive pediatric patients treated with intrathecal baclofen therapy: 14-year experience. J Neurosurg Pediatr. 2014;13(3):301–306.
+
Okun
M, Nadeau
S, Rossi
F, Triggs
W. Immobilization dystonia. J Neurol Sci. 2002;201(1):79–83.
+
Ostrem
JL, San Luciano
M, Dodenhoff
KA,
et al. Subthalamic nucleus deep brain stimulation in isolated dystonia: A 3-year follow-up study. Neurology. 2017;88(1):25–35.
+
Petrucci
S, Valente
EM. Genetic issues in the diagnosis of dystonias. Frontiers in Movement Disorders. 2013;4:1–7.
+
Phukan
J, Albanese
A, Gasser
T, Warner
T. Primary dystonia and dystonia-plus syndromes: clinical characteristics, diagnosis, and pathogenesis. Lancet Neurol. 2011;10(12):1074–1085.
+
Pont-Sunyer
C, Martí
M, Tolosa
E. Focal limb dystonia. Eur J Neurol. 2010;17:22–27.
+
Ramdharry
G. Case Report: Physiotherapy cuts the dose of botulinum toxin. Physiother Res Int. 2006;11:117–122.
+
Schneider
SA, Edwards
MJ, Grill
SE, Goldstein
S, Kanchana
S, Quinn
N, Bhatia
K, Hallett
M, Reich
S. Adult-Onset Primary Lower Limb Dystonia. Mov Disord. 2006;21:767–771.
+
Schwarz
C, Bressman
S. Genetics and treatment of dystonia. Neurol Clin. 2009;27(3):697.
+
Spector
JT, Brandfonbrener
AG. A new method for quantification of musician's dystonia: the frequency of abnormal movements scale. Med Probl Perform Art. 2005;20:152–162.
+
Svetel
M, Pekmezović
T, Kostić
V. Spread of primary dystonia in relation to initially affected region. J Neurol. 2007;254(7):879–883.
+
Tarsy
D, Simon
D. Current Concepts: Dystonia. N Engl J Med. 2006;355:818–829.
+
Tassorelli
C, Mancini
F, Martignoni
E, . Botulinum toxin and neuromotor rehabilitation: An integrated approach to idiopathic cervical dystonia. Mov Disord. 2006;21(12):2240–2243.
+
Truong
D. Botulinum toxins in the treatment of primary focal dystonias. J Neurol Sci. 2012;316(1-2):9–14.
+
Van den Dool
J, Visser
B, Koelman
TM, Engelbert
R, Tijssen
M. Cervical dystonia: effectiveness of a standardized physical therapy program; study design and protocol of a single blind randomized controlled trial. BMC Neurology. 2013;13:1–8.
+
Vercueil
L, Krack
P, Pollak
P. Results of deep brain stimulation for dystonia: a critical reappraisal. Mov Disord. 2002;17 Suppl 3:S89–S93.
++
Definition:
Inflammation and damage to cells primarily in the gray matter of the Central Nervous System (CNS) that may result from infection with any number of viruses and present as an alteration in consciousness, fever, headache, seizure, and/or focal neurologic signs (Ringold, 2005; NIH, 2007; Glaser, 2006)
For cases of infectious encephalitis, viral pathogens make up roughly 61% of all encephalitis cases (Glaser, 2006; Rodrigo 2016) followed by bacteria, parasites, prions, and fungi (Glaser, 2006). Noninfectious encephalitis makes up only 3.4% of all cases (Rodrigo, 2016).
Encephalitis is most commonly caused by viruses which lead to cerebral cortex inflammation via two mechanisms:
Primary encephalitis (acute viral)
Caused by direct viral infection of the brain or spinal cord (NIH, 2007)
May be a reactivation of a virus that had previously been latent after previous illness
Secondary (post-infective)
Infection first occurs elsewhere in the body and then travels to the brain (NIH, 2007).
Instead of solely attacking the infection-causing cells, the immune system mistakenly attacks healthy brain cells.
Can be due to:
Bacterial causes- such as bacterial meningitis or Lyme's disease, tuberculosis, cat scratch fever, typhoid fever (Polhill, 2007)
Complications of other infectious diseases- such as measles, mumps, hepatitis A, rabies, syphilis, and chicken pox.
Herpes Simplex virus Encephalitis (HSE) type I is the most common cause of fatal encephalitis in the United States (about 10% of all cases) and of infantile encephalitis (about 50% of all infants that contract HSE will develop CNS symptoms) (Ringold, 2005; NIH, 2007).
Typical patients will be either younger than 20 yo or older than 50 yo (NINDS, 2004).
HSE typically involves frontotemporal changes and lesions in limbic system (Tie, 2001), can affect both hemispheres simultaneously (Kennedy, 2013), and may rarely target the brainstem (Kennedy, 2013).
A rare form of relapsing HSE exists that is autoimmune in nature with N-methyl-D-aspartate (NMDA) receptor antibodies present in CSF (Graus, 2016).
Arboviruses (arthropod-borne viruses) such as:
Tick borne encephalitis (TBE) virus (sometimes referred to as Powassan virus)
The most common arthropod-transmitted viral infection of humans in Europe and central and eastern Asia, hospitalizing >1000 patients each year (Heinz, 2013)
Humans are dead-end hosts only and do not play any role in the maintenance of TBE in nature. (Heinz, 2013)
The European TBE virus subtype may cause asymptomatic infection, flu-like disease or biphasic disease with central nervous system involvement in the later phase. It can then present as meningitis of varying severity, from self-limited to life-threatening (Grygorczuk, 2015).
Japanese Encephalitis virus (JEV) (Kennedy, 2004; NIH, 2007; Alatoom,2009) is most prevalent in Southeast Asia and the Far East but spreading into previously non-endemic areas, and pigs and wild birds are the natural reservoirs (Shi, 2014)
Involves thalamic hemorrhages (Tie, 2001)
JEV is a major public health problem because of large endemic areas in the country, the high case fatality rate (20-30%) and frequent residual neuropsychiatric damage (50-70%). (Sharma, 2012)
Eastern equine encephalitis: associated with progressive basal ganglia and brain stem lesions (Polhill, 2007)
Western equine encephalitis: associated with a lower mortality but is more severe in infants/children
Autoimmune Encephalitis is a relatively new differential diagnosis within encephalopathies with an impact on disease diagnosis and treatment (Lancaster, 2011). It involves antibodies against a variety of CNS proteins, some of which are critical for synaptic transmission such as the NMDA receptor, GABA receptor, and AMPA receptor (Gastaldi, 2016; Lancaster, 2011). Certain clinical signs can be clues for the type of antibodies being produced, such as the NMDA receptor antibodies which are associated with psychologic symptoms (Lancaster, 2015)
St. Louis Encephalitis Virus and West Nile Virus → the most recent causes of epidemics in the US. HHV-6 is a possible pathogen in a subset of encephalitis cases.(Yao, 2009) If a peripheral infection is able to defeat the body's immune system, that infection can travel into cerebrospinal fluid, causing inflammation which can be detrimental to nerves and vessels travelling to the brain (NINDS, 2004)
Roughly 1 in 150 individuals infected with West Nile Virus will develop encephalitis, meningitis, or a combination of both (Rath, 2012).
Due to swelling of the brain, increased intracranial pressure is exerted on the brain stem pushing the brain stem downward. Resulting herniation will impair or terminate brain stem function.
Areas other than the cerebral cortex that can be inflamed include: brain stem, cerebellum, anterior horns of spinal cord, dorsal root of the spinal cord, white matter, sympathetic ganglia (Davis, 2006).
Incidence
In Western industrialized countries such as the United States, the incidence of encephalitis in children is ~12 per 100,000 and near 2 per 100,000 in adults (Jmor, 2008)
Several thousand cases are reported each year, but many more may occur with mild-moderate flu-like symptoms (NIH, 2007).
Climate change and increasing international travel raises the possibility of a wider geographic spread of microbes, which may have important public health implications (Granerod, 2013)
A higher incidence (adjusted for year, age, and region) of encephalitis observed among male patients, consistent with previous studies, and patients <1 and > 65 years of age, perhaps related to hyporesponsive immune system in early life and later immunosenescene. (Granerod, 2013)
Toxoplasmic encephalitis and West Nile Virus were each approximately two-fold more prevalent in males than females in the U.S. (George, 2014).
Strong evidence that the incidence of encephalitis is higher than that previously estimated in England with some models indicating it could be as high as 8.66 cases/100,000/year (Granerod, 2013)
Herpes simplex virus has an incidence of 1-3/million in western countries (Sharma, 2012)
In children, anti-NMDA (N-methyl D-aspartate) encephalitis was more common in warm months, but tumor-related incidence of encephalitis was more frequent during cold months (Adang, 2014).
Due to advances in medicine, there is an increase in the population of immunocompromised patients resulting from HIV (Human Immunodeficiency Virus) infections, transplant successes, and weakening cancer treatments. This has resulted in an increase in the incidence of opportunistic viral infections such as encephalitis. (Kneen, 2008)
Between January 1, 2000 and December 31, 2010, nearly 250,000 patients were admitted to acute inpatient hospital care in the United States with a diagnosis of encephalitis. Approximately 53% of encephalitis patients were female and the average age was 44.8 years. Among all encephalitis admissions, 7.7% were found to have comorbid HIV/AIDS (Acquired Immune Deficiency Syndrome) while 6.8% had concurrent cancer (George, 2014).
Risk Factors
Non-modifiable:
Anyone with a compromised immune system is at higher risk
A genetic mutation (single nucleotide polymorphism) in OAS1 has been linked as a risk factor for progression of West Nile virus to West Nile encephalitis (Bingham, 2011)
Modifiable:
Human exposure to ticks through increased time spent in wooded areas (Randolph, 2010)
Travel to endemic areas, especially for Japanese encephalitis (Hills, 2010)
A correlation was found between dense vegetation and increased occurrence of infections in regards to the geographical grouping of Saint Louis encephalitis (Rotella, 2011)
++
++
++
++
++
++
++
Computed Tomography (CT) scan is used most often - may show an area of bleeding which occurs in patients with HSE type I.
Magnetic Resonance Imaging (MRI) on T2-weighted, fluid-attenuated inversion recoveries (FLAIR) (Cole, 2007) are used along with CT scans to determine the extent and location of swelling (usually done before a spinal tap).
Encephalitis typically involves the temporal lobe and is best visualized by (MRI) of the brain. (Sili, 2014).
MRI is more sensitive and specific than CT for the diagnosis of HSE. Brain MRI had compatible findings in 95% of cases, while brain CT was positive in only half of the cases tested (Sili, 2014).
HSE has characteristic abnormalities of edema and/or abnormal enhancement in one or both temporal and frontal lobes, the insular cortex and the angular gyrus with relative sparing of the basal ganglia (Kennedy, 2013; Rath, 2012). MRIs of HIV Encephalitis indicate volume loss and periventricular and deep white matter abnormality (Rath, 2012).
Electroencephalogram (EEG) abnormality may show sharp waves in one or more temporal lobes. However, a normal EEG does not rule out encephalitis.
Normal early EEG can predict survival in patients with acute encephalitis and altered level of consciousness independent from clinical outcome predictors in evaluating patients (Sutter, 2014).
Early EEG patterns did not differ significantly among patients with infectious, autoimmune, or unknown causes. More EEGs were nonreactive to stimuli and revealed frontotemporal and occipital slowing in patients with Herpes Simplex Virus Encephalitis (Sutter, 2014).
Chest x-ray (CXR) is used to detect chest manifestations for infections cause by Mycoplasma, TB, and Legionella (Polhill, 2007)
++
++
Lumbar puncture used only after prominent brain swelling is excluded by a CT scan and determines if any organism or evidence of inflammation present in CSF (cerebrospinal fluid) which helps with differential diagnosis of viruses, autoimmune disorders and chronic disease (Cole, 2007).
Blood testing-
In those with encephalitis with an infectious agent as the source, higher CSF WBC counts were found than in patients who had a noninfectious agent diagnosed. However, the difference in protein levels was not significant (Glaser, 2006).
The West Nile Virus IgM Capture ELISA test is a blood test used to detect encephalitis in a patient with the West Nile virus (Rados, 2003).
Immunologic testing-done to confirm the infectious agent and diagnose autoimmune encephalitis
Specific antibody testing (tissue based assay, immune-blotting, or cell based immunoassay) can expedite the differential diagnosis of encephalitis and autoimmune encephalitis, to select the correct immunotherapy if appropriate (Lee, 2016)
++
++
A diagnosis of encephalitis can be reached after an exam by either general practitioners (GPs) or neurologists.
Difficulties are often encountered by GPs in the diagnosis of Myalgic Encephalitis, which can lead to an increased time window between onset of symptoms and the onset of treatment. (Chew-Graham, 2010.)
Delays in the onset of treatment for viral encephalitis beyond the 48 hours after hospital admission results in a worse prognosis. (Solomon, 2011)
GP may find skin rashes (Rickettsial fever, Colorado tick fever) upon examination, mucous membrane involvement (HSE), or lymph node issues (common with mumps) (Polhill, 2007).
An association of focal piloerection with HSE has been identified and may prove useful in clinical detection (Brazg, 2014).
The Glasgow Coma Scale should be utilized early in treatment. It can be performed quickly and is helpful to standardize examinations, quantify the degree of neurologic dysfunction, and monitor for clinical deterioration or improvement (Simon, 2013).
++
Usually non-progressive with a varied prognosis. Rarely the disease is severe and potentially life-threatening.
In the acute phase, patients may have a gradual or sudden dissolution of fever and neurological symptoms. The acute phase is defined as up to 14 days (Sharma, 2012).
In mild cases some neurological symptoms may require several months to recover. • Expected sequelae include a wide range of functional impairments:
Hearing and/or speech loss, blindness, nerve and brain damage, cognitive impairment, change in personality, lack of muscle control, seizures and memory loss (NIH, 2007). There are also possibilities of secondary bacterial infection, respiratory failure and a number of cardiac abnormalities (Kennedy, 2004; Kneen, 2008).
In severe cases (~10%), permanent impairment or even death is possible. It is also possible for patients to experience brief or prolonged comatose states.
Herpes Simplex Encephalitis (HSE):
HSE has a better prognosis in those under 30 yo with disease for 4 days or less (Whitley, 2002).
Poor prognostic factors include:
Glasgow Coma Scale of 6 or less, older than 30, more than 4 days from onset to start of treatment, abnormal lesion on cranial CT at start of treatment, and more than 100 copies/ml of the HSE-DNA in the CSF (Khaled, 2014)
The most significant factor affecting the HSE prognosis is starting effective antiviral treatment as early as possible (Sili, 2014). With every hour that antiviral administration is delayed, ICU stay is increased by nine hours and death is 1.033 times more likely (Khaled, 2014)
If left untreated, death will typically happens within 10-14 days (Khaled, 2014).
Most patients will suffer from lifelong disabilities and less than 20% are able to return to work (Khaled, 2014).
Tick Borne Encephalitis (TBE):
Has a good prognosis if only found in meninges
Severe symptoms if spreads to brain, upper spinal cord, nerve roots (Vene, 2007).
Far Eastern Encephalitis has a mortality rate of 5-20%) and European Encephalitis has a mortality rate of .5-20% (Dumpis, 1999). Those with TBE have a long-term morbidity of 26-50% (Vene, 2007).
NMDA Encephalitis:
A more severe form of encephalitis, that can a poor prognosis and be potentially fatal in children
This form of encephalitis is frequently accompanied with cognitive changes and seizures (Houtrow, 2012)
In one study, all patients experiencing acute encephalitis with a normal EEG survived. None of these patients had a normal Glasgow Coma Scale at admission nor during EEG, so EEG provides important prognostic information beyond the patients' cognitive conditions (Sutter, 2014).
+++
IV. MEDICAL/SURGICAL MANAGEMENT
++
Mild cases may be treated with rest and a healthy diet to allow the immune system to fight the virus.
To maximize favorable neurologic recovery, efforts should be directed at identification and treatment of neurologic and systemic complications, which could potentially exacerbate brain damage (Kramer, 2013).
On arrival, the initial evaluation should begin with assessment of airway, breathing, and circulation. Hypoxia, hypotension, fever, and seizures should be promptly treated to reduce the likelihood of secondary brain injury (Simon, 2013).
Antiviral medications:
May help treat a viral infection
If anti-viral medication is not administered to patients with HSE, the mortality rate is over 70% (Kennedy, 2004).
Acyclovir
Intravenous (IV) Acyclovir achieves best results when given promptly, however, some patients may experience a relapse and require an additional treatment (Tunkel, 2008).
In a study with 106 adult patients experiencing HSE, Acyclovir was started within five (5) days of onset of main symptoms and continued for ≥ fourteen (14) days. Case fatality rate was 8%, while 69% of patients recovered with sequelae. Favorable prognosis was observed in 73% of patients (Sili, 2014). As soon as the diagnosis of HSE is suspected, the patient should start intravenous (IV) acyclovir,10 mg/kg 3x daily for 14 days. (Kennedy, 2013)
Nitazoxanide (NTZ)
Antibiotic medications:
Prevents secondary bacterial infection
During the appropriate season, if the patient presents with exposure or clinical features of rickettsial or ehrlichial infection, empiric antibacterial treatment should also include doxycycline (Simon, 2013).
Anticonvulsants:
Corticosteroids:
Immunotherapy
For encephalitis characterized by antibodies to neural proteins, immunotherapy treatments have been shown to be extremely successful; on some occasions with a complete return to prior functional status (Gastaldi, 2016)
Other Medications
Sedatives for irritability or restlessness (NINDS, 2014)
Over-the-counter medications for fever and headache (NINDS, 2014)
Surgery:
Implantation of a stent can be performed to relieve increased intracranial pressure (Ramachandran-Nair, 2005)
A new surgical option that can be employed to help control seizures is the implantation of a vagus nerve stimulator (Grujic, 2011)
Supportive care:
Mechanical ventilation is used in severe cases
Monitor breathing, heart rhythm, fluid balance, prevention of DVT
Prevention:
Human vaccination is the only effective, long term, cost effective measure against Acute Encephalitis Syndrome (AES) (Sharma, 2012)
Acute encephalitis syndrome (AES) is a medical and neurological emergency, requiring immediate consideration of key issues including immediate life support, identification of cause, and when available, the institution of specific therapy (Sharma, 2012).
The best way to prevent viral encephalitis is to avoid exposure to disease-causing agents. This can include practicing appropriate hygiene and maintaining current vaccinations.
Herpes Simplex Virus (HSV) (Anzivino, 2009)
22% of pregnant women are infected with HSV-2 and 2% acquire it during pregnancy.
Women who contract the virus during the third trimester or those who test positive for detection test at the time of delivery are encouraged to deliver through caesarean section in order to reduce the risk of transmission to the infant.
60-75% of babies with disseminated HSV-2 develop encephalitis
+++
V. THERAPEUTIC MANAGEMENT
++
Practice Patterns (APTA, 2017)
5A: Primary Prevention/Risk Reduction for Loss of Balance and Falling
5C: Impaired Motor Function and Sensory Integrity Associated With Nonprogressive Disorders of the Central Nervous System—Congenital Origin or Acquired in Infancy or Childhood
5D: Impaired Motor Function and Sensory Integrity Associated with Nonprogressive Disorders of the Central Nervous System – Acquired in Adolescence or Adulthood
5E: Impaired Motor Function and Sensory Integrity Associated With Nonprogressive Disorders of the Central Nervous System - Congenital Origin or Acquired in Infancy or Childhood),
5I: Impaired Arousal, Range of Motion, and Motor Control Associated With Coma, Near Coma, or Vegetative State
7A: Pattern 7A: Primary Prevention/Risk Reduction for Integumentary Disorders
Physical Therapists may play a larger and longer role in the rehabilitation of encephalitis that affects areas in the brain or specific cell populations critical to motor function (Tham, 2012)
For children with NMDA receptor encephalitis, a more severe form of encephalitis, inpatient rehabilitation (including all rehabilitative services) has been reported to last at least 3 hours a day for up to six days a week. However, length of stay is hard to estimate due to the varying severity and course of the disease between individuals (Houtrow, 2012)
Children diagnosed with encephalitis showed functional improvements with inpatient rehabilitation, although the amount of improvement varied; longer inpatient rehabilitation duration was correlated with lower functional status at the time of discharge (Tailor, 2013)
For Treatment Of:
General deconditioning
Strengthening with resistive exercises
A training regime for the deep cervical flexors may be effective for the alleviation of associated neck pain and dizziness after a long period of bed rest. When the neck of a patient has been immobilized, the efficiency of stabilizing muscles should be investigated (Graaf, 2012).
Impaired gait, motor functions, and balance deficits (Miller, 2006)
Stiffness and contractures
Gentle PROM, progressing towards AROM and stretches
Early physical therapy in Acute Encephalitis Syndrome (AES) to prevent the development of contractures. (Sharma, 2012)
Pain
Patient Education
Vector control, educating the public to avoid high-risk areas, wearing protective clothing, and using DEET-containing insect repellents (Alatoom, 2009).
In patients with AES, regular posture change must be done to prevent the development of bed sores. (Sharma, 2012)
In the early stages of rehabilitation the patient may benefit from aquatic exercises for strengthening. Goals should focus on preventing patient fatigue. Bobath/NDT may be used to improve muscle strength and co-contraction (Miller, 2006). Severe Encephalitis may require long-term therapy, medication and supportive care (NIH, 2007).
+++
VI. CONSUMER AND PROFESSIONAL RESOURCES
++
Mayo Clinic: http://www.mayoclinic.com/health/encephalitis/DS00226.
Website to provide more information on diagnosis, symptoms, treatment, causes, risk factors, complications, and prevention for health care professionals
MD Guidelines: http://www.mdguidelines.com/encephalitis
The Encephalitis Society: http://www.encephalitis.info/
Website to provide to patients with resources including counseling via telephone, email, outreach, links to information on equipment, family support, books on encephalitis, newsletter, resources for teachers, retreats for families, and meetings about encephalitis
Encephalitis Global http://www.encephalitisglobal.org/
Charity based organization that connects encephalitis survivors and caregivers globally
Discussion forums for individuals, promotes awareness, books, videos of people with encephalitis
Daily Strength: Encephalitis Support Group http://www.dailystrength.org/c/Encephalitis/support-group
Encephalitis Global Support Community: http://www.inspire.com/groups/encephalitis-global/
Online support community sponsored by Encephalitis Global Inc.
Connects survivors and caregivers to share information and support, raise awareness and alleviate unnecessary suffering
ThirdAge.com: Encephalitis Resource Guide: http://www.thirdage.com/hc/c/encephalitis-resource-guide
ExperienceProject.com: Anonymous Encephalitis Support Group: http://www.experienceproject.com/groups/Have-Herpes-Simplex-Encephalitis/94471
An anonymous support group with information on diagnosis, treatment, symptoms, along with personal stories and experiences
The link is directed to the support group for Herpes Simplex Encephalitis. There are, however, support groups for most forms of encephalitis.
Medline Plus: https://www.nlm.nih.gov/medlineplus/encephalitis.html
Website created by the National Institute of Health from the U.S National Library of Medicine that provides general information about encephalitis as well as link to numerous other reputable resources.
Sub-sections include basic information regarding encephalitis, research, resources to find experts and more.
Also available in Español.
+
Adang
LA, Lynch
DR, Panzer
JA. Pediatric anti-NMDA receptor encephalitis is seasonal. Ann Clin Transl Neurol. 2014;1(11):921–925.
+
Aksamit
AJ. Acute viral encephalitis. In: Goldman
L, Schafer
AI. Cecil Medicine. 24th ed. Philadelphia, Pa: Saunders Elsevier;2011: chap 422.
+
Alatoom
A, Toom
D. CE update. An overview of arboviruses and Bunyaviruses. Lab Med. 2009;40(4):237–41.
+
Anzivino
E, Fioriti
D, Mischitelli
M, Bellizzi
A, Barucca
V, Chiarini
F, Pietropaolo
V. Herpes simplex virus infection in pregnancy and in neonate: status of art of epidemiology, diagnosis, therapy and prevention. Virol J. 2009;6:40. doi:10.1186/1743-422X-6-40.
+
Aryee
A, Thwaites
G. Viral encephalitis in travellers. Clin Med. 2015;15(1):86–90.
+
Bigham
AW, Buckingham
KJ, Husain
S,
et al. Host Genetic Risk Factors for West Nile Virus Infection and Disease Progression. PLoS One. 2011;6(9).
+
Bleck
TP. Arthropod-borne viruses affecting the central nervous system. In: Goldman
L, Schafer
AI. Cecil Medicine. 24th ed. Philadelphia, Pa: Saunders Elsevier; 2011: chap 391.
+
Brazg
J, Gillett
B. A hair-raising diagnosis: goosebumps as sign of herpes simplex encephalitis. Am J Emerg Med. 2014;33(5):742-e1. doi: 10.1016/j.ajem.2014.11.030.
+
Chew-Graham
C, Dowrick
C, Wearden
A,
et al. Making the diagnosis of Chronic Fatigue Syndrome/Myalgic Encephalitis in primary care: a qualitative study. BMC Fam Pract. 2010;11:16. doi: 10.1186/1471-2296-11-16.
+
Cole
A, Henson
J, Roehrl
M. et al. A 20-year-old pregnant woman with altered mental status. N Engl J Med. 2007;357(6):589.
+
Davis
L, DeBasi
R, Goade
D,
et al. West nile virus neuroinvasive disease. Ann Neurol. 2006;60:286–300.
+
Dumpis
U, Crook
D, Oksi
J. Tick-borne encephalitis. Clin InfectDis. 1999;28(4):882e90.
+
Eckhardt
P, Gniel
D, Zent
O. Tick-borne encephalitis (TBE) trends in epidemiology and current and future management. Travel Med Infect Dis. 2010;8:233–245.
+
Encephalitis Global. Available at: http://www.encephalitisglobal.org. Accessed on: January 20, 2013.
+
Gastaldi
M, Thouin
A, Vincent
A. Antibody-Mediated Autoimmune Encephalopathies and Immunotherapies. NeuroRx. 2016;13(1):147–162.
+
George
BP, Schneider
EB, Venkatesan
A. Encephalitis Hospitalization Rates and Inpatient Mortality in the United States, 2000-2010. PLoS ONE 9(9):e104169.
+
Glaser
CA, Honarmand
S, Anderson
J, Schnurr
DP, Forghani
B, Cossen
CK, Schuster
FL, Christie
LJ, Tureen
JH. Beyond Viruses: Clinical Profiles and Etiologies Associated with Encephalitis. Clin Infect Dis. 2006;43(12):1565–1577.
+
Graaf
M, Schmitt
M. The effect of training the deep cervical flexors on neck pain, neck mobility, and dizziness in a patient with chronic nonspecific neck pain after prolonged bed rest: A Case Report. J. Orthop Sports Phys Ther. 2012:42(10):853–860.
+
Granerod
J, Cousens
S, Davies
NW, Crowcroft
NS, Thomas
SL. New estimates of incidence of encephalitis in England. Emerging Infect Dis. 2013;19(9):1455–1462. doi: 10.3201/eid1909.130064.
+
Grujic
J, Bien
CG, Pollo
C,
et al. Vagus nerve stimulator treatment in adult-onset Rasmussen's encephalitis. Epilepsy Behav. 2011;20:123–125.
+
Grygorczuk
S, Parczewski
M, Moniuszko
A, Swierzbinska
R, Kondrusik
M, Zaikowska
J, Czupryna
P, Dunai
J, Boron-Kaczmarsak
A, Pancewica
S. Increased concentration of interferon lambda-3, interferon beta and interleukin-10 in the cerebrospinal fluid of patients with tick-borne encephalitis. Cytokine. 2015:71(2):125–131.
+
Graus
F, Titulaer
MJ, Balu
R,
et al. A clinical approach to diagnosis of autoimmune encephalitis. Lancet Neurol. 2016;15:391–404.
+
Houtrow
AJ, Bhandal
M, Pratini
NR, Davidson
L, Neufeld
JA. The Rehabilitation of Children with Anti-NMDA-Receptor Encephalitis: A Case Series. Am J Phys Med Rehabil. 2012;91(5):435.
+
Hills
SL, Griggs
AC, Fischer
M. Japanese Encephalitis in Travelers from Non-Endemic Countries, 1973-2008. Am J Trop Med Hyg. 2010;82(5):930–936.
+
Hong
S, Klein-Gitelman
M, Wainwright
MS. Recognition and Treatment of Anti-N-Methyl-D-Aspartate Receptor Encephalitis. Clin Pediatr Emerg Med. 2015;16(1):3–10.
+
Jmor
F, Emsley
HC, Fischer
M, Solomon
T, Lewthwaite
P. The incidence of acute encephalitis syndrome in Western industrialised and tropical countries. Virol. J. 2008;5:134. doi:10.1186/1743-422X-5-134.
+
Kennedy
P..G.E. Viral encephalitis: causes, differential diagnosis, and management. J Neurol Neurosurg Psychiatry. 2004;75(Supplement 1):i10–15.
+
Kennedy
PG, Steiner
I. Recent issues in herpes simplex encephalitis. J Neurovirol. 2013;19(4):346–50.
+
Khaled
M, Youssof
AMA, Mourad
HS. Clinical and Laboratory Predictors of Outcome in Patients with Herpes Simplex Encephalitis. Egypt J Neurol Psychiatry Neurosurg. 2014;51(2):173–180.
+
Kneen
R, Solomon
T. Management and outcome of viral encephalitis in children. Paediatrics and Child Health. 2008;18(1):7–16.
+
Kramer,
A. Viral Encephalitis in the ICU. Crit Care Clin. 2013;3:621–649.
+
Lancaster
E. The Diagnosis and Treatment of Autoimmune Encephalitis. J Clin Neurol. 2016;12(1):1–13.
+
Lancaster
E, Martinez-Hernandez
E, Dalmau
J. Encephalitis and antibodies to synaptic and neuronal cell surface proteins. Neurology. 2011;77(2):179–189.
+
Lee
SK, Lee
ST. The Laboratory Diagnosis of Autoimmune Encephalitis. J Epilepsy Res. 2016;6(2):45–52.
+
Lin
K-L, Lin
J-J, Hsia
S-H, Chou
M-L, Hung
P-C, Wang
H-S. Effect of Antiepileptic Drugs for Acute and Chronic Seizures in Children with Encephalitis. PLoS ONE. 2015;10(10):1–11. doi:10.1371/journal.pone.0139974.
+
Mann
A, Machado
NM, Liu
NM, Mazin
A, Silver
K, Afzal
KI. A Multidisciplinary Approach to the Treatment of Anti-NMDA-Receptor Antibody Encephalitis: A Case and Review of the Literature. J Neuropsychiatry Clin Neurosci. 2012;24:247–254.
+
Miller
N, Miller
D, Goldberg
J. Physical therapist examination, evaluation, and intervention for a patient with West Nile virus paralysis. Phys Ther. 2006;86:843–856.
+
Pandey
S, Rathore
C, Michael
BD. Antiepileptic drugs for the primary and secondary prevention of seizures in viral encephalitis. Cochrane Database of Systematic Reviews 2014; 10: CD010247.
+
Polhill
S, Soni
N. Encephalitis in the ICU setting. Current Anaesthesia & Critical Care. 2007;18:107–116.
+
Rados
C. First test approved to detect West Nile virus. FDA Consumer. 2003;37:18–19.
+
Ramachandran-Nair
R. Hemicraniectomy to reduce raised intracranial pressure in a child with acute disseminated encephalomyelitis. J Pediatr Neurol. 2005;3:45–48.
+
Randolph
SE. Human activities predominate in determining changing incidence of tick-borne encephalitis in Europe. Euro Surveill. 2010;15(27):24–31.
+
Rath
T, Hughes
M, Arabi
M, Shah
G. Imaging of Cerebritis, Encephalitis, and Brain Abscess, Neuroimag Clin N Am. 2012;22:585–607.
+
Rotela
CH, Spinsanti
LI, Lamfri
MA,
et al. Mapping environmental susceptibility to Saint Louis encephalitis virus, based on a decision tree model of remotely-sensed data. Geospatial Health. 2011:6(1);85–94.
+
Sharma
S, Mishra
D, Aneja
S, Kumar
R, Jain
A, Vashishtha
V. Consensus guidelines on evaluation and management of suspected acute viral encephalitis in children in India. Indian Pediatr. 2012;49(11):897–910.
+
Shi
Z, Wei
J, Deng
X, Li
S, Qui
Y, Shao
D, Li
B, Zhang
K, Xue
F, Wang
X, Ma
Z. Nitazoxanide inhibits the replication of Japanese encephalitis virus in cultured cells and in a mouse model. Virol J. 2014;11(1):10.
+
Simon
D, Yong
Sing Da S, Zuccoli
G, Clark
R. Acute Encephalitis. Crit Care Clin. 2013;29:259–277.
+
Solomon
T, Michael
BD, Smith
PE, Sanderson
F, Davies
NW, Hart
IJ, Holland
M, Easton
A, Buckley
C, Kneen
R, Beeching
NJ. Management of suspected viral encephalitis in adults–association of British Neurologists and British Infection Association National Guidelines. J Infect. 2012;64(4):347–73.
+
Sutter
R, Kaplan
P, Cervenka
M, Thakur
K, Asemota
A, Venkatesan
A, Geocadin
R. Electroencephalography for diagnosis and prognosis of acute encephalitis. Clin Neurophysiol. 2015;126(8):1524–31.
+
Tailor
YI, Suskauer
SJ, Sepeta
LN,
et al. Functional status of children with encephalitis in an inpatient rehabilitation setting: A case series. J Pediatr Rehabil Med. 2013;6(3):163–173.
+
Tie
ML. Basic head CT for intensivists. Crit Care Resusc. 2001;3(1):35–44.
+
Tham
SL, Kong
KH. A case of Anti-NMDAR (N-Methyl-D-Aspartate Receptor) Encephalitis: A rehabilitation perspective. Neurorehabilitation. 2012;30(2):109–112.
+
Tunkel
Allan R R, Glaser
Carol A A, Bloch
Karen C C
et al.. The Management of Encephalitis: Clinical Practice Guidelines by the Infectious Diseases Society of America. Clin Infect Dis. 2008;47(3):303–27.
+
Vene
S, Haglund
M, Lundkvist
A°, Lindquit
L, Forsgren
M. Study of the serological response after vaccination against tick-borne encephalitis in Sweden. Vaccine. 2007;25(2):366e72.
+
Whitley
RJ, Gnann
JW. Viral encephalitis: familiar infections and emerging pathogens. Lancet. 2002;359(9305):507–13.
+
Yao
K
et al.. Detection of human herpesvirus-6 in cerebrospinal fluid of patients with encephalitis. Ann Neurol. 2009 Mar; 65(3): 257–67.
+++
Fetal Alcohol Syndrome
++
Prenatal exposure to alcohol causing brain damage manifested by functional impairments of neurocognition, self-regulation, and adaptive behavior, termed neurobehavioral disorder associated with prenatal alcohol exposure (ND-PAE) in most recent Diagnostic Statistical Manual, ed. 5 (DSM-5) (Hagan, 2016; American Psychological Association, 2013);
Fetal Alcohol Spectrum Disorders (FASD) better signifies the variable presentation in terms of severity and symptoms than fetal alcohol syndrome (FAS) (Bertrand, 2005; Brown, 2010);
Three categories of conditions (Miller, 2013).
Fetal alcohol syndrome (FAS): severe type; can result in death. Characteristics include: facial feature abnormalities, lacking proper growth development, and central nervous system (CNS) issues. Other issues include: " learning, memory, attention span, communication, vision, or hearing." Difficulty with school and social situations also possibly observed.
Alcohol-related neurodevelopmental disorder (ARND): Possible intellectual disabilities or behavior and learning. At risk for doing unsatisfactory in school and problems with "math, memory, attention, judgment, and poor impulse control."
Alcohol-related birth defects (ARBD): Possible issues include: heart, kidneys, bones, or hearing.
[ARND and ARBD categories are secondary to prenatal alcohol exposure; however, they do not exhibit the physical signs associated with FAS.]
Center for Disease Control and Prevention (CDC) monitors alcohol consumption in women of childbearing age (18-44 years) in United States via the Behavioral Risk Factor Surveillance System; women asked about drinking habits in the 30 days prior to the survey using categories of "any alcohol use" and "binge drinking (4 or more drinks on any one occasion)"; data is available by state on the webpage. (https://www.cdc.gov/brfss/)
Alcohol consumption during pregnancy preventable and prevention would result in considerable cost savings for society (Klug, 2003; Popova, 2012)
Characteristics of mothers of children with FASD:
Age 35-44 years (14.3%), white (8.3%), college graduates (10.0%), or employed (9.6%) (CDC, 2012)
10.2% of pregnant women reported consuming alcohol during past 30 days and 3.1% reported binge drinking within past 30 days (Tan, 2015)
Population Based Studies
7 fold higher prevalence in African Americans compared to Caucasians (Ornoy, 2010)
10 fold higher prevalence in Native Americans (Thackray, 2001)
Maternal risk factors
Age over 30, low socioeconomic status, ethnicity, genetic sensitivity to alcohol, malnutrition, and a history of binge drinking.
Other adverse effects on pregnancy/birth/infancy: spontaneous abortion, placental abruption, preterm delivery, amnionitis, stillbirth, and sudden infant death syndrome.
Prevalence of FASD - 0.2 to 1.5 per 1000 live births (Bertrand, 2005; Muenke, 2005; Tsai, 2007)
Based on testing of 6-7 year olds (record review), incidence of FASD in United States estimated at 2-5%, many not diagnosed (per review of medical records)
Pathogenesis: (CDC, 2017)
Based on MRI studies, brain volume, isocortical volume, isocortical thickness, and isocortical surface area all reduced; regional patterns in cortical thickness differences suggested primary sensory areas were particularly vulnerable to gestational EtOH exposure. (Leigland, 2013)
Ethanol along with its metabolite acetaldehyde] alters development through disturbance of cellular differentiation and growth, impedance of DNA and protein synthesis, and inhibition of cell migration.
Changes metabolism of carbohydrates, proteins, and fats
Decreases the transport of amino acids, glucose, folic acid, zinc, and other nutrients through the placenta affecting fetal growth due to intrauterine nutrient deprivation
Controversy in research about whether habitual exposure to alcohol or episodic binge drinking has a greater effect on the fetus
Critical time periods throughout fetal development; alcohol exposure will have different effects depending on timing (Thackray,2001; Ornoy, 2010)
++
DSM-5 "Proposed Criteria for Neurobehavioral Disorder Associated with Prenatal Alcohol Exposure" provides an important diagnostic framework; note the absence of criteria related to physical features (i.e., dysmorphia and growth restriction) (American Psychological Association, 2013; Hagan, 2016)
More than minimal exposure to alcohol during gestation, including prior to pregnancy recognition. Confirmation of gestational exposure to alcohol may be obtained from maternal self-report of alcohol use in pregnancy, medical or other records, or clinical observation. ["more than minimal exposure to alcohol" is defined as >13 drinks per month during any phase of pregnancy]
Impaired neurocognitive functioning as manifested by one or more of the following:
Impairment in global intellectual performance (i.e., IQ of 70 or below, or a standard score of 70 or below on a comprehensive developmental assessment).
Impairment in executive functioning (e.g., poor planning and organization; inflexibility: difficulty with behavioral inhibition).
Impairment in learning (e.g., lower academic achievement than expected for intellectual level; specific learning disability).
Memory impairment (e.g., problems remembering information learned recently; repeatedly making the same mistakes; difficulty remembering lengthy verbal instructions).
Impairment in visual–spatial reasoning (e.g., disorganized or poorly planned drawings or constructions; problems differentiating left from right).
Impaired self-regulation as manifested by one or more of the following:
Impairment in mood or behavioral regulation (e.g., mood lability; negative affect or irritability; frequent behavioral outbursts).
Attention deficit (e.g., difficulty shifting attention; difficulty sustaining mental effort).
Impairment in impulse control (e.g., difficulty waiting turn; difficulty complying with rules).
Impairment in adaptive functioning as manifested by two or more of the following, one of which must be (1) or (2):
Communication deficit (e.g., delayed acquisition of language; difficulty understanding spoken language).
Impairment in social communication and interaction (e.g., overly friendly with strangers; difficulty reading social cues; difficulty understanding social consequences).
Impairment in daily living skills (e.g., delayed toileting, feeding, or bathing; difficulty managing daily schedule).
Impairment in motor skills (e.g., poor fine motor development; delayed attainment of gross motor milestones or ongoing deficits in gross motor function; deficits in coordination and balance)
Onset of the disorder (symptoms in Criteria B, C, and D) occurs in childhood.
The disturbance causes clinically significant distress or impairment in social, academic, occupational, or other important areas of functioning.
The disorder is not better explained by the direct physiologic effects associated with postnatal use of a substance (e.g., a medication, alcohol or other drugs), a general medical condition (e.g., traumatic brain injury, delirium, dementia), another known teratogen (e.g., fetal hydantoin syndrome), a genetic condition (e.g., Williams syndrome, Down syndrome, Cornelia de Lange syndrome), or environmental neglect.
Signs and Symptoms – as specified in DSM-5 criteria as well as variable occurrence of the following (Gurerri, 2009; Hagan, 2016; Osborn, 1993):
Neuromuscular: fine motor, gross motor and visual motor deficits; balance deficits, neurological soft signs; sensory processing deficits
Gross motor impairments; problems with balance control including static balance in standing (increased postural sway with perturbations), coordination, and ball skills (Osborn, 1993; Harris, 1993; Lucas, 2014; Jirikowic, 2016)
Musculoskeletal: characteristic facial anomalies, joint anomalies with abnormal position or function, maxillary hypoplasia, growth restrictions (height and/or weight), microcephaly
Cardiopulmonary: cardiac defects
Poor sleep patterns, eating disorders, depression, phobias
Microcephaly in >80% (Osborn, 1993)
Diagnostic Testing
Multidisciplinary testing, typically using standardized, norm-referenced measurement tools: cognitive, adaptive, motor, anthropometric measures
Triggers for diagnostic evaluation (Bertrand, 2005)
Known prenatal exposure to alcohol; an evaluation is necessary if the mother confirms drinking 7 or more drinks per week, and/or 3 or more drinks on multiple occasions.
Unknown prenatal exposure to alcohol with reports of concern by parent or caregiver; presence of diagnostic facial features; concern related to family history or environmental situation
Withdrawal symptoms may be present if the infant was exposed to high levels of alcohol: jitteriness, tremors, hypotonia, and gastrointestinal (GI) symptom
+++
III. Prognosis and Sequelae (Bertrand, 2005; Kahn-D'Angelo, 2000)
++
Non-progressive—prognosis depends on specific toxic exposure, timing of exposure, severity of substance abuse and severity of symptoms
Physical anomalies and developmental delays are the primary manifestations from birth to childhood; at childhood, impairments in executive cognitive function (judgment, impulsivity, problem solving) and behavioral aspects become more pronounced and remain throughout the child's life often leading to maladaptive social functioning.
Clinical manifestations likely to continue into adulthood with increasing importance of impairments of executive cognitive function (judgment, impulsivity, problem solving) and adaptive behavior as child matures and progresses in educational and vocational attainment (Kully- Martens, 2012).
Adverse outcomes and risk/protective factors examined by Streissguth et al (2004) for 415 patients with FASD (ages 6-51; median age 14 years):
80% not raised by biological mothers
For adolescents and adults, the life span prevalence: 61% for disrupted school experiences, 60% for trouble with the law, 50% for confinement (i.e., detention, prison, or psychiatric or alcohol/drug inpatient setting), 49% for inappropriate sexual behaviors on repeated occasions, and 35% for alcohol/drug problems
92% problems with independent living or employment for complex reasons
Factors that help reduce the effects of FASD and promote successful long term outcomes (Leigland, 2013; Streissguth, 2004; Peadon, 2009):
Early diagnosis and intervention (before age 6 years)
Supportive, stable home environment without violence
Involvement in special education and social services
+++
IV. Medical/Surgical Management
++
No cure for FASD; intervention focused on management of symptoms
High levels of exposure to alcohol in the womb may result in withdrawal symptoms at birth; including irritability, tremors, apnea, and seizures; withdrawal may be treated symptomatically with Phenobarbital or Morphine. (Kahn-D'Angelo, 2000)
Children born with the medical complications of FASD, such as otitis media, pneumonia and central nervous system damage, often need hospitalization to combat these conditions (Kvigne, 2004)
Since many children with complete or incomplete FASD require hospitalization and social service intervention, some families often have higher medical bills and may require assistance with funding and continued care (Kvigne, 2004)
Uncertainty continues about association of congenital heart defects (CHDs) with FASD; meta-analysis indicated prenatal alcohol exposure was not associated with overall CHDs or some subtypes; marginally significant association with conotruncal defects and statistically significant association with transposition of great arteries (Yang, 2015).
No specific medication exists to treat FASD, but behavior impairments may be pharmacologically managed by psycho-stimulant medications (ADHD), antidepressants, anxiety and anti-psychotic medications (Kodituwakku, 2011; CDC, 2017)
+++
V. Key Aspects of Therapeutic Management
++
Overview of evidence for pharmacological and non-pharmacological interventions, focused on neurobehavioral problems (Peadon, 2009)
Preventative Education
The best therapeutic treatment is preventative education regarding risks of drinking (Bertrand, 2005; Kumpfer, 2007)
Family centered care and early intervention for optimal rehabilitation results (Bertrand, 2009)
Maternal and child advocacy to maximize functional outcomes
Multidisciplinary approach
Behavioral, educational, social, physical, and occupational therapy may all be necessary, depending on the extent of the child's deficits. (Bertrand, 2005)
Specifically, physical and occupational therapy may be needed for children who exhibit sensory motor impairments and difficulty with state regulation (Bertrand, 2005; Jirikowic, 2008)
Physical therapy evaluation of motor proficiency and intervention to address motor delays advisable to intervene as early as possible, transitioning from direct to indirect or consultative delivery model as appropriate (Osborn, 1993; Lucas, 2014; Harris, 1993)
Occupational therapy for fine motor delays, sensory processing disorder and emerging self-regulation problems
Task-specific practice using virtual game technology to train balance control under altered sensory conditions; using Sensorimotor Training to Affect Balance, Engagement and Learning (STABEL); results of pilot study indicated improved sensory adaptation, balance and motor performance (Jirikowic, 2016)
Speech and language therapy for any expressive or receptive speech-related delays or articulation problems
Referral to mental health system as child matures (psychologist and/or psychiatrists)
Caregiver involvement and education crucial
Special Education and Support Interventions (Bertrand, 2005)
Special education for individualized learning and vocational needs; many children with a FASD may not qualify for special education but may still need support
Explicit teaching techniques, repetitive presentation (Hagan, 2015)
Individualized intervention strategies to stabilize home environment and improve parent-child relationships
Neurocognitive habilitation therapy (Wells, 2012)
Alternative Approaches (CDC, 2017)
Biofeedback
Auditory training
Relaxation therapy, visual imagery and meditation
Creative art therapy
Yoga and exercise
Acupuncture and acupressure
Massage, Reiki, and energy healing
Vitamins, herbal supplements, and homeopathy
Animal-assisted therapy
+
American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. 5th edition. Washington, DC: American Psychiatric Association; 2013.
+
Bertrand
J. Interventions for children with fetal alcohol spectrum disorders (FASDs): overview of findings for five innovative research projects. Res Dev Disabil. 2009;30(5):986–1006.
+
Bertrand
J, Floyd
LL, Weber
MK. Guidelines for identifying and referring persons with fetal alcohol syndrome. MMWR Recomm Rep. 2005;28(54):1–14.
+
Brown
C, Olson
H, Croninger. Maternal alcohol consumption during pregnancy and infant social, mental, and motor development. J Early Interv. 2010;32(2):110–126. 8. PubMed PMID: 22810267.
+
Guerri
C, Bazinet
A, Riley
EP. Fetal alcohol spectrum disorders and alterations in brain and behavior. Alcohol. 2009;44:108–114.
+
Hagan
JF, Balachova
T, Bertrand
J,
et al. Neurobehavioral disorder associated with prenatal alcohol exposure. Pediatr. 2016;138(4):e20151553.
+
Harris
SR, Osborn
JA, Weinberg
J, Loock
C, Junaid
K. Effects of prenatal alcohol exposure on neuromotor and cognitive development during early childhood: a series of case reports. Phys Ther. 1993;73(9):608–617.
+
Hellemans
KGC, Sliwowska
JH, Verma
P, Weinberg
J. Prenatal alcohol exposure: fetal programming and later life vulnerability to stress, depression and anxiety disorders. Neurosci Biobehav Rev. 2010;34(6):791–807.
+
Jirikowic
T, Olson
HC, Kartin
D. Sensory processing, school performance, and adaptive behavior of young school-age children with fetal alcohol spectrum disorders. Phys Occup Ther Pediatr. 2008; 28(2):117–136.
+
Jirikowic
T, McCoy
SW, Price
R, Ciol
MA, Lin-Ya-H, Kartin
D. Virtual sensorimotor training for balance: pilot study results for children with fetal alcohol spectrum disorders. Pediatr Phys Ther. 2016;28:460–468.
+
Klug
M, Burd
L. Fetal alcohol syndrome prevention: annual and cumulative cost savings. Neurotoxicol Teratol. 2003;25:763–765.
+
Kodituwakku
PW, Kodituwakku
EL. From research to practice: an integrative framework for the development of interventions for children with fetal alcohol spectrum disorders. Neuropsychol Rev. 2011;21(2):204–223.
+
Kully-Martens
K, Denys
K, Treit
S, Tamana
S, Rasmussen
C. A review of social skills deficits in individuals with fetal alcohol spectrum disorders and prenatal alcohol exposure: profiles, mechanisms, and interventions. Alcohol Clin Exp Res. 2012;36(4):568–576.
+
Kumpfer
K, Fowler
M. Parenting skills and family support programs for drug-abusing mothers. Semin Fetal Neonatal Med. 2007;12:134–142.
+
Kvigne
K, Leonardson
G, Neff-Smith
M, Brock
E, Borzelleca
J, Welty
T. Characteristics of children who have full or incomplete fetal alcohol syndrome. J Pediatr. 2004;145:635–640.
+
Leigland
L, Ford
MM, Lerch
JP, Kroenke
CD. The influence of fetal ethanol exposure on subsequent development of the cerebral cortex as revealed by magnetic resonance imaging. Alcohol Clin Exp Res. 2013;37(6):924–932.
+
Lucas
BR, Latimer
J, Pinto
RZ,
et al. Gross motor deficits in children prenatally exposed to alcohol: a meta-analysis. Pediatr. 2014;134(1):e192–e209.
+
McGee
CL, Riley
EP. Fetal alcohol spectrum disorders: an overview with emphasis on changes in brain and behavior. Exp Biol Med. 2005; 230: 357–365.
+
Miller
AR. Diagnostic nomenclature for foetal alcohol spectrum disorders: the continuing challenge of causality. Child Care Health Dev. 2013 Jan 7. doi: 10.1111/cch.12017. [Epub ahead of print] PubMed PMID: 23294157.
+
Miodovnik
A. Environmental neurotoxicants and developing brain. Mt Sinai J Med. 2011;78(1):58–77.
+
Muenke
M, Wattendorf
DJ. Fetal alcohol spectrum disorders. Am Fam Physicians. 2005; 72: 279–285.
+
Ornoy
A, Ergaz
Z. Alcohol abuse in pregnant women: effects on the fetus and newborn, mode of action and maternal treatment. Int J Environ Res Public Health. 2010;7(2):364–379.
+
Osborn
JA, Harris
SR, Weinberg
J. Fetal alcohol syndrome: review of the literature with implications for physical therapists. Phys Ther. 1993;73:9;599–607.
+
Paintner
A, Williams
AD, Burd
L. Fetal alcohol spectrum disorders-- implications for child neurology, part 1: prenatal exposure and dosimetry. J Child Neurol. 2012;27(2):258–263.
+
Peadon
E, Phys-Jones
B, Bower
C, Elliot
EJ. Systematic review of interventions for children with fetal alcohol spectrum disorders.
BMC Pediatr. 2009;9:35.
https://doi.org/10.1186/1471-2431-9-35.
+
Popova
S, Lange
S, Burd
L, Rehm
J. Health care burden and cost associated with fetal alcohol syndrome: based on official Canadian data. PLoS ONE. 2012;7(8):e43024. doi:10.1371/journal.pone.0043024.
+
Streissguth
AP, Bookstein
FL, Barr
HM, Sampson
PD, O'Malley
K, Young
JK. Risk factors for adverse life outcomes in fetal alcohol syndrome and fetal alcohol effects. Dev Behav Pediatr. 2004;5(4):228–238.
+
Tan
CH, Denny
CH, Cheal
NE, Sniezek
JE, Kanny
D. Centers for Disease Control and Prevention. Alcohol use and binge drinking among women of childbearing age: United States, 2011-2013. MMWR Morb Mortal Wkly Rep. 2015;64(37):1042–1046.
+
Tsai
J, Floyd
RL, Green
PP, Boyle
CA. Patterns and average volume of alcohol use among women of childbearing age. Matern Child Health J. 2007; 11:5 437–445.
+
Thackray
HM, Tifft
C. Fetal alcohol syndrome. Pediatr Rev. 2001;22(2):47–55.
+
Wells
AM, Chasnoff
IJ, Schmidt
CA, Telford
E, Schwartz
L. Neurocognitive habilitation therapy for children with fetal alcohol spectrum disorders: an adaptation of the Alert Program. Am J Occup Ther. 2012;66:24–34.
+
Yang
J, Qiu
H, Qu
P,
et al. Prenatal alcohol exposure and congenital heart defects: a meta-analysis. PLoS One. 2015;10(6):e0130681.
++
Incidence and Prevalence
Affects 5 million people in the US (about 2-4% of population)
More prevalent in women
Affects 9 times more women than men (Staud, 2006);
80-90% are women (Vitorino, 2006)
Affects 2%-6% of pediatric population (Sherry, 2015).
Onset most common in individuals ages 40-75
3rd most prevalent rheumatologic disorder in US (Peterson, 2007).
Etiology is unknown
Research is now ongoing to determine the cause of fibromyalgia; most likely the initiation of this condition is multifactorial, resulting from a combination of both physical and emotional stress
Thought to be caused by a short-term stressor that causes a chronic illness (Pall, 2001)
Ineffectiveness in lowering peroxynitrite levels also plays a role (Nathan, 1992)
Central sensitization, considered a main mechanism: "spontaneous nerve activity, enlarged receptive fields, and augmented stimulus responses transmitted by primary afferent fibers is excessive in fibromyalgic patients" (Bellato, 2012)
Genetic predisposition; triggered by traumatic event or injury (motor vehicle accident, surgery) and is often seen in the craniocervical region (Watson, 2009).
Physiological abnormalities include:
Low levels of blood flow to the thalamus, low levels of serotonin and tryptophan, increased levels of substance P in the spinal cord, abnormalities in cytokine function (NFA, 2007).
Sensory processing abnormalities of the central nervous system, hypothalamic-pituitary axis, and peripheral tissues (Crofford, 2002; Crofford, 2004)
Risk factors:
Age (early to middle adulthood), gender (female), disturbed sleep patterns, family history of the disease -transmission may be polygenic (Bellato, 2012)
Rheumatic disease (Dadabhoy 2006; Mayo Clinic, 2014).
Commonly associated with psychiatric and psychological symptoms such as depression, anxiety, sleep disturbance, and cognitive dysfunction (Chang, 2015)
Patients with depression have increased risk of developing fibromyalgia (Chang, 2015)
++
No blood tests, x-rays or muscle biopsies can detect fibromyalgia (Move Forward PT, 2011)
Signs:
Widespread muscle pain (Watson, 2009; Mannerkorpi, 2017)
Most common sites are neck, back, shoulders, hands, and pelvic girdle
Sleep disturbances (Staud, 2006; Watson, 2009)
Fatigue (Krsnich-Shriwise, 1997; Staud, 2006; Watson, 2009)
Various neurological complaints (Watson, 2009)
Paresthesia, blurred vision, numbness, weakness (Krsnich-Shriwise, 1997; NFA, 2007)
May have tingling and burning or numbness with pain worse in the morning (NFA, 2007).
Pain Sensitivity and Tenderness (Mannerkorpi, 2017)
Stabbing, shooting, throbbing, aching deep muscular pain (NFA, 2007)
Diffuse, multifocal, deep, gnawing, burning, periods of waxing-waning, and migratory pain (Bellato, 2012)
Impaired pain inhibition implies pain perception increases during physical activities (Mannerkorpi, 2017)
Associated with impaired physical fitness (Mannerkorpi, 2017)
Stiffness
In addition, tender points, soft tissue swelling, heightened sensitivity, muscle spasms or nodules (Krsnich-Shriwise, 1997; Bellato, 2012).
Increased emotions (Staud, 2006)
American College of Rheumatology Standards (Krsnich-Shriwise, 1997; Longley, 2006; Bellato, 2012).
History of widespread pain greater than 3 months, accompanied by pain in 11/ 18 tested tender points on both sides of the body including: arms, buttocks, chest, knees, lower back, neck, rib cage, shoulders, and thighs.
Diagnosis:
Medical history, FIQ questionnaire, physical exam, and neurological exam needed; no lab tests available to rule in fibromyalgia, but lab tests often required to rule out (Krsnich-Shriwise, 1997, NFA, 2007, Peterson, 2007; Bennett, 2009).
A differential diagnosis of five conditions should be considered to decrease risk of misdiagnosis: mental health disorders, hypothyroidism, rheumatoid arthritis, adrenal dysfunction, multiple myeloma (Bellato, 2012).
Complications:
Headaches and migraines, impaired memory and concentration, dry eyes and mouth, depression, anxiety, irritable bowel and bladder, and dizziness (NFA, 2007).
++
Non-progressive disease (Mayo Clinic, 2007)
Symptoms tend to wax and wane with chronic fibromyalgia (NFA, 2007).
People with mild symptoms have a positive long-term outcome
Most people experience ongoing symptoms of fibromyalgia for a long period of time and possibly even for the rest of their life.
Fibromyalgia was determined to have a better prognosis than chronic fatigue syndrome with the length of illness only 51 days/year as compared to 80 days/year (Hamilton, 2005).
Patients who were younger in age and had shorter duration of symptoms at diagnosis had better outcomes (Kennedy, 1996).
Does not lead to the development of other conditions or diseases (Mayo Clinic, 2007).
Subjective complaints:
Economic problems due to inability to work; isolation and hopelessness; decreased social activities (Foresth, 2006).
Combination of exercise, education, and psychological assistance with medications improves well-being significantly (Peterson, 2007).
Research has shown that patients with fibromyalgia participating in aerobic conditioning, aquatics, stretching, strengthening, yoga, tai chi, deep breathing and manual therapy exercises experience a decrease in pain and an improvement in symptoms (Krsnich-Shriwise, 1997; Busch, 2011).
Aerobic exercise interventions reduce pain, fatigue, and depression and improve health-related quality of life and physical fitness, while strength training is associated with large improvements in global well-being and physical function (Busch, 2011).
Self-efficacy and social support significant predictors of an overall health promoting lifestyle (Beal, 2009).
Self-management skills in conjunction with a multidisciplinary health care team can significantly improve symptoms Sarzi-Puttini, 2011).
++
No specific surgery for fibromyalgia has proven effective (Russell, 2008).
Medications should target patient's symptoms: (Russell, 2008).
Pregabalin (Lyrica): first treatment approved by FDA (FDA, 2007, Pfizer Inc., 2007); significantly reduces pain and fatigue, improves sleep and other quality-of-life measures (Crofford, 2005); improvement of pain independent of baseline anxiety and depression levels (Arnold, 2007).
Cymbalta: approved by FDA for treatment of fibromyalgia. Serotonin and norepinephrine reuptake inhibitor relieves pain and tenderness (Cymbalta, 2009; Chappell, 2009).
Acetaminophen or ibuprofen: pain relief. Non-narcotic pain relievers, low dose antidepressants, or benzodiazepines used. Tender points injected with lidocaine for local pain (NFA, 2007).
Muscle relaxants: treat pain and spasm; anticonvulsants relieve chronic pain (Mayo Clinic, 2007)
Tramodol hydrochloride (Zydol): interrupts pain messages at spinal cord level (Goldenberg, 2004).
Tricyclic antidepressants: like amitriptyline, improves quality and duration of sleep and re-establishes normal sleep patterns (Longley, 2006).
5-hydroxytryptophan at 100 mg 3 times daily (over the counter medication) to promote serotonin synthesis and increase sleep (Caruso, 1990). Fibromyalgia patients may be sensitive or intolerant to some medications; start all medications at low dose and titrate slowly for desired result (Longley, 2006).
All treatment should be multidimensional for best results (Russell, 2008).
Physical Therapy is essential along with exercise and fitness program
+++
V. Therapeutic Management
++
+++
VI. Consumer and Professional Resources
++
National Fibromyalgia & Chronic Pain Association
Education on fibromyalgia on: symptoms, diagnosis, treatment, prevalence, causes and prognosis. Provides links to research, resources and advocacy (2014).
National Fibromyalgia Research Association
Provides up-to-date research, education and treatments. Provides patients with organizations, newsletters, events and videos
National Fibromyalgia Partnership, Inc. (NFP)
Highly regarded, non-profit, educational organization whose members include FM patients and their families, as well as healthcare professionals, both research and clinical.
Offers high quality, medically-accurate information on fibromyalgia symptoms, diagnosis, treatment, and research.
Dutch Fibromyalgia Association
Education, social interaction among patients, and communication with health care workers are important when struggling with fibromyalgia since it is invisible to most (Greenan, 2009).
DFA is an international, professional organization with over 12,000 members (Greenan, 2009).
+
Arnold
L, Crofford
L, Martin
S, Young
J, Sharma
U. The effect of anxiety and depression on improvements in pain in a randomized, controlled trial of pregabalin for treatment of fibromyalgia.
Pain Med. 2007;8(8):633–638.
[PubMed: 18028041]
+
Beal
CC, Stuifbergen
AK, Brown
A. Predictors of a health promoting lifestyle in women with fibromyalgia syndrome.
Psychol Health Med. 2009;14(3):343–53.
[PubMed: 19444712]
+
Bellato
E, Marini
E, Castoldi
F, Barbasetti
N, Mattei
L, Bonasia
D, Blonna
D. Fibromyalgia syndrome: etiology, pathogenesis, diagnosis, and treatment.
Pain Res Treat. 2012;2012:426130. doi: 10.1155/2012/426130.
[PubMed: 23213512]
+
Bennet
RM, Bushmakin
AG, Cappelleri
JC, Ziateva
G, Sadosky
AB. Minimal clinically important difference in the fibromyalgia impact questionnaire.
J Rheumatol. 2009;36(6):1304–1311.
[PubMed: 19369473]
+
Brosseau
L, Wells
GA, Tugwell
P,
et al. Ottawa panel evidence-based clinical practice guidelines for aerobic fitness exercises in the management of fibromyalgia: part 1.
Phys Ther. 2008;88(7):857–871.
[PubMed: 18497301]
+
Brosseau
L, Wells
GA, Tugwell
P,
et al. Ottawa panel evidence-based clinical practice guidelines for strengthening exercises in the management of fibromyalgia: part 2.
Phys Ther. 2008;88(7):873–886.
[PubMed: 18497302]
+
Burckhardt
CS. Nonpharmacologic management strategies in fibromyalgia.
Rheum Dis Clin North Am. 2002;28(2);291–304.
[PubMed: 12122918]
+
Busch
A, Webber
S, Brachaniec
M
et al.. Exercise Therapy for Fibromyalgia.
Curr Pain Headache Rep. 2011;15(5):358–367.
[PubMed: 21725900]
+
Caruso
I, Sarzi
Puttini P, Cazzola
M, Azzolini
V. Double-blind study of 5-hydroxy-tryptophan versus placebo in the treatment of primary fibromyalgia syndrome. J Internat Med Res. 1990;18(3):201–209.
+
Castel
C, Salvat
M, Sala
J, Rull
M. Cognitive-behavioural group treatment with hypnosis: a randomized pilot trial in fibromyalgia. Contemp Hypnosis. 2009;26(1):48–59.
+
Chang
M, Hsu
J, Huang
K
et al.. Bidirectional Association Between Depression and Fibromyalgia Syndrome: A Nationwide Longitudinal Study.
J Pain. 2015;16(9):895–902.
[PubMed: 26117813]
+
Chappell
AS. A 1-year safety and efficacy study of duloxetine in patients with fibromyalgia.
Clin J Pain, 2009; 25(5):365–75.
[PubMed: 19454869]
+
Crofford
L
et al.. Pregabalin for the treatment of fibromyalgia syndrome: Results of a randomized, double-blind, placebo-controlled trial.
Arthritis Rheum. 2005;52(4):1264–1273.
[PubMed: 15818684]
+
Crofford
LJ. The hypothalamic-pituitary-adrenal axis in fibromyalgia: Where are we in 2001? J Musculoskelet Pain. 2002;10:215–220.
+
Crofford
LJ, Young
EA, Engleberg
NC, Korszun
A, Brucksch
CB, McClure
LA, Brown
MB, Demitrack
MA: Basal circadian and pul- satile ACTH and cortisol secretion in patients with fibromyal- gia and/or chronic fatigue syndrome.
Brain Behav Immun. 2004;18:314–325.
[PubMed: 15157948]
+
Dadabhoy
D, Clauw
DJ. Fibromyalgia: A Different Type of Pain Needing a Different Type of Treatment.
Nat Clin Pract Rheumatol. 2006;2(7):364–372.
[PubMed: 16932722]
+
Dogru
MT. Correlations between autonomic dysfunction and circadian changes and arrhythmia prevalence in women with fibromyalgia syndrome.
Anadolu Kardiyol Derg, 2009;9(2):110–117.
[PubMed: 19357052]
+
Foresth
K, Gran
J. Management of Fibromyalgia. Drugs. 2006;62:577–592.
+
Greenan
R. Educating patients and the struggle for validation of fibromyalgia syndrome: the Dutch way. J Musculoskel Pain. 2009;17(1):80–85.
+
Hamilton
WT, Gallagher
AM, Thomas
JM, White
PD. The prognosis of different fatigue diagnostic labels: a longitudinal survey.
Family Practice. 2005;22(4):383–388.
[PubMed: 15805126]
+
Jones
KD, Clark
SR, Bennett
RM. Prescribing exercise for people with fibromyalgia.
AACN Clinical Issues. 2002; 13:277–293.
[PubMed: 12011599]
+
Kalichman
L. Massage therapy for fibromyalgia symptoms.
Rheumatol Int. 2010;30(9):1151–1157.
[PubMed: 20306046]
+
Katz
DL, Greene
L, Ali
A, Faridi
Z. The pain of fibromyalgia syndrome is due to muscle hypoperfusion induced by regional vasomotor dysregulation.
Med Hypotheses. 2007;69:517–525.
[PubMed: 17376601]
+
Kennedy
M, Felson
DT. A prospective long-term study of fibromyalgia syndrome.
Arthritis Rheum. 1996;39(4):682–685.
[PubMed: 8630121]
+
Klyak
EK. A new nonpharmacological method in fibromyalgia: the use of wool.
J Altern Complement Med. 2009;15(4):399–405.
[PubMed: 19388862]
+
Krsnich-Shriwise
S. Fibromyalgia Syndrome: An Overview.
Phys Ther. 1997;77(1):68–75.
[PubMed: 8996465]
+
Leventhal
J. Management of fibromyalgia.
Ann Intern Med. 1999;131:850–858.
[PubMed: 10610631]
+
Longley
K. Fibromyalgia: aetiology, diagnosis, symptoms, and management.
Br J Nurs. 2006;15:729–733.
[PubMed: 16926725]
+
Mannerkorpi
K, Landin-Wilhelmsen
K, Larsson
A, Cider
Å, Arodell
O, Bjersing
J. Acute effects of physical exercise on the serum insulin-like growth factor system in women with fibromyalgia. BMC Musculoskelet Disord. 2017;18(1). doi:10.1186/s12891-017-1402-y.
+
Musekamp
G, Gerlich
C, Ehlebracht-König
I, Faller
H, Reusch
A. Evaluation of a self-management patient education program for patients with fibromyalgia syndrome: study protocol of a cluster randomized controlled trial.
BMC Musculoskelet Disord. 2016;17:1–7.
[PubMed: 26728594]
+
Nathan
C. Nitric oxide as a secretory product of mammalian cells.
FASEB J. 1992;6:3051–3064.
[PubMed: 1381691]
+
Nijs
J, Mannerkorpi
K, Descheemaeker
F, Van Houdenhove
B. Primary Care Physical Therapy in People With Fibromyalgia: Opportunities and Boundaries Within a Monodisciplinary Setting.
Phys Ther. 2010;90(12):1815–1822.
[PubMed: 20847036]
+
Pall
ML. Common etiology of posttraumatic stress disorder, fibromyalgia, chronic fatigue syndrome and multiple chemical sensitivity via elevated nitric oxide/peroxynitrite. Med Hypoth. 2001;57:139–145.
+
Peterson
E. Fibromyalgia -- management of a misunderstood disorder. J Am Acad Nurs Prac. 2007;19(7):341–348.
+
Russell
IJ. Fibromyalgia syndrome: approach to management.
CNS spectrums. 2008;13(3):27–33.
[PubMed: 18323771]
+
Sarzi-Puttini
P, Atzeni
F, Salaffi
F, Cazzola
M, Benucci
M, Mease
PJ. Multidisciplinary approach to fibromyalgia: What is the teaching?
Best Pract Res Clin Rheumatol. 2011;25(2):311–319.
[PubMed: 22094204]
+
Sherry
D, Brake
L, Tress
J
et al.. The Treatment of Juvenile Fibromyalgia with an Intensive Physical and Psychosocial Program.
J Pediatr. 2015;167(3):731–737.
[PubMed: 26209526]
+
Staud
R. Biology and therapy of fibromyalgia: pain in fibromyalgia syndrome.
Arthritis Res Ther. 2006;8(3):208. doi: 10.1186/ar1950.
[PubMed: 16684376]
+
Stucki
MO G. Physical therapy in the treatment of fibromyalgia. Scand J Rheumatol. 2000;29(113):78–85.
+
Vitorino
DF de M, Carvalho
LBC de, Prado
GF do. Hydrotherapy and conventional physiotherapy improve total sleep time and quality of life of fibromyalgia patients: Randomized clinical trial.
Sleep Medicine. 2006;7(3):293–296.
[PubMed: 16564209]
+
Watson
NF, Buchwald
D, Goldberg
J, Noonan
C, Ellenbogen
RG. Neurologic signs and symptoms in fibromyalgia.
Arthritis Rheum. 2009;60(9):2839–2844.
[PubMed: 19714636]
+
Wood
PB, Glabus
MF, Simpson
R, Paterson
JC 2nd. Changes in gray matter density in fibromyalgia: correlation with dopamine metabolism. J Pain, 2009;10(6):609–18.
++
Friedreich's Ataxia (FA) is an autosomal recessive neurodegenerative disorder (Vogel, 2014)
It is the most common inherited ataxia
1:29,000 affected by FA worldwide (Delatycki, 2000)
1:50,000 affected in the United States (Gilliland, 2012); 9,000 affected individuals at any given time (Koeppen, 2011)
Point prevalence possibly as high as 3/100,000 (Koeppen, 2011)
Estimated carrier frequency ranges from 1:50 to 1:100 in people of European, North African, Middle Eastern, and Indian descent (Koeppen, 2011).
Hereditary autosomal recessive mutation in the gene labeled FXN (Gilliland, 2012)
Nervous system changes (central and peripheral) include: loss of cells in dorsal root ganglia, degeneration in posterior columns/spinocerebellar tracts. Later: deep cerebellar nuclei, efferent pathways of the cerebellum, corticospinal tracts degenerate (Koeppen, 2011; Maring, 2007).
Average age of onset is 10 (Vogel, 2014).
The major clinical signs of Friedreich's ataxia are progressive ataxia, scoliosis, dysarthria (the most common type of speech deficiency), cardiomyopathy, diabetes mellitus, foot deformity (pes cavus), and proprioceptive loss (Vogel, 2014; McCormick 2017).
++
"Progressive, unremitting, mixed cerebellar and sensory ataxia characterizes the disease" (Massimo, 2003).
"Neurological dysfunction in patients with Friedreich's ataxia is characterized by progressive gait and limb ataxia, impairments of fine motor skills such as hand-writing and swallowing, dysarthria, lower limb areflexia, decreased vibration sense, disturbed proprioception, fatigue and muscular weakness" (Metz, 2012).
Essential (Primary) criteria for diagnosis (Santos, 2010):
Autosomal recessive inheritance
Age of onset before 25 (in rare cases, can occur as late as the 4th decade of life)
Progressive limb and gait ataxia
Absent tendon reflexes in the legs
Extensor plantar response (+Babinski sign)
Axonal sensory neuropathy
Dysarthria (if after five years from onset)
Secondary criteria for diagnosis (present in over 66%) (Santos, 2010):
Scoliosis
Pyramidal weakness in lower limbs
Absent reflexes in upper limbs
Distal loss of position and vibration sense
Abnormal ECG (cardiomyopathy)
Additional criteria for diagnosis (must be present if secondary criteria are absent) (present in less than 50%) (Santos, 2010):
Nystagmus
Optic atrophy
Deafness
Distal amyotrophy (weakness and wasting)
Pes cavus
Diabetes
Affected sibling fulfilling primary and secondary criteria
Median motor nerve conduction velocities > 40 m/s (Delatycki, 2005)
Signs/Symptoms (occur primarily between the ages of 5 and 15 years, and vary extensively among patients) (Maring, 2007; Massimo, 2003; Santos, 2010)
Cardinal sign - gait and limb ataxia (difficulty with fine motor actions)
Dysmetria, intention tremors, dysarthria, and dysphagia (usually in late stages), pes cavus and pes equinovarus; morbidity secondary to progressive equinovarus deformity (Delatycki, 2005)
Children with early FA may appear clumsy with their motor skills not matching those of unaffected siblings (Koeppen, 2011).
Scoliosis, cardiac hypertrophy associated with heart failure and sudden cardiac death in later stages (Sutton, 2013), diabetes mellitus, optic atrophy, and nephrotic syndrome are also associated with FA (Hart, 2005; Shinnick, 2016)
Neurologic Examination Findings:
Deep sensory loss, loss of position sense and vibration sense
Light touch, pain and temperature is initially normal but decreases with advanced disease
Abnormal smooth pursuit and dysmetric eye saccades are common
EKG – inverted T-waves seen in almost all patients with FA associated with cardiomyopathy accompanied by left ventricular hypertrophy
Muscle tone generally normal in arms but usually hypotonic in legs (Koeppen, 2011)
Decreased verbal fluency, working memory, and social cognition can occur in individuals with FA (Dogan, 2016)
MRI and CT scans
Used to observe abnormalities of the cerebellum and spinal cord, some of which include cerebellar blood flow, mitochondrial dysfunction and atrophy (Massimo, 2003)
MRI: The most common abnormality observed is cerebellar atrophy (thin cortex folia and large cerebellar fissures), usually most pronounced in the vermis. The volume of the cerebellum can progressively diminish as seen on sequential scans (Vedolin, 2013).
Other diagnostic tests include: nerve conductions studies, electromyogram, muscle biopsy, x-rays of the chest and spine, blood tests, and echocardiogram (NIH, 2011; NCBI, 2011; Sutton, 2013).
Rating Scales (Delatycki, 2009)
International Cooperative Ataxia Rating Scale (ICARS)
Developed by a committee within the World Federation of Neurology (Pandolfo, 2008).
A cross sectional study analyzed rate of disease progression in 603 patients with FA. It gave further validation of the ICARS and provided predictable rates of change in patients with both early and late onset FA (Metz, 2012).
Friedreich Ataxia Rating Scale (FARS)
Designed specifically for Friedreich's Ataxia and validated in 2005 (Subramony, 2005).
A screening tool useful to identify patients requiring assistive device for walking (Croarkin, 2009)
Scores > 50 tend to indicate independent walkers vs. those requiring assistance (Croarkin, 2009).
Scale for the Assessment and Rating of Ataxia (SARA)
Diagnosis of FA is confirmed by genetic testing of the chromosomal abnormality (Maring, 2007).
++
The disease continues to progress and slowly gets worse over time.
Although there is no known cure, progression of signs and symptoms can be ameliorated through surgical, pharmacological, and rehabilitative interventions. (Maring, 2007).
Heart Failure is the leading cause of death (~60%) in all FA cases (McCormick, 2017).
The median age of death in individuals with FA who die from heart failure is 26 years. The median age of death in individuals without cardiac dysfunction is 41 years (McCormick, 2017).
Intercalated Discs (ICD) in the heart can be altered in individuals with FA, this microscopic disorganization of the ICD usually precedes cardiomyopathy (Koeppen, 2016).
Most lose ambulation and require some sort of assistive aid for ADL's within 15-20 years after 1st appearance of symptoms (Gottesfeld, 2007).
Patients with FA use a WC for mobility at an average age of 25 (~15.5 yrs. after disease onset)
Symptoms tend to progress more quickly in those who present with FA onset in childhood versus those who present with onset in adulthood (Friedman, 2010).
Female patients tend to progress more quickly to aided or WC mobility than males (Maring, 2007).
Some patients with FA maintain the upper extremity coordination necessary to control a wheelchair, but others require a more specialized wheelchair that uses head controls to move the chair (De Souza, 2015)
Individuals with FA require regular review of assistive devices by an interdisciplinary team to ensure that the device fits the changing needs of the individual (De Souza, 2015)
"A direct correlation has been firmly established between the size of GAA repeats and earlier age of onset, earlier age of confinement to a WC, more rapid disease progression, and more widespread neuro-degeneration" (Massimo, 2003).
Expected sequelae include: heart disease, diabetes, scoliosis, kyphosis, pes cavus, optic atrophy, nystagmus, dysmetria, truncal ataxia, areflexia, hypotonia, and carbohydrate intolerance (NIH, 2011; Kurul, 2013).
+++
IV. Medical/Surgical Management
++
There is no cure or treatment to slow down the disease process
Current medical research is aimed at treating the lack of frataxin, mitochondrial iron accumulation, and oxidative stress (Wilson, 2012)
Cytokine recombinant human erythropoietin has been found to increase frataxin expression in various cell types from patients with FA in a dose-dependent manner especially in cardiac fibroblasts, which may protect the heart from cardiomyopathy. This is significant because the main cause of death in individuals with FA is cardiomyopathy. (Sturm, 2005).
An iron chelator, such as Deferiprone, may provide beneficial effects when used in low doses. Studies demonstrate the ability of deferiprone to remove excess iron from the central nervous system, which may result in a small but significant improvement in ataxia (Pandolfo, 2013).
Removal of excess mitochondrial iron via antioxidant or chelating medication may be attempted to protect against damage caused by free radicals (Massimo, 2003).
Idebenone, a short-chain benzoquinone has been shown to improve total, emotional, school, and social components of quality of life in patients with FA (Brandsema, 2010).
Stem cell therapy, protein replacement, and gene therapy are being researched for their abilities to correct frataxin deficits (Gottesfeld, 2007).
Surgical heart transplantation in patients with eminent heart failure has been shown to increase life expectancy by 11 years (McCormick, 2017).
Braces or surgeries have been used to treat the scoliosis and bony foot deformities that often accompany this disease (NIH, 2011).
Upcoming treatments for impairments needing more research:
Implantable ventricular assist device (VAD) –good for patients with inability to ambulate or live independently, or if heart transplant is contraindicated secondary to risks of respiratory failure and infection due to immunosuppressive therapy (Yoda, 2006).
Intrathecal baclofen in the treatment of painful, disabling spasms secondary to spinal automatism (Smail, 2005)
Combined coenzyme Q10 (CoQ10) and vitamin E therapy may lead to improvement in cardiac and muscle energy metabolism (Hart, 2005)
Additional studies concerning peripherally administered erythropoietin, histone deacetylase inhibitors, and iron-chelating therapies will need to be performed to determine their effectiveness (Mancuso, 2010).
Long-term thiamine treatment may improve neurological symptoms and up-regulate frataxin concentration. This would aid in the removal of iron from the cell (Costantini, 2016)
Liver Growth Factor has potential as a treatment option in combination with other therapies. It has been shown to have a neuroprotective effect and increase the levels of frataxin in the spinal cord and heart (Calatrava-Ferreras, 2016)
A "wearable proprioceptive stabilizer" which uses low energy vibration (less than 0.8 N) that has been converted from body heat has been shown in a pilot study to improve gait in individuals with ataxia. Scale for Assessment and Rating of Ataxia (SARA) and 6MWT improved in these individuals (Leonardi, 2016).
+++
V. Preview of Therapeutic Management
++
Physical Therapy Practice Pattern
Multidisciplinary approach:
Physical therapist, occupational therapist, neurologist, cardiologist, orthopedist, ophthalmologist, speech therapist, endocrinologist, urologist, and physiatrist (Powers, 2007)
Goals
Optimize function, minimize pain, minimize disability, minimize deformity, prolong ambulatory skills, and construct a home exercise program that can be modified every year (Powers, 2007)
Compensate for loss of coordination, sensation, and aerobic endurance while minimizing secondary complications such as joint contractures and deconditioning (Harris-Love, 2004)
Treatments include:
Prevention of weakness/atrophy via strengthening
Stretching exercises especially with patients confined to wheelchairs
Prevention of musculoskeletal deformity via splinting orthotics, supportive devices, and posture education
Recommend assistive devices such as rollators, walkers, and wheelchairs for appropriate patients
Improving cardiovascular function, individuals with FA have been shown to have a reduced cardiovascular recovery time (Bossie, 2016).
Electrical stimulation may be beneficial for acute improvements in perception of energy level in this population (Bossie, 2016).
Home management and adaptation
Support groups
Interventions to address visual, spatial, and perceptual proprioception due to the high incidence of early undetected proprioception deficits (Borchers, 2013)
Prevention of loss of balance and falls including gait training and safety education
Decline in balance is correlated with greater walking impairment and increased occurrence of falls. Individuals with FA have significantly worse limits of stability (LOS) measures using the Biodex Balance System SD. Furthermore, the participants with FA demonstrated slower gait velocity, reduced cadence, decreased time in swing phase, and increased time in stance phase (Stephenson, 2015)
Physical therapy for Friedreich's Ataxia has no specific guideline
Outcome Measures:
The Friedreich Ataxia Impact Scale (FAIS), the only patient-reported outcome tool specific to Friedreich Ataxia, was developed to examine clinically relevant areas in FA. It is made up of 126 items grouped into eight subscales, measuring three areas identified as being clinically important to individuals with the disease: symptoms, physical functioning, and psychological and social impact. The FAIS can provide insight into the patient's perspective on their health status, but is not very responsive to change over two years. Further study should be done to measure the efficacy in measuring health impact of FA (Tai, 2015).
The Barthel Index is a subjective measure that includes 10 different ADLs and the individual scores their ability and need for assistance for each task. This measure is normally used in other populations (geriatric, stroke), however it can be used in places of the FAIS because it allowed for a self-report by the individual (Bossie, 2016).
International Co-operative Ataxia Rating Scale (ICARS).
Scale for Assessment and Rating of Ataxia (SARA).
+++
VI. Consumer and Professional Resources
++
Friedreich's Ataxia Research Alliance: national, public, non-profit organization dedicated for finding a cure for Friedreich's Ataxia (FARA)
Friedreich's Ataxia for Parent's group: support group for parents of affected children with resources for living with the disease, research, fundraising, and connecting with others (FAPG)
National Ataxia Foundation: provides support groups, information about various diseases, resources, and the latest news and information (NAF)
Muscular Dystrophy Association: provides support for families, research, Quest (magazine for MDA), and also offers summer camps for children with disabilities (MDA)
Genetic and Rare Diseases Information Center (GARD) provides information about Friedreich's Ataxia that is helpful for professionals to review. It also contains links to other FA information pages.
+
Becker
E, Richardson
R. Frataxin: Its role in iron metabolism and the pathogenesis of Friedreich's ataxia. Int J Biochem Cell Biol. 2000;33:1–10.
+
Brandsema
J, Stephens
D, Hartley
J, Yoon
G. Intermediate-Dose Idebenone and Quality of Life in Friedreich Ataxia.
Pediatr Neurol. 2010;42(5):338–342.
[PubMed: 20399388]
+
Borchers
S, Synofzik
M, Kiely
E, Himmelbach
M. Routine clinical testing underestimates proprioceptive deficits in Friedreich's ataxia.
Cerebellum. 2013;12(6):916–922.
[PubMed: 23893553]
+
Bossie
HM, Willingham
TB, Schoick
RAV, O'conner
PJ, Mccully
KK. Mitochondrial capacity, muscle endurance & low energy in friedreich ataxia. Muscle Nerve. 2016. Doi:10.1002/mus.25524.
+
Calatrava-Ferreras
LCAD, Gonzalo-Gobernado
R, Reimers
D,
et al. Liver Growth Factor (LGF) Upregulates Frataxin Protein Expression and Reduces Oxidative Stress in Friedreich's Ataxia Transgenic Mice. Int J Mol Sci. 2016;17(12):2066.
+
Chakravarty
A. Friedreich's Ataxia—yesterday, today, and tomorrow.
Neurology India. 2003;51:176–182.
[PubMed: 14570998]
+
Costantini
A, Laureti
T, Pala
MI,
et al. Long-term treatment with thiamine as possible medical therapy for Friedreich ataxia.
J Neurol. 2016;263(11):2170–2178.
[PubMed: 27488863]
+
Croarkin
E, Maring
J, Pfalzer
L, Harris-Love
M, Siegel
K, DiProspero
N. Characterizing gait, locomotor status, and disease severity in children and adolescents with Friedreich ataxia.
J Neurol Phys Ther. 2009;33(3):144–149.
[PubMed: 19809393]
+
Delatycki
M. Evaluating the progression of Friedreich ataxia and its treatment.
J Neurol. 2009;256:36–41.
[PubMed: 19283349]
+
Delatycki
M, Williamson
R, Forrest
S. Friedreich ataxia: an overview.
J Med Genet. 2000;37(1):1–8.
[PubMed: 10633128]
+
Delatycki
MB
et al.. Surgery for equinovarus deformity in Friedreich's ataxia improves mobility and independence. Clin Orthop Related Res. 2005;430:138–41.
+
Dogan
I, Tinnemann
E, Romanzetti
S, Mirzazade
S, Costa
AS, Werner
CJ, Heim
S, Fedosov
K, Schulz
S, Timmann
D, Giordano
IA. Cognition in Friedreich's Ataxia: A Behavioral and Multimodal Imaging Study.
Ann Clin Transl Neurol. 2016:3(8):572–87.
[PubMed: 27606341]
+
Friedman
LS, Farmer
JM, Perlman
S, Wilmot
G, Gomez
CM, Bushara
KO, Mathews
KD, Subramony
SH, Ashizawa
T, Balcer
LJ, Wilson
RB, Lynch
DR. Measuring the rate of progression in Friedreich ataxia: implications for clinical trial design.
Mov Disord. 2010;25(4):426–432.
[PubMed: 20063431]
+
Gilliland
K, Knight
A. Friedreich's Ataxia and Gait Changes through Participation in Therapeutic Horseback Riding. Clin Kinesiol. 2012;66(1):1–6.
+
Gottesfeld
JM. Small molecules affecting transcription in Friedreich ataxia.
Pharmacol Ther. 2007;116:236–248.
[PubMed: 17826840]
+
Harris-Love
MO, Siegel
KL, Paul
SM, Benson
K. Rehabilitation Management of Friedreich Ataxia: Lower Extremity Force-Control Variability and Gait Performance.
Neurorehabil Neural Repair. 2004;18(2):117–124.
[PubMed: 15228808]
+
Koeppen
AH. Friedreich's ataxia: pathology, pathogenesis, and molecular genetics.
J Neurol Sci. 2011;303(1-2):1–12.
[PubMed: 21315377]
+
Kurul
S, Yiş
U, Güzel
A, Kasap
H, Başak
N, Dirik
E. Evaluation of the Cases with Friedreich Ataxia. Gulhane Medical Journal. 2013;55(2):123–127.
+
Leonardi
L, Aceto
MG, Marcutulli
C,
et al. A wearable proprioceptive stabilizer for rehabilitation of limb and gait ataxia in hereditary cerebellar ataxias: a pilot open-labeled study. Neurol Sci. 2016;1–5. Doi: 10.1007/s10072-016-2800-x.
+
Li
K, Reichmann
H. Role of iron in neurodegenerative diseases. J Neural Transm. January 2016. doi:10.1007/s00702-016-1508-7.
+
Mancuso
M, Orsucci
D, Choub
A, Siciliano
G. Current and emerging treatment options in the management of Friedreich ataxia.
Neuropsychiatr Dis Treat. 2010;6:491–499.
[PubMed: 20856912]
+
Maring
JR, Croarkin
E. Presentation and progression of Friedreich ataxia and implications for physical therapist examination.
Phys Ther. 2007;87:1687–1696.
[PubMed: 17911272]
+
Martin
B, Holian
A, Corben
L,
et al. Surgery for equinovarus deformity in Friedreich's ataxia improves mobility andindependence. Clin Orthop Relat Res. 2004;430:138–141.
+
Massimo,
P. Friedreich ataxia.
Seminars in Pediatric Neurology. 2003;10:163–172.
[PubMed: 14653404]
+
McCormick
A, Shinnick
J, Schadt
K,
et al. Cardiac transplantation in Friedreich Ataxia: Extended follow-up J Neurol Sci 2017. doi:10.1016/j.jns.2017.01.027.
+
Metz
G, Coppard
N, Meier
T, Metz
G, Coppard
N, Cooper
JM, Delatycki
MB, Dürr
A, Di Prospero
NA, Giunti
P, Lynch
DR, Schulz
JB, Rummey
C, Meier
T Rating disease progression of Friedreich's ataxia by the International Cooperative Ataxia Rating Scale: analysis of a 603-patient database.
Brain. 2013;136(Part 1):259–268.
[PubMed: 23365101]
+
Pandolfo,
M. Drug Insight: Antioxidant Therapy in Inherited Ataxias: Measures of Friedreich Ataxia.
Nat Clin Pract Neurol. 2008;4(2):86–96.
[PubMed: 18256680]
+
Pandolfo
M, Hausmann
L. Deferiprone for the treatment of Friedreich's ataxia.
J Neurochem. 2013;126:142–146.
[PubMed: 23859349]
+
Santos
R, Lefevre
S, Sliwa
D, Seguin
A, Camadro
J-M, Lesuisse
E. Friedreich ataxia: molecular mechanisms, redox considerations, and therapeutic opportunities.
Antioxid Redox Signal. 2010;13(5):651–690.
[PubMed: 20156111]
+
Schmitz-Hübsch
T, du Montcel
S, Klockgether
T,
et al. Scale for the assessment and rating of ataxia: development of a new clinical scale.
Neurology. 2006;66(11):1717–1720.
[PubMed: 16769946]
+
Shinnick
JE, Isaacs
CJ, Vivaldi
S, Schadt
K, Lynch
DR. Friedreich Ataxia and nephrotic syndrome: a series of two patients. BMC Neurol. 2016;16(1). doi:10.11.86/212883-016-0526-2.
+
Smail
DB, Jacq
C, Denys
P, Bussel
B. Intrathecal baclofen in the treatment of painful, disabling, spasms in Friedreich's ataxia.
Mov Disord. 2005;20:758–759.
[PubMed: 15756654]
+
Souza
LHD, Frank
AO. Rare diseases: matching wheelchair users with rare metabolic, neuromuscular or neurological disorders to electric powered indoor/outdoor wheelchairs (EPIOCs).
Disabil Rehabil. 2015;38(16):1547–1556.
[PubMed: 26714619]
+
St John
Sutton M, Ky
B, Regner
SR, Schadt
K, Plappert
T, He
J, D'Souza
B, Lynch
DR. Longitudinal strain in Friedreich Ataxia: a potential marker for early left ventricular dysfunction.
Echocardiography. 2014;31(1):50–57.
[PubMed: 23834395]
+
Stephenson
J, Zesiewicz
T, Gooch
C, Wecker
L, Sullivan
K, Jahan
I, Kim
S K. Gait and balance in adults with Friedreich's ataxia.
Gait Posture. 2015;41(2):603–607.
[PubMed: 25662043]
+
Sturm
B, Stupphann
D, Kaun
C, Boesch
S, Schranzhofer
M, Woita
J, Goldenberg
H, Scheiber-Mojdehkar
B. Recombinant human erythropoietin: effects on frataxin expression in vitro.
Eur J Clin Invest. 2005;35(11):711–717.
[PubMed: 16269021]
+
Subramony
SH, Subramony
SH, May
W, Lynch
D, Gomez
C, Fischbeck
K, Hallett
M, Taylor
P, Wilson
R, Ashizawa
T. Measuring Friedreich ataxia: interrater reliability of a neurologic rating scale.
Neurology. 2005;64:1261–1262.
[PubMed: 15824358]
+
Tai
G, Yiu
EM, Corben
LA, Delatycki
MB. A longitudinal study of the Friedreich Ataxia Impact Scale.
J Neurol Sci. 2015;352(1-2):53–57.
[PubMed: 25840637]
+
Vedolin
L, Gonzalez
G, Souza
CF, Lourenço
C, Barkovich
AJ. Inherited Cerebellar Ataxia in Childhood: A Pattern-Recognition Approach Using Brain MRI.
AJNR Am J Neuroradiol. 2013;34(5):925–934.
[PubMed: 22595899]
+
Vogel
A, Folker
J, Poole
M. Treatment for speech disorder in friedreich ataxia and other hereditary ataxia syndromes. Cochrane Db Syst Rev. 2014:10.0.1002/14651858.CD008953.
+
Wilson
RB. Therapeutic Developments in Friedreich Ataxia.
J Child Neurol. 2012;27(9):1212–1216.
[PubMed: 22791549]
+
Yoda
M, El-Banayosy
A, Arusoglu
L,
et al. Permanent use of a ventricle assist device for dilated cardiomyopathy in Friedreich's ataxia.
J Heart Lung Transplant. 2006;25(2):251–256.
[PubMed: 16446231]
+++
Guillain-Barré Syndrome
++
A syndrome, including multiple debilitating features, related to myelin damage and axon destruction in peripheral nerves resulting in impairment in motor-unit recruitment due to transmission problem. As a result, there are fewer numbers of successfully recruited muscle fibers available to produce sufficient forces for functioning (Basille, 1996)
Incidence and Prevalence
Rare with 1 to 2 cases per 100,000 individuals (McGrogan, 2009).
The rate of incidence is almost 3 cases per 100,000 among those in their ninth decade of life, and males have a higher incidence than women (Sejvar, 2011).
Peak incidence in Australia is between 55 and 65 years of age, with no difference in the age of onset between males and females (Blum, 2013).
Very little evidence regarding incidence in Asian and African countries (McGrogan, 2009).
GBS is rare in children, with an incidence of 0.6 cases per 100,000 children (Watson, 2014).
Subtypes of Guillain-Barré
Pathogenesis
Considered an autoimmune disease: Lymphocyte-mediated autoimmune reaction
Often preceded by an infection 1-3 weeks prior
Swedish study: 70% of 53 subjects had at least one preceding event one month before symptoms.
47% respiratory infections (cytomegalovirus, Mycoplasma pneumoniae) including recent viral infections like influenza (Haemophilus influenza), Epstein-Barr virus, cytomegalovirus and M. pneumoniae (Seneviratne, 2000; Rajabally, 2013) and Zika Virus (Watrin, 2016).
11% gastrointestinal infection (Cheng, 2000) including recent bacterial infections such as campylobacter jejuni (Seneviratne, 2000)
According to a systematic review, 70% of cases reported were preceded by either a gastrointestinal or respiratory infection (McGrogan, 2009; Sudalagunta, 2015)
German study: 82% of 95 children had previous infections (most common being Coxsackie virus) (Schessl, 2006)
40% of patients experienced no precipitating event leading up to the disease (Lugg, 2010)
Seasonally higher rates of occurrence in winter than in summer, perhaps due to more frequent upper respiratory infections in winter months (Blum, 2013; Sudalagunta, 2015).
Also evidence of increased incidence following surgery and vaccination
Popular theories:
Less popular theories:
++
Rapidly ascending symmetrical motor weakness and sensory impairments typically beginning distally and moving proximally with weakness and deep tendon reflex loss
Motor weakness doesn't usually last more than 4 weeks (Sudulagunta, 2015).
Signs and Symptoms (Eldar, 2014)
Motor Impairments in affected areas
Ascending weakness of symmetrical limbs, neck muscle, respiratory muscles
Areflexia
Atrophy of limb muscles
Ataxia
GBS is a common cause of acute weakness in children (Talebian, 2016; Roodbol, 2010).
Sensory Impairments
Numbness, paresthesias
Loss of kinesthesia, vibration, and touch
Paresthesia of the toes is often the first neurologic symptom
Cranial Nerve Palsies in more severe cases (if it progresses that high)
Especially bulbar, facial, and occulomotor nerves
About 50% of people with GBS will develop cranial neuropathy (Talebian, 2016)
The facial nerve is the most common cranial nerve involved in GBS (Talebian, 2016).
Delayed facial weakness is the appearance of facial palsy with an onset after the peak of limb weakness or ataxia, seen in 54% of patients with AIDP and 23% of patients with AMAN developed facial weakness (Tatsumoto, 2015).
10% of patients with GBS have ocular muscle weakness (Talebian, 2016).
Autonomic dysfunction
Variable blood pressure and heart rate disturbances often occur in those with GBS; 3-10% die presumably due to sudden autonomic failure (Samadi, 2013).
Sinus tachycardia and bradycardia, HTN, postural hypotension, fluctuations in pulse and blood pressure
Abnormal vasomotor tone causing venous pooling and facial flushing
Hypertension was also common in childhood GBS, with 44% of children requiring treatment (Watson, 2014).
Bowel/bladder dysfunction
Urinary sphincter disturbances
Constipation
Bladder dysfunction is common in childhood GBS with the presence of urinary retention due to associated paralysis (Watson, 2014).
Others
Pain, tonic pupils, anhydrosis, papilloedema
Ptosis without opthalamoplegia is very rarely the first manifestation of GBS. Therefore, the diagnosis of GBS should be considered with isolated ptosis and ptosis with ascending/descending paralysis (Talebian, 2016).
30% of cases of GBS progress to respiratory failure requiring Endotracheal Mechanical Ventilation (ETMV) (Bilan, 2015).
Patients with paraparetic vs quadriparetic GBS have less frequent cranial nerve deficits (van den Berg, 2014)
Diagnostic Criteria
Other Laboratory Tests:
Antibodies directed against gangliosides GM1, GD1a, GM1b, GalNAcGD1a;
Stool culture for C. jejuni; antibodies to C. jejuni;
Viruses including: cytomegalovirus, Epstein-Barr Virus, Herpes Simplex Virus, HIV, M pneumoniae;
Biochemical screening and testing for:
Disease severity can be quantified using the GBS Disability Scale (Blum, 2013)
Normal=0
Minimal signs and symptoms= 1
Able to walk without assistance= 2
Able to walk without assistance for 5 meters= 3
Bed or chair bound= 4
Requires ventilator support= 5
Death= 6
++
Prognosis and outlook for people with Guillain-Barre Syndrome is generally good with high likelihood of recovery (Kahn, 2010; van den Berg, 2014) However, patients who require mechanical ventilation typically have increased mortality, complications, and poor prognosis (Witsch, 2012).
Progression
After a few weeks, the demyelination process usually stops and remyelination of the axons begins.
Progression can cease at any body level (ankle weakness only to involvement of whole body, cranial nerves, and respiration)
Phases
Acute phase: Onset of symptoms with rapid progression until no more deterioration occurs, lasting up to 4 weeks; first neurologic symptom is paresthesia in toes/fingers prior to progressive paralysis
Plateau phase: Symptoms constant from acute phase and do not change; days to weeks
Recovery phase: patient's condition begins to improve; lasts few weeks to 2 years
Prognosis
Most recover completely but it may take weeks to years to fully recover (NINDS, 2011)
Patients with paraparetic GBS show a faster and more complete recovery than those with quadriparetic GBS (van den Berg, 2014).
20%-30% of patients have reported a reduced physical functioning status long-term. (Forsberg, 2015)
30% of individuals are left with residual weakness (NINDS, 2011)
Severe fatigue is a frequently reported long-term symptom (Garssen, 2006).
At 2 years, 50% of subjects showed impairments in motor and sensory areas (Forsberg, 2004)
There is a 10% mortality rate in patients with GBS and 20-25% have a resulting disability following recovery (Kuwabara, 2004)
Outcomes are related to the patient's status at the time of admittance, including older age, decreased strength, presence of diarrhea prior, and speed of symptom development (Rajabally, 2013).
Factors that predict a poor outcome include:
Poor prognosis if > 40-50 years of age, rapidly progressive disease, axonal loss, and extended mechanical ventilation (higher levels)
A longer period before recovery begins
5%-10% patients die in acute phase (especially old age, cardiac disease, pulmonary embolism, or respiratory infection)
Mortality rate varies by source but all agree that it is low for most individuals (Hiraga, 2005; NINDS, 2011)
+++
IV. Medical Treatment
++
Main goal: decrease disease severity, duration, and pain incurred by patient
Medical emergency: ICU for monitoring and mechanical ventilation if needed
Medical Treatments:
Corticosteroids: ineffective alone. However, pain may be managed via different drugs such as methylprednisolone or corticosteroids in some cases, because pain is caused by many factors (Ruts, 2007).
Intravenous (IV) immunoglobulin (Ig): found to be safe and effective in GBS treatment; 5 to 10% of GBS patients deteriorate after initial improvement or stabilization following IV Ig treatment (Van Doorn, 2014)
Plasmapheresis: within first 2 weeks, reduced hospitalization, mechanical ventilation, and time to reach ambulation
CSF filtration: more research needed
Mechanical ventilation may be used during the most severe phases of GBS for assistance with breathing (Witsch, 2012).
Experimental treatments: nerve growth factors, combined use of steroids and IV Ig, B-interferon, and repeated doses of IV Ig
+++
V. Implications for Therapeutic Management
++
Possible Preferred Practice Patterns (APTA, 2017)
5F: Impaired Peripheral Nerve Integrity and Muscle Performance Associated with Acute or Chronic Polyneuropathies
5G: Impaired Motor Function and Sensory Integrity Associated with Acute or Chronic Polyneuropathies
6B: Impaired Aerobic Capacity/Endurance Associated with Deconditioning
6E: Impaired Ventilation and Respiration/Gas Exchange Associated with Ventilatory Pump Dysfunction or Failure
7A: Primary Prevention/Risk Factors Reduction for Integumentary Disorders
Physical therapy has been shown to be beneficial to producing good outcomes for those who receive acute care at the onset of the syndrome (Davidson, 2009; Dennis, 2013)
Role of the PT:
Three parameters physical therapist should use when retraining/exercising these patients (Basille, 1996)
Recruited muscle fibers are at risk for overworking. Assessment for overwork weakness is essential. (Basille, 1996)
As a precaution in early treatment, provide gentle stretching and AAROM exercise at the level that is consistent to the person's strength. Overstretching and overuse can result in prolonged recovery time.
Educate patient and family regarding what to expect and how to deal with the effects of residual neuropathy (de la Cour, 2005)
Acute and Ascending Phase
Physical therapy initiated early to maintain the patient's available joint ROM and muscle strength during the initial onset of the syndrome.
Important to understand patient may become fatigued easily or may have a low pain tolerance
Therapy focus: prevent complications with immobilization
Prevention of skin breakdown:
PTs also need to be aware of the risk of DVTs due to lack of mobility of some patients (Hughes, 2005)
Prevention of muscle and joint pain: PTs can provide appropriate splinting to help alleviate joint and muscle pain.
Cardiopulmonary monitoring is important due to severe cases causing respiratory dysfunction (in 25%) or failure of the respiratory muscles (Van Doorn, 2014)
Arterial gas measurements (PO2). Signs of decreasing PO2 such as hypoxia.
Encourage clearing of airways, place patient in sidelying if needed.
Prevention of orthostatic hypotension: Elevate the head of the bed 30 degrees to decrease the risk or orthostatic hypotension when getting up from bed
Some patients may have catheters (Hughes, 2005)
Therapists must follow universal precautions to prevent respiratory infections
Therapy in pool or Hubbard tank can be useful for beginning movement immediately after acute phase (during the ascending phase)
Descending Phase
Paralysis is receding with returning physical function. The patient can begin neuromuscular facilitation techniques
Treatment and Prevention of fatigue: endurance and strength training (in the resolving phase) (Garssen, 2004; ElMhandi, 2007), active isometric and isotonic exercises but not to the point of fatigue
Promotion of functional training: incorporate assistive devices and AFOs
High intensity exercise was shown to be more beneficial than low intensity exercise in patients in the chronic phase of recovery of GBS in reducing disability (Khan, 2011)
+++
VI. Consumer and Professional Resources
++
There are many websites and support groups that explain the syndrome and how to help those who have suffered from it.
GBS/CIDP Foundation International
http://www.gbs-cidp.org/
For those who have been diagnosed with GBS/CIDP as well for health professionals who want to know more about the disease process
Includes
Location of local chapters or communities for support
Information for friends or loved ones of those suffering from GBS/CDIP
Information about the disease and about insurance coverage and treatment for health professionals
Information about research being done for the treatment of GBS/CDIP
Document17 http://www.gbs-cidp.org/wp-content/uploads/2012/01/PTOTGuidelines.pdf
CIDP USA
A support group with a site that has information about all autoimmune diseases with a focus on GBS. It has information on things like diet and things that individuals can try at home to help them cope.
http://www.cidpusa.org/gbs.html
DailyStrength GBS Support group
Guillain-Barre Support
MDJunction
Medscape
A website geared more toward the professional medical team member. This website is geared by WebMD, LLC and comes with a disclaimer stating the contained information for the use of professionals. It includes general GBS statistics, information, and rehabilitation needs.
http://emedicine.medscape.com/article/315632-treatment#aw2aab6b6b3
US National Library of Medicine National Institutes of Health
+
Atkinson
SB, Carr
RL, Maybee
P, Haynes
D. The challenges of managing and treating guillain-barré syndrome during the acute phase.
Dimens Crit Care Nurs. 2006; 25(6):256–263.
[PubMed: 17122655]
+
Bassile,
CC. Guillain-Barre syndrome and exercise guidelines. Neurology Report. 1996;20:31–36.
+
Bersano
A, Fiorini
M, Allaria
S
et al.. Detection of CSF 14-3-3 protein in guillain-barré syndrome.
Neurology. 2006; 67:2211–2216.
[PubMed: 17190946]
+
Bilan
N, Barzegar
M, Habibi
P. Predictive factors of respiratory failure in children with Guillain-Barre Syndrome. Int J Pediatr. 2015:3(15):33–37.
+
Blum
S, Reddel
S, Spies
J, McCombe
P. Clinical features of patients with Guillain-Barré syndrome at seven hospitals on the East Coast of Australia. J Peripher Nerv Syst. 2013.
+
Bowyer
H, Glover
M. Guillain-Barre Syndrome: Management and Treatment Options for Patients with Moderate to Severe Progression. J Neurosci Nurs 2017;42(5):288–293.
+
Cheng
Q
et al.. Clinical epidemiology of guillain-barré syndrome in adults in Sweden 1996-97: a prospective study.
European Journal of Neurology. 2000; 7:685–692.
[PubMed: 11136356]
+
Davidson
I, Wilson
C, Walton
T, Brissenden
S. Physiotherapy and Guillain-Barré syndrome: results of a national survey. Physiotherapy [serial online]. September 2009;95(3):157–163. Available from: MEDLINE with Full Text, Ipswich, MA. Accessed January 26, 2012.
+
De la Cour
CD, Jakobsen
J. Residual neuropathy in long-term population-based follow-up of guillain-barre syndrome.
Neurology, 2005; 64:246–253.
[PubMed: 15668421]
+
Dennis
D, Mullins
R. Guillain-Barre syndrome patient's satisfaction with physiotherapy: A two-part observational study.
Physiother Theory Pract. 2013;29(4):301–308.
[PubMed: 23126501]
+
Derksen
A, Ritter
C, Lehmann
H, Athar
P, Kieseier
B, Mancias
P, Hartung
H, Sheikh
K. Sural sparing pattern discriminates Guillain-Barré syndrome from its mimics. Muscle Nerve. November 2014;50(5):780–784.
+
Elings
J
et al.. Physiotherapy in patients with acute and chronic inflammatory polyneuropathies: a survey of clinical practice among physiotherapists associated with the dutch organization for neuromuscular diseases [Dutch]. Ned Tijdschr Fysyother, 2009; 119(1): 10–6
+
Eldar
AH, Chapman
J. Guillain Barré syndrome and other immune mediated neuropathies: Diagnosis and classification. Autoimmun Rev. 2014. doi:10.1016/j.autrev.2014.01.033.
+
El Mhandi
L, Calmels
P, Camdessanche
J, Gautheron
V, Feasson
L. Muscle strength recovery in treated Guillain-Barre syndrome: a prospective study for the first 18 months after onset.
Am J Phys Med Rehabil, 2007; 86:716–724.
[PubMed: 17709995]
+
Forsberg
A, Press
R, Einarsson
U, de Pedro-Cuesta
J, Holmqvist
LW. Impairment in guillain-barré syndrome during the first 2 years after onset: a prospective study.
Journal of the Neurological Sciences. 2004; 227:131–138.
[PubMed: 15546603]
+
Forsberg
A, Widén-Holmqvist
L, Ahlström
G. Balancing everyday life two years after falling ill with Guillain-Barré syndrome: a qualitative study.
Clinical Rehabilitation. 2015;29(6):601–610 10p.
[PubMed: 25200880]
+
Fisher
T, Stevens
J. Rehabilitation of a marathon runner with guillain-barre syndrome. Journal of Neurologic Physical Therapy. 2008; 32(4): 203–209.
+
Frenzen
PD. Economic cost of guillain-barré syndrome in the United States.
Neurology. 2008; 71(1): 21–7.
[PubMed: 18591502]
+
Garssen
M, Bussman
J, Schmitz
P, Zandbergen
A, Welter
T, Merkies
I, Stam
H, van Doorn
P. Physical training and fatigue, fitness, and quality of life in Guillain-Barre syndrome and CIDP.
Neurology, 2004; 63:2393–2395.
[PubMed: 15623709]
+
Garssen
MPJ, Schmitz
PIM, Merkies
ISJ, Jacobs
BC, Meche
FGA, Doorn
PA. Amantadine for treatment of fatigue in Guillain-Barré syndrome: a randomised, double blind, placebo controlled, crossover trial. J Neurology Neurosurg Psychiatry. 2006;77(1):61–65.
+
Hiraga
A, Mori
M, Kuwabara
S,
et al. Recovery patterns and long term prognosis for axonal Guillain-Barré syndrome. Journal Of Neurology, Neurosurgery & Psychiatry [serial online]. May 2005;76(5):719–722. Available from: CINAHL Plus with Full Text, Ipswich, MA. Accessed January 25, 2012.
+
Kaida
K
et al.. GM1/GalNAc-GD1a complex: a target for pure motor guillain-barre syndrome. Neurology, 2008 .18;71 (21): 1683–90.
+
Khan
F, Pallant
J, Amatya
B, Ng
L, Gorelik
A, Brand
C. Outcomes of high- and low-intensity rehabilitation programme for persons in chronic phase after Guillain-Barré syndrome: A randomized controlled trial.
J Rehabil Med. 2011;43(7):638–646.
[PubMed: 21667009]
+
Kuitwaard
K, Koningsveld
R, Ruts
L, Jacobs
B, Doorn
P. Recurrent Guillain-Barre syndrome. Journal of Neurology, Neurosurgery and Psychiatry. 2009; 80:56–59.
+
Kuwabara
S. Guillain-Barre Syndrome Epidemiology, Pathophysiology, and Management. Drugs. 2014;64(5):1–15.
+
Lugg
J. Recognising and managing Guillain-Barré syndrome. Emerg Nurse. June 2010;18(3):27–30.
+
Malekian
A, Barzegar
M, Malaki
M, Shiva
S. Different subtypes of Guillane-Barré syndrome in children. J Of Pediatr Neurol. July 2013;11(3):159–163.
+
McGrogan
A, Madle
GC, Seaman
HE, De Vries
CS. The Epidemiology of Guillain-Barre Syndrome Worldwide.
Neuroepidemiology. 2009;32(2):150–163.
[PubMed: 19088488]
+
Petzold
A, Hinds
N, Murray
N, Hirsch
N, Grant
D, Keir
G, Thompson
E, Reilly
M. CSF neurofilament levels: A potential prognostic marker in Guillain-Barre syndrome. Neurology, 2006; 67:1071–1073.
+
Rajabally
YA, Uncini
A. Outcome and its predictors in Guillain–Barré syndrome.
J Neurol Neurosurg Psychiatry. 2012;83(7):711–718.
[PubMed: 22566597]
+
Ruts
L, Koningsveld
R, Jacobs
BC, Doorn
PA. Determination of pain and response to methylprednisolone in Guillain-Barré syndrome.
Journal of Neurology. 2007;254(10):1318–1322.
[PubMed: 17426908]
+
Samadi
M, Kazemi
B, Oskoui
S, Barzegar
M. Assessment of Autonomic Dysfunction in Childhood Guillain-Barré Syndrome. J Cardiovasc Thorac Res. September 2013;5(3):81–85.
+
Schessl
J, Luther
B, Kirschner
J, Mauff
G, Korinthenberg
R. Infections and vaccinations preceding childhood guillain-barré syndrome: a prospective study.
European Journal of Pediatrics. 2006; 165(9):605–612.
[PubMed: 16691408]
+
Sejvar
JJ, Baughman
AL, Wise
M, Morgan
OW. Population incidence of Guillain-Barré syndrome: a systematic review and meta-analysis.
Neuroepidemiology. 2011;36(2):123–133.
[PubMed: 21422765]
+
Sudulagunta
SR, Sodalagunta
MB, Sepehrar
M, Khorram
H, Bangalore
Raja SK, Kothandapani
S, Noroozpour
Z, Aheta
Sham M, Prasad
N, Sunny
SP, Mohammed
MD, Gangadharappa
R, Nidsale
Sudarshan R. Guillain-Barré syndrome: clinical profile and management. Ger Med Sci. 2015;13(16):1–15.
+
Talebian
A, Soltani
B, Talebian
M. Bilateral Ptosis as the First Presentation of Guillain-Barre Syndrome. Iranian Journal of Child Neurology. 2016;10(1):70–73 4p.
[PubMed: 27057192]
+
Tatsumoto
M, Misawa
S, Kokubun
N, Sekiguchi
Y, Hirata
K, Kuwabara
S, Yuki
N. Delayed facial weakness in Guillain-Barré and miller fisher syndromes.
Muscle Nerve. 2015;51(6):811–814.
[PubMed: 25287079]
+
Tatsumoto
M, Koga
M, Gilbert
M, Odaka
M, Hirata
K, Kuwabara
S, Yuki
N. Spectrum of neurological diseases associated with antibodies to minor gangliosides GM1b and GalNAc-GD1a.
Journal of Neuroimmunology, 2006; 177:201–208.
[PubMed: 16844234]
+
Van den Berg
B, Fokke
C, Drenthen
J, van Doorn
P, Jacobs
B. Paraparetic Guillain-Barre syndrome.
Neurology. 2014;82(22):1984–1989.
[PubMed: 24808021]
+
Van Doorn
PA. Diagnosis, treatment and prognosis of Guillain-Barré syndrome (GBS).
Presse Med. 2013;42(6 Pt 2):e193–201.
[PubMed: 23628447]
+
Van Doorn
P, Ruts
L, Jacobs
B. Clinical features, pathogenesis, and treatment of Guillain-Barré syndrome.
Lancet Neurol.2008;7(10):939–950.
[PubMed: 18848313]
+
Watrin
L, Ghawché
F, Larre
P, Neau
J, Mathis
S, Fournier
E. Guillain–Barré Syndrome (42 Cases) Occurring During a Zika Virus Outbreak in French Polynesia.
Medicine. 2016;95(14):e3257.
[PubMed: 27057874]
+
Watson
L, Aziz
M, Vassallo
G, Plant
N, Webb
N. Bladder dysfunction and hypertension in children with Guillain-Barre syndrome.
Pediatr Nephrol. 2014;29(9):1637–1641.
[PubMed: 24651942]
+
Willison
H. Gangliosides as targets for autoimmune injury to the nervous system.
Journal of Neurochemistry, 2007; 103:143–149.
[PubMed: 17986149]
+
Witsch
J, Galldiks
N, Bender
A, Kollmar
R, Bosel
J, Hobolm
C, Gunther
A, Schirotzek
I, Fuchs
K, Juttler
E. Long-term outcome in patients with Guillain–Barré syndrome requiring mechanical ventilation [published online ahead of print January 9, 2013].
Journal of Neurology. 2013:1–8.
http://link.springer.com.libproxy2.usouthal.edu/article/10.1007/s00415-012-6806-x/fulltext.html. Accessed January 31, 2013.
+
Ye
Y, Wang
K, Deng
F, Xing
Y. Electrophysiological subtypes and prognosis of Guillain–Barré syndrome in Northeastern China.
Muscle & Nerve. 2013;47(1):68–71.
[PubMed: 23042578]
+++
High Risk Infant / Prematurity (Intraventricular Hemorrhage)
++
Intraventricular hemorrhage (IVH) is the most common form of intracranial hemorrhage in infants born preterm. In the United States, approximately 12,000 infants born prematurely develop an IVH (Ballabh, 2010).
Globally, 20-40% of premature infants (birth weight less than 1,500 grams) have been observed to have IVH (El-Atawi, 2016), although the incidence decreased from a high of 50.9% in 1991 to a low of 11.9% in 2005 in one cohort study (Marba, 2011).
There is a direct correlation between the extent of prematurity and the severity of IVH (Shooman, 2009).
++
++
IVH occurs in the periventricular germinal matrix located on the head of the caudate nucleus and underneath the ventricular ependyma, and is a highly metabolically active site of neuronal proliferation and differentiation during fetal development (Ballabh, 2010).
++
++
IVH is attributed to multifactorial involvement including: immaturity of the germinal matrix vasculature, disturbance of cerebral blood flow, and platelet and coagulation disorders.
The periventricular germinal matrix is highly vascularized, with a very fragile and primitive capillary network stemming from the Heubner artery and lateral striate arteries, with venous drainage through the terminal vein that leads into the internal cerebral vein and vein of Galen (Wildrick, 1997).
Infants delivered before 36 weeks gestational age are more vulnerable to IVH because the intact primitive germinal matrix has fragile, irregular, thin-walled vessels susceptible to slight changes in cerebral blood flow (Allen, 2013).
The neonate's autoregulatory abilities are proportional to the gestational age at time of birth. Thus, a premature baby is more susceptible to IVH with a loss in the body's ability to regulate systemic blood pressure (BP) (Wildrick, 1997).
Loss of systemic BP regulation coupled with rapid changes in cerebral blood flow (due to respiratory distress syndrome, ventilator support, tracheal suctioning, systemic hyper- or hypotension, hypoxic events, patent ductus arteriosus, seizures, transfusions, coagulation, or metabolic disorders) can overwhelm the neonate and result in grades I, II, or III hemorrhage (Wildrick, 1997; Allen, 2013; Ballabh, 2010).
Grade IV hemorrhages result primarily from increased venous pressure that develops from the progression of lower-grade hemorrhages and are referred to as periventricular hemorrhagic infarctions (PHI) (El-Atawi, 2016).
Grading of IVH (El-Atawi, 2016; Ahn, 2015; Wildrick, 1997; Volpe, 1995):
++
++
Clinical presentation: 1) Silent: asymptomatic; 2) Saltatory: evolves more slowly over hours to days after birth with gradual alteration in consciousness with spontaneous movement, hypotonia, respiratory distress, and changes in eye movements and position; and 3) Catastrophic: evolves within minutes to hours after birth with signs including seizures, stupor or coma, hypoventilation, decerebrate posturing, bulging anterior fontanelle, muscular weakness and usually has poor outcome (Ballabh, 2010; Robinson, 2012).
Neuropathological patterns in premature infants: periventricular leukomalacia (PVL), diffuse white matter gliosis (DWMG), neuronal-axonal injury of white and gray matter, germinal matrix hemorrhage-IVH (GMH-IVH), and periventricular hemorrhagic infarction (Hinojosa-Rodriguez, 2017).
++
++
Routine screening with cranial ultrasonography of all infants <30 weeks gestation between 7-14 days of age and repeated between 36-40 weeks gestation should be done to detect IVH, PVL, and low- pressure ventriculomegaly (Ment, 2002; McCrea, 2008).
Transcranial doppler ultrasonography: Non-invasive tool to monitor cerebral circulation.
Near infra-red spectroscopy: Real-time monitoring of cerebral oxygenation; non-invasive measure of changes in oxygenation, cerebral blood flow and imaging of brain activity in the neonate
Advanced magnetic resonance imaging (MRI) techniques: MR spectroscopy is helpful in measurement of the neurodevelopmental outcomes; functional MRI measures neuronal activity in different parts of the brain in response to various tasks; susceptibility-weighted imaging detects paramagnetic substances and is an accurate detector of small hemorrhages (Panigrahy, 2012; Smyser, 2013; Intrapiromkul, 2013; El-Atawi, 2016).
Biomarkers: the following biomarkers have shown promise in recent studies for early diagnosis of IVH. Activin A is a growth factor that plays a role in the physiologic response to brain injury. Preterm neonates with IVH have increased activin levels. S100b is a predictor of IVH and neonatal mortality. IL-6 and erythropoietin are also potential biomarkers for IVH (Andrikopoulou, 2014; Douglas-Escobar, 2013).
+++
III. Prognosis & Sequelae
++
++
++
++
Premature birth accounts for 45% of all children with CP, 35% with visual impairment, and 25% with cognitive or hearing impairment (Allen, 2008; Jarjour, 2015).
Cystic PVL secondary to PHI: cerebral palsy (CP) with diplegia or quadriplegia depending on location of infarct; motor cortex leads to hemiplegic CP (Hinojosa-Rodriguez, 2017).
Diffuse cystic PVL with moderate to severe white matter abnormalities (WMA): cognitive impairment/behavioral deficits (Hinojosa-Rodriguez, 2017).
Non-cystic PVL with mild to moderate WMA: cognitive impairment/behavioral deficits (Hinojosa-Rodriguez, 2017).
Neuronal loss and gliosis of gray matter: cognitive impairment/behavioral deficits (Hinojosa-Rodriguez, 2017).
Breaking of the germinal matrix vessels: sequelae depends on location and severity; Grade III/PVH leads to CP, cognitive, behavioral, and visual deficits (Hinojosa-Rodriguez, 2017).
PVL combined with neuronal and axonal loss: CP and autism spectrum disorders, motor, cognitive, attentional, behavioral, and socialization deficits (Hinojosa-Rodriguez, 2017).
+++
IV. Medical/Surgical Management
++
++
Manage BP, intracranial pressure, coagulation disorders, and respiratory stress to minimize complications of continued or reoccurrence of hemorrhage (El-Atawi, 2016).
Prevention of IVH is the primary goal for the health care team; the best way to prevent IVH is to prevent premature births. This may not always be possible, and thus the medical team should screen all babies weighing under 1800 grams at birth for IVH (Wildrick, 1997).
The use of Indomethacin to reduce the incidence is controversial, as one study has shown that the perinatal use of indomethacin was not associated with improved outcomes in very low birth weight infants and another showed a reduction in severe IVH (Luque, 2014; Fowlie, 2010).
Antenatal corticosteroids increase survival rate and reduce rate or IVH in children born prematurely. Vitamin K and antenatal corticosteroids decrease the incidence of IVH (Shooman, 2009; Wong, 2014).
Prevention: optimal peripartum management including resuscitation, body temperature > 36 degrees C, optimal surfactant delivery, minimize pain and stress through developmental care and minimal handling, neutral head position, fluid volume therapy for hypotension, indomethacin prophylaxis first 24 hours after birth, adequate ventilation, avoid routine suctioning, and limit use of sodium bicarbonate and postnatal dexamethasone (Whitfield, 2001; Mclendon, 2003; El-Atawi, 2016).
++
++
Ventricular peritoneal (VP) shunting for treatment of post-hemorrhagic hydrocephalus (PHH). Complications include poor long-term outcomes, shunt infections, and shunt obstruction leading to failure. These complications have led to development of temporary options to allow the infant time to mature. Ventricular reservoirs and ventriculo-subgaleal shunts are temporary cerebrospinal fluid diverters (El-Atawi, 2016).
Researchers are currently working on a developing fibrinolytic therapy to offset the grow factors (tPA) that are present in posthemorrhagic CSF (Whitelaw, 2003).
+++
V. Key Aspects of Therapeutic Management
++
Possible Preferred Practice Pattern: Pattern 5C: Impaired Motor Function and Sensory Integrity Associated with Non-Progressive Disorders of the Central Nervous System— Congenital Origin or Acquired in Infancy or Childhood (APTA, 2014)
In the neonatal intensive care unit (NICU), great care should be taken to properly manage the infant and the environment to avoid unnecessary stressors. The entire NICU experience affects brain development and all intervention in the NICU is brain care (Als, 2004).
Early intervention for the infant born prematurely can aide in more regulated brain maturation and recovery attributed to neuroplasticity of the neonatal brain. Developmentally appropriate interventions when the infant is deemed receptive and physiologically ready may aid in decreasing long-term functional impairment (Santos, 2015; Bodkin, 2003).
Developmental interventions that focus on the parent-infant relationship and infant development have shown better outcomes for preterm infants. Interventions geared towards responsive parenting to support conditions for child development within the family unit have better cognitive outcomes (Spittle, 2016; van Wassenaer-Leemhuis, 2016).
Parent-administered physical therapy promoting postural control, head control, and midline orientation under the supervision of a physical therapist showed greater improvement in motor performance at 37 weeks for infants born < 32 weeks compared to conventional care (Ustad, 2016).
Physical therapists working within a model that incorporates the parent/child dyad show improved motor outcomes for preterm infants (Wu, 2014).
Positioning to: "(1) support posture and movement; (2) optimize skeletal development and biomechanical alignment; (3) provide controlled exposure to varied proprioceptive, tactile, and visual stimuli; and (4) promote calm, regulated behavioral state" (Sweeney, 2002).
Outcome measures: Test of Infant Motor Performance (TIMP), Neonatal Individualized Developmental Care and Assessment Program (NIDCAP), Qualitative Assessment of General Movement of Infants (GMA), The Newborn Behavioral Observations (NBO) System Handbook, Neonatal Infant Pain Scale (NIPS), The Premature Infant Pain Profile (PIPP).
+
Ahn
SY, Shim
SY, Sung
IK. Intraventricular hemorrhage and post hemorrhagic hydrocephalus among very-low-birth-weight infants in Korea.
J Korean Med Sci. 2015;30(Suppl 1):S52–58.
[PubMed: 26566358]
+
Allen
KA. Treatment of intraventricular hemorrhages in premature infants: where is the evidence?
Adv Neonatal Care. 2013;13(2):127–130.
[PubMed: 23532032]
+
Allen
MC. Neurodevelopmental outcomes of preterm infants.
Curr Opin Neurol. 2008;21(2):123–128.
[PubMed: 18317268]
+
Als
H, Duffy
F, McAnulty
GB,
et al. Early experience alters brain function and structure.
Pediatrics. 2004;113(4):846–857.
[PubMed: 15060237]
+
American Physical Therapy Association. Guide to Physical Therapist Practice 3.0. Alexandria, VA: American Physical Therapy Association; 2014. Available at:
http://guidetoptpractice.apta.org/. Accessed December 29, 2017.
+
Andrikopoulou
M, Almalki
A, Farzin
A, Cordeiro
CN, Johnston
MV, Burd
I. Perinatal biomarkers in prematurity: early identification of neurologic injury.
Int J Dev Neurosci. 2014;36:25–31.
[PubMed: 24768951]
+
Ballabh
P. Intraventricular hemorrhage in premature infants: mechanism of disease.
Pediatr Res. 2010;67(1):1–8.
[PubMed: 19816235]
+
Bodkin
AW, Baxter
RS, Heriza
CB. Treadmill training for an infant born preterm with a grade III intraventricular hemorrhage.
Phys Ther. 2003;83:1107–18.
[PubMed: 14640869]
+
Douglas-Escobar
M, Weiss
MD. Biomarkers of brain injury in the premature infant. Front Neurol. 2012;3:e185. doi: 10.3389/fneur.2012.00185.
+
El-Atawi
K, Elhalik
M, Kulkarni
T, Abdelsamed
A, Alexander
L, Satyan
AD. Risk factors, diagnosis, and current practices in the management of intraventricular hemorrhage in preterm infants: a review. Acad J Pediatr Neonatol. 2016;1(3):001–007.
+
Fowlie
PW, Davis
PG, McGuire
W. Prophylactic intravenous Indomethacin for preventing mortality and morbidity in preterm infants. Cochrane Database Syst Rev. 2010(7):CD000174.
+
Hinojosa-Rodriguez
M, Harmony
T, Carrillo-Prado
C,
et al. Clinical neuroimaging in the preterm infant: diagnosis and prognosis.
NeuroImage Clin. 2017;16:355–368.
[PubMed: 28861337]
+
Intrapiromkul
J, Northington
F, Huisman
TA, Izbudak
I, Meoded
A, Tekes
A. Accuracy of head ultrasound for the detection of intracranial hemorrhage in preterm neonates: comparison with brain MRI and susceptibility-weighted imaging.
J Neuroradiol. 2013;40(2):81–88.
[PubMed: 22633043]
+
Jarjour
IT. Neurodevelopmental outcome after extreme prematurity: a review of the literature. Pediatri Neurol. 2015;52(2):143–152.
+
Luque
MJ, Tapia
JL, Villarroel
L,
et al. A risk prediction model for severe intraventricular hemorrhage in very low birth weight infants and the effect of prophylactic indomethacin.
J Perinatol. 2014;34(1):43–48.
[PubMed: 24113396]
+
Marba
ST, Caldas
JP, Vinagre
LE, Pessoto
MA. Incidence of periventricular/intraventricular hemorrhage in very low birth weight infants: a 15-year cohort study.
J Pediatr (Rio J). 2011;87(6):505–511.
[PubMed: 22015432]
+
McCrea
HJ, Ment
LR. The diagnosis, management, and postnatal prevention of intraventricular hemorrhage in the preterm neonate.
Clin Perinatol. 2008;35(4):777–792.
[PubMed: 19026340]
+
McLendon
D, Check
J, Carteaux
P,
et al. Implementation of potentially better practices for the prevention of brain hemorrhage and ischemic brain injury in very low birth weight infants.
Pediatrics. 2003;111(4 Pt 2):e497–503.
[PubMed: 12671170]
+
Ment
LR, Bada
HS, Barnes
P,
et al. Practice parameter: neuroimaging of the neonate: report of the Quality Standards Subcommittee of the American Academy of Neurology and the Practice Committee of the Child Neurology Society.
Neurology. 2002;58(12):1726–1738.
[PubMed: 12084869]
+
Panigrahy
A, Wisnowski
JL, Furtado
A, Lepore
N, Paquette
L, Bluml
S. Neuroimaging biomarkers of preterm brain injury: toward developing the preterm connectome.
Pediatr Radiol. 2012;42 Suppl 1:S33–61.
[PubMed: 22395719]
+
Robinson
S. Neonatal posthemorrhagic hydrocephalus from prematurity: pathophysiology and current treatment concepts.
J Neurosurg Pediatr. 2012;9(3):242–258.
[PubMed: 22380952]
+
Santos
J, Pearce
SE, Stroustrup
A. Impact of hospital-based environmental exposures on neurodevelopmental outcomes of preterm infants.
Curr Opin Pediatr. 2015;27(2):254–260.
[PubMed: 25635585]
+
Shooman
D, Portess
H, Sparrow
O. A review of the current treatment methods for posthaemorrhagic hydrocephalus of infants. Cerebrospinal Fluid Res. 2009;6:e1. doi: 10.1186/1743-8454-6-1
+
Smyser
CD, Snyder
AZ, Shimony
JS, Blazey
TM, Inder
TE, Neil
JJ. Effects of white matter injury on resting state fMRI measures in prematurely born infants.
PLoS One. 2013;8(7):e68098.
[PubMed: 23874510]
+
Spittle
A, Treyvaud
K. The role of early developmental intervention to influence neurobehavioral outcomes of children born preterm.
Semin Perinatol. 2016;40(8):542–548.
[PubMed: 27817913]
+
Sweeney
JK, Gutierrez
T. Musculoskeletal implications of preterm infant positioning in the NICU.
J Perinat Neonatal Nurs. 2002;16(1):58–70.
[PubMed: 12083295]
+
Ustad
T, Evensen
KA, Campbell
SK,
et al. Early parent-administered physical therapy for preterm infants: a randomized controlled trial.
Pediatrics. 2016;138(2): e20160271
[PubMed: 27440658]
+
van Wassenaer-Leemhuis
AG, Jeukens-Visser
M, van Hus
JW,
et al. Rethinking preventive post-discharge intervention programmes for very preterm infants and their parents.
Dev Med Child Neurol. 2016;58 Suppl 4:67–73.
[PubMed: 27027610]
+
Volpe,
JJ. Neurology of the Newborn. 3rd ed. Philadelphia, PA: WB Saunders; 1995.
+
Wildrick
D. Intraventricular hemorrhage and long-term outcome in the premature infant.
J. Neurosci Nurs. 1997;29(5):281–9.
[PubMed: 9361998]
+
Whitelaw
A, People
I, Cherian
S, Evans
D, Thoresen
M. Phase 1 trial of prevention of hydrocephalus after intraventricular hemorrhage in newborn infants by drainage, irrigation, and fibrinolytic therapy.
Pediatrics. 2003;111(4 Pt 1):759–765.
[PubMed: 12671109]
+
Whitfield
J, Charsha
D, Sprague
P. In search of excellence--the Neonatal Intensive Care Quality Improvement Collaborative. Proc (Bayl Univ Med Center). 2001;14(1):94–97; discussion 97-98.
+
Wong
D, Abdel-Latif
M, Kent
A. Antenatal steroid exposure and outcomes of very premature infants: a regional cohort study.
Arch Dis Child Fetal Neonatal Ed. 2014;99(1):F12–20.
[PubMed: 24142624]
+
Wu
YC, Leng
CH, Hsieh
WS,
et al. A randomized controlled trial of clinic-based and home-based interventions in comparison with usual care for preterm infants: effects and mediators.
Res Dev Disabil. 2014;35(10):2384–2393.
[PubMed: 24973546]
++
Neurodegenerative disorder affecting motor coordination, behavior, and cognition
Incidence and Prevalence
Prevalence of disease is 5-10/100,000 people worldwide (Pringsheim, 2012)
Equal frequency between sexes (Leffler, 2012)
Most common in people of western European descent (Pringsheim, 2012)
Symptoms begin between 30 and 60 years of age (mean age 40)
# of CAG repeats determinant for age of onset (Morrison, 1995); >55 CAG repeats is often seen in Juvenile Huntington's Disease (JHD) (Roos, 2010)
5-10% of cases are juvenile onset with symptoms beginning before the age of 20 (Frank, 2013)
Pathogenesis:
Caused by a cytosine-adenine-guanine (CAG) expansion mutation with >35 trinucleotide repeats on the gene
Results in degeneration of neurons in certain motor control regions of the brain (mainly the putamen and caudate nuclei substructures of the basal ganglia) and degeneration of the temporal, frontal, and occipital lobes (responsible for higher order function, movements, and sensation) (Leffler, 2012)
Brain weight may be reduced 25-30% in advanced cases (Margolis, 2003).
Degeneration of gray matter contributes to loss of cognitive function (Cruickshank, 2015)
Dopamine, glutamate, and y-aminobutyric acid are the most affected neurotransmitters in HD (Frank, 2013)
Increased TRPC5 (transient receptor potential cation 5) S-glutathionylation by oxidative stress and decreased TRPC1 (transient receptor potential cation 1) expression contribute to neuronal damage in the striatum and may underlie neurodegeneration in Huntington's disease (Hong, 2015)
++
Signs and Symptoms
Diagnostic requirements include:
Medical history
Family history
Neurological examination
Chorea, emotional and behavioral disturbances, progressive dementia (Leffler, 2012; Langbehn, 2007)
Brain imaging tests (MRI, CT)
MRI more sensitive than CT because it can detect putamen degeneration before the caudate
In moderate to severe cases, brain imaging reveals increased size of the butterfly-shaped frontal horns of the lateral ventricles due to loss of striatal volume (Kumar, 2014)
Voxel Based Morphometry (VBM) imaging has shown degeneration of grey matter in Huntington's patients (Cruickshank, 2015)
Laboratory tests, and genetic testing: Gold standard is DNA determination to identify CAG repeats (Roos, 2010)
Differential diagnosis:
A definitive molecular diagnosis can be made, even in pre-symptomatic patients, which can rule out the other 8 disorders, which are caused by the CAG repeat expansions (Margolis, 2003)
++
Progressive disorder with no known cure
Life expectancy from time of diagnosis is about 24 years (Rodriques, 2014); with mean duration 17-20 years (Roos, 2010)
Juvenile HD in 10% of cases (Huntington's Disease Society of America, 2009)
Onset before age 20
Early signs include difficulty learning and disturbances in behavior at school (Roos, 2010)
May exhibit more Parkinsonian signs (bradykinesia, dystonia, tremors) and this variation is called the Westphal Variant of HD (Frank, 2013).
Seizures and cerebellar dysfunction occur as complications (Kumar, 2014)
Decline in academic performance can occur (Kumar, 2014)
Death as a result of complications (infection, falls, pneumonia, dysphagia) (Zinzi, 2007)
Most frequent cause of death is pneumonia (Rodriques, 2014); Second most common cause is suicide (Roos, 2010)
Patients and family members are at increased risk for suicide than general population
Suicide 5 to 10 times more likely (Kumar, 2014)
Suicide is most frequent in early stages of clinical diagnosis (Baliko, 2004; Di Maio, 1993; Schoenfield, 1984)
Variations in onset and progression of Huntington's disease mostly depend on the length of the CAG repeat in DNA sequence (Harper, 1999)
<40 repeats: may never develop symptoms (Andrew, 1993)
>60 repeats usually have onset before age 20 (Andrew, 1993)
+++
IV. Medical management
++
Aimed at alleviating symptoms and focused on preserving function and quality of life (Frich,2016)
Multi-disciplinary approach can slow deterioration
Pharmacological Management
Typical/atypical neuroleptics to block dopamine receptors (Roos, 2010)
Tetrabenazine (Xenazine)
Monoamine (dopamine) depleting (Jankovic, 2014, Roos, 2010)
FDA approved treatment of chorea in HD (Fasano, 2008)
Dosage varies from 25 mg to 100 mg per day (Kumar, 2014)
Patients treated chronically with this should be monitored for possible side effects including sedation, insomnia, depression, akathisia (feeling of inner restlessness and need to move), and Parkinsonism (Kumar, 2014)
Ethyl-EPA is not effective for improving motor function in patients with HD (Ferreira, 2015)
Antipsychotic- Halozol (haloperidol)
Benzodiazapines (Klidopine)
PBT2 (a metal protein-attenuating compound that reduces huntingtin) in doses up to 250mg daily, is generally safe and well tolerated in patients with HD (Angus, 2015).
Juvenile HD may use antiparkinsonian agents (Mestra, 2009)
Surgical Techniques
Removal of medial globus pallidus—believed to be "consequently overexcited by neuronal loss"—has shown mixed results
Implantation of adrenal medullary grafts: not believe to be beneficial
Stem cell therapy: Results of transplanting stem cells into damaged areas of the brain, in an attempt to improve neural function have been inconclusive (Clelland, 2008)
Feeding tube: helps prevent patient choking on food (Panagiotakis, 2008)
Caudate-putaminal transplantation of fetal striatal tissue: Grafted patients experienced less motor/cognitive decline and associated brain metabolism improvement compared to subjects without transplantation at long-term follow-up (Paganini, 2013)
Pallidal deep brain stimulation can cause improvements in choreatic symptoms but further research is needed before this treatment can be implemented globally (Loutfi, 2014)
Genetically-Engineered Mesenchymal Stem Cells (MSC) (Deng, 2016)
Studies are looking into the use of MSC to secrete brain-derived neurotrophic factor (BDNF)
Testing in rodents has shown improvements in either neuropathological or behavioral deficits
Transplantation of MSCs in rodents has resulted in reduced striatal atrophy and medium spiny neuron loss, decreased Htt aggregation, and endogenous neurogenesis stimulation
Improvements in cognitive and motor function have also been observed
Counseling/Education
Because symptoms are often ignored or not recognized, patients and their families may need counseling to gain a better understanding of what they can and cannot do (Jankovic, 2014)
Especially important when discussing precautions to ensure safe driving, hunting, and other activities that require a large amount of motor control and judgment skills
Because some symptoms can be exacerbated by stress, relaxation techniques and stress management may be used to improve quality of life (Jankovic, 2014)
+++
V. Implications for Therapeutic Management
++
Practice Patterns (APTA, 2017)
5A: Primary Prevention/Risk Reduction for Loss of Balance and Falling
5E: Impaired Motor Function and Sensory Integrity Associated with Progressive Disorders of the Central Nervous System (APTA, 2017)
Impairments addressed by stage
Pre-symptomatic stage
Early stage
Chorea, rigidity (in some patients), impaired balance, slow saccadic eye movement, and depression (Paulsen, 2005)
Middle stage
Late stage
Pain, skin breakdown, respiratory limitations, dysphagia (Monaco, 2014)
Pneumonia (Busse, 2008; Walker, 2007)
Apathy (Thompson, 2012)
Interventions
Prevent/reduce risk of falls
Increase strength and flexibility, especially core strength
Balance, coordination, and core stability training (EHDN, 2009)
To reduce and/or prevent the risk of falls
Should be task-specific and occur in their own normal environment if possible
Progressions include:
Practice getting up from the floor
Practice recovering from perturbations using step response
Instruct in focusing on one task at a time to decrease fall risk
Gait training
Identify functionally limiting components of gait (EHDN, 2009)
May be more harmful to implement "normalized" gait
Assistive devices may increase prevalence of falls
Gait stability and safety are consistently better with a 4-wheeled walker, with faster times and fewer stumbles in a figure-eight course when compared to other ADs (Kloos, 2012)
Canes are more hazardous due to chorea of the upper extremities (EHDN, 2009)
Treadmill training at moderate intensity may improve gait speed and stride length. More research is needed (Kloos, 2014)
Functional training (EHDN, 2009)
Address specific daily tasks such as ADLs
Includes training for equipment and assistive devices
Can also include injury reduction and prevention
Chest physical therapy
Airway clearance, pursed lip breathing and diaphragmatic breathing
Optimal body positioning and ventilation-perfusion (EHDN, 2009)
Respiratory Muscle Training (RMT)
4 months of RMT can increase maximum inspiratory and expiratory pressures, forced vital capacity, and peak expiratory flow. Overall pulmonary function is improved (Reyes, 2014)
Patient and family education
Skin breakdown
Reduce risk of Pneumonia
Counseling and support groups
Individualized Home Exercise Program (Busse, 2008)
Safe transfers for caregivers and adaptive equipment to improve quality of life
Multidisciplinary Rehabilitation Intervention:
Considered gold standard for delivery of care (Frich, 2016)
Based on interdisciplinary team including physical therapists, exercise physiologists, occupational therapists, strength and conditioning specialists
Incorporated exercise program, home-base exercise program, occupational therapy
Approach is effective in increasing regional GM volume in cortical and subcortical regions (Cruickshank, 2015)
Outcome Measures
Unified HD rating scale
Specifically for Huntington's Disease
High validity and reliability for motor scores
Can be used to track patient progression over time (Siesling, 1998; Busse, 2008)
Berg Balance Scale
Activity-Specific Balance Confidence Scale
Rivermead Mobility Index
Timed Up and Go (Busse, 2008)
Performance may reflect cognitive dysfunction (Nordin, 2006)
Very high test-retest reliability and MDC in HD is 2.98s (Quinn, 2013).
Physical Performance Test (Quinn, 2013)
Hospital Anxiety and Depression Scale (Dale, 2015)
Montreal Cognitive Assessment (Van Liew, 2016)
30-point global function assessment
Examines recall and recognition memory, and can differentiate areas of memory deficit
High sensitivity and specificity in detecting cognitive dysfunction in patients with HD
Precautions/Contraindications
High fall risk related to cognitive and motor dysfunction
Cognitive/Psychological States
Because there is high risk for depression and suicidal tendencies, collaboration with mental health professionals is warranted. (Baliko, 2004; Di Maio, 1993; Schoenfield, 1984)
Aerobic Exercise Capacity
Patients with HD may have reduced/altered aerobic exercise capacity
Necessary to monitor patient's response during training (Dawes, 2014)
Breathing/Airway Clearance
+++
VI. Consumer and Professional Resources
++
Huntington's Disease Society of America
Huntington's Disease Advocacy Center
European Huntington's Disease Network Working Group of Physiotherapy: provides EBP guidelines for physical therapy and Huntington's disease
Huntington's Disease Support Group
Hereditary Disease Foundation
+
Andrew
SE, Goldberg
YP, Kremer
B, Telenius
H, Theilmann
J, Adam
S, Starr
E, Squitieri
F, Lin
B, Kalchman
MA, Graham
RK, Hayden
MR. The relationship between trinucleotide (CAG) repeat length and clinical features of Huntington's disease.
Nat Genet. 1993;4(4):398–403.
[PubMed: 8401589]
+
Angus
D, Herd
C, Stone
C, Stout
J, Wieler
M, Reilmann
R, Ritchie
CW, Dorsey
ER, Helles
K, Kayson
E, Oakes
D.. Safety, tolerability, and efficacy of PBT2 in Huntington's disease: a phase 2, randomised, double-blind, placebo-controlled trial.
Lancet Neurol. 2015;14(1):39–47.
[PubMed: 25467848]
+
Baliko
L, Csala
B, Czopf
J. Suicide in Hungarian Huntington's disease patients.
Neuroepidemiology. 2004;23(5):258–260.
[PubMed: 15316254]
+
Busse
M, Khalil
H, Quinn
L, Rosser
A. Physical Therapy Intervention for People with Huntington Disease.
Physical Therapy. 2008;88:820–821.
[PubMed: 18467429]
+
Ciarmiello
A, Cannella
M,
et al. Brain white-matter volume loss and glucose hypometabolism precede the clinical symptoms of Huntington's disease.
J Nucl Med 2006;47:215–22.
[PubMed: 16455626]
+
Clelland
CD, Barker
RA, Watts
C. Cell therapy in Huntington disease.
Neurosurg Focus. 2008;24(3-4):E9.
[PubMed: 18341412]
+
Craufurd
D, Thompson
JC, Snowden
JS. Behavioural changes in Huntington's disease.
Neuropsychiatry Neuropsychol Behav Neurol. 2001; 14:219–226.
[PubMed: 11725215]
+
Cruickshank
TM, Thompson
JA, Domínguez
D JF, Reyes
A, Bynevelt
M, Georgiou-Karistianis
N, Ziman
M. The effect of multidisciplinary rehabilitation on brain structure and cognition in Huntington's disease: an exploratory study. Brain Behav. 2015;5(2): Epub ahead of print. doi:10.1002/brb3.312.
+
Dale
M, Maltby
J, Martucci
R, Shimozaki
S, the REGISTRY investigators of the European Huntington's Disease Network. Factor analysis of the hospital anxiety and depression scale among a Huntington's disease population.
Mov Disord. 2015;30(14):1954–1960.
[PubMed: 26443751]
+
Dawes
H, Collett
J, Debono
K, Quinn
L, Jones
K, Kelson
M, Simpson
S, Playle
R, Backx
K, Wasley
D, Nemeth
A, Rosser
A, Izardi
H, Busse
M. Exercise testing and training in people with Huntington's disease. Clin Rehabil. 2015 Feb;29(2):196–206
+
Deng
P, Torrest
A, Pollock
K,
et al. Clinical trial perspective for adult and juvenile Huntington's disease using genetically-engineered mesenchymal stem cells.
Neural Regen Res. 2016;11(5):702–705.
[PubMed: 27335539]
+
Di Maio
L, Squitieri
F, Napolitano
G, Campanella
G, Trofatter
JA, Conneally
PM. Suicide risk in Huntington's disease.
J Med Genet 1993;30(4):293–295.
[PubMed: 8487273]
+
Drouin-Ouellet
J, Sawiak
SJ, Cisbani
G, Lagace
M, Kuan
W, Saint-Pierre
M, Dury
R, Alata
W, St-Amour
I, Mason
S, Calon
F, Lacroix
S, Gowland
P, Francis
S, Barker
R, Cicchetti
F. Cerebrovascular and blood-brain barrier impairments in Huntington's disease: Potential implications for its pathophysiology: Vascular impairments in HD.
Ann Neurol. 2015;78(2):160–177.
[PubMed: 25866151]
+
Ehret
JC, Day
PS, Wiegand
R, Wojcieszek
J, Chambers
RA. Huntington disease as a dual diagnosis disorder: data from the national research roster for Huntington disease patients and families.
Drug and Alcohol Dependence. 2007; 86:283–286.
[PubMed: 16930866]
+
Fasano
A, Cadeddu
F, Guidubaldi
A, Piano
C, Soleti
F, Zinzi
P, Bentivoglio
A, The Long-term Effect of Tetrabenazine in the Management of Huntington Disease.
Clinical Neuropharmacology. 2008; 31(6): 313–318.
[PubMed: 19050408]
+
Ferreira
JJ, Rosser
A, Craufurd
D, Squitieri
F, Mallard
N, Landwehrmeyer
B. Ethyl-eicosapentaenoic acid treatment in Huntington's disease: A placebo-controlled clinical trial: ETHYL-EPA FOR HUNTINGTON'S DISEASE.
Mov Disord. 2015;30(10):1426–1429.
[PubMed: 26175332]
+
Frank
S. Treatment of Huntington's Disease. Neurotherapeutics. 2014; 11(1): 153–160.
+
Frich
JC, Rae
D, Roxburgh
R,
et al. Health Care Delivery Practices in Huntington's Disease Specialty Clinics: An International Survey.
J Huntingtons Dis. 2016;5(2):207–213.
[PubMed: 27372053]
+
Harper
PS. Huntington's disease: a clinical, genetic and molecular model for polyglutamine repeat disorders.
Philos Trans R Soc Lond B Biol Sci. 1999;354(1386):957–961.
[PubMed: 10434293]
+
Ho
AK, Sahakian
BJ, Brown
RG, Barker
RA, Hodges
JR, Ané
MN, Snowden
J, Thompson
J, Esmonde
T, Gentry
R, Moore
JW, Bodner
T, NEST-HD Consortium. Profile of cognitive progression in early Huntington's disease.
Neurology. 2003;61(12):1702–1706.
[PubMed: 14694033]
+
Hong
C, Seo
H, Kwak
M, Jeon
J, Jang
J, Jeong
E, Myeong
J, Hwang
Y, Ha
K, Kang
M, Lee
K, Yi
E, Kim
I, Jeon
J, Ryu
H, So
I. Increased TRPC5 glutathionylation contributes to striatal neuron loss in Huntington's disease.
Brain. 2015;138(10):3030–3047.
[PubMed: 26133660]
+
Jankovic
J, Roos
RAC. Chorea associated with Huntington's disease: To treat or not to treat?
Mov Disord. 2014;29(11):1414–1418.
[PubMed: 25156927]
+
Kirkwood
SC, Siemers
E, Hodes
ME, Conneally
PM, Christian
JC, Foroud
T. Subtle changes among presymptomatic carriers of the Huntington's disease gene.
J Neurol Neurosurg Psychiatry. 2000;69:773–9.
[PubMed: 11080230]
+
Kloos
AD, Kegelmeyer
DA, White
SE, Kostyk
SK. The impact of different types of assistive devices on gait measures and safety in Huntington's disease.
PLOS ONE. 2012;7(2):e30903.
[PubMed: 22363511]
+
Kloos
A, Kostyk
S, Fritz
N, Fiumedora
M, Kegelmeyer
D. J06 Safety And Feasibility Of Treadmill Training In Ambulatory Individuals With Huntington's Disease. J Neurol Neurosurg Psychiatry. 2014;85(Suppl 1):A67–A67.
+
Kumar
A. Textbook of Movement Disorders. New Delhi, India: P Medical Ltd; 2014.
+
Langbehn
D, Paulsen
J. Predictors of diagnosis in Huntington disease.
Neurology. 2007;68:1710–1717.
[PubMed: 17502553]
+
Lemiere
J, Decruyenaere
M, Evers-Kiebooms
G, Vandenbussche
E, Dom
R. Longitudinal study evaluating neuropsychological changes in so-called asymptomatic carriers of the Huntington's disease mutation after 1 year.
Acta Neurol Scand. 2002;106:131–41.
[PubMed: 12174172]
+
Loutfi
G, Linder
J, Harlz
G, Blomstedt
P. Pallidal Deep Brain Stimulation in the Treatment of Huntington's Chorea. Brain Disord Ther. 2014; 3:4. doi:10.4172/2168-975X.1000136
+
Margolis
RL, Ross
CA. Diagnosis of Huntington Disease.
Clinical Chemistry. 2003;49(10):1726–1732.
[PubMed: 14500613]
+
Mestra
T, Ferreira
J, Coelho
M, Rosa
M, Sampaio
C. Therapeutic Treatment for Symptomatic Treatment in Huntington's Disease. Cochrane Database System Review. 2009;3:CD006456. Available at:
http://www.ncbi.nlm.nih.gov/pubmed/19588393.
+
Monaco
AD, Nuzzi
A, Parente
A, lavermicocca
V, Chiarelli
T, Tommaso
M, Fiore
P, Megna
M. I03 Swallowing Function In The Early, Middle And Late Stages Of Huntington's Disease. J Neurol Neurosurg Psychiatry. 2014;85(Suppl 1):A58–A58.
+
Morris
M, Tyler
A: Management and therapy. In Harper
P: Huntington's disease, Philadelphia, 1991, Saunders, pp 205–250.
+
Morrison
P, Johnston
W, Nevin
N. The epidemiology of Huntingon's disease in Northern Ireland. J of Med Gen 1995, 32(7), pp. 524–530.
+
Niccolini
F, Haider
S, Marques
TR, Muhlert
N, Tziortzi
A, Searle
G, Natesan
S, Piccini
P, Kapur
S, Rabiner
E, Gunn
R, Tabrizi
SJ, Politis
M. Altered PDE10A expression detectable early before symptomatic onset in Huntington's disease. Brain. July 2015:awv214. doi:10.1093/brain/awv214.
+
Nordin
E, Rosendahl
E, Lundin-Olsson
L. Timed "Up & Go" Test: reliability in older people dependent in activities of daily living focus on cognitive state.
Phys Ther. 2006;86:646–655.
[PubMed: 16649889]
+
Paganini
M, Biggeri
A, Romoli
AM,
et al. Fetal striatal grafting slows motor and cognitive decline of Huntington's disease. J Neurol Neurosurg Psychiatr. 2013. doi:10.1136/jnnp-2013-306533.
+
Panagiotakis
PH, DiSario
JA, Hilden
K, Ogara
M, Fang
JC. DPEJ tube placement prevents aspiration pneumonia in high-risk patients.
Nutr Clin Pract. 2008;23(2):172–175.
[PubMed: 18390785]
+
Paulsen
JS, Nehl
C, Hoth
KF, Kanz
J, Benjamin
M, Conybeare
R, McDowell
B, Turner
B. Depression and Stages of Huntington's Disease. JNP. 2005;17(4):496–502.
+
Paulsen
JS, Ready
RE, Hamilton
JM, Mega
MS, Cummings
JL. Neuropsychiatric aspects of Huntington's disease.
J Neurol Neurosurg Psychiatry. 2001;71:310–314.
[PubMed: 11511702]
+
Pringsheim
T, Wiltshire
K, Day
L, Dykeman
J, Steeves
T, Jette
N. The incidence and prevalence of Huntington's disease: A systematic review and meta-analysis.
Movement Disorders. 2012;27(9):1083–1091.
[PubMed: 22692795]
+
Quinn
L, Khalil
H, Dawes
H,
et al. Reliability and minimal detectable change of physical performance measures in individuals with pre-manifest and manifest Huntington disease.
Phys Ther. 2013;93(7):942–956.
[PubMed: 23520147]
+
Reyes
A, Cruickshank
T, Nosaka
K, Ziman
M. The Effects Of Respiratory Muscle Training On Pulmonary And Swallowing Function In Huntington's Disease Patients. J Neurol Neurosurg Psychiatry. 2014;85(Suppl 1):A67–A67.
+
Rodrigues
F, Abreu
D, Damásio
J, Goncalves
N, Correia-Guedes
L, Coelho
M, Ferreira
J. Causes Of Death In A European Huntington's Disease Cohort (registry). J Neurol Neurosurg Psychiatry. 2014;85(Suppl 1):A64–A65. doi:10.1136/jnnp-2014-309032.183.
+
Roos
RA. Huntington's disease: a clinical review.
Orphanet Journal of Rare Diseases. 2010;5:40. doi:10.1186/1750-1172-5-40.
[PubMed: 21171977]
+
Siesling
S, van Vugt
JP, Zwinderman
KA, Kieburtz,
K, Roos
RA. Unified Huntington's disease rating scale: a follow up.
Mov Disord. 1998;13: 915–919.
[PubMed: 9827615]
+
Schoenfeld
M, Myers
RH, Cupples
LA, Berkman
B, Sax
DA, Clark
E. Increased rate of suicide among patients with Huntington's disease. J Neurol Neurosurg Psychiatr. 1984;47(12):1283–1287.
+
Snowden
JS, Craufurd
D, Thompson
J, Neary
D. Psychomotor, executive, and memory function in preclinical Huntington's disease.
J Clin Exp Neuropsychol. 2002;24:133–45.
[PubMed: 11992196]
+
Thompson
JC, Harris
J, Sollom
AC, Stopford
CL, Howard
E, Snowden
JS, Craufurd
D. Longitudinal Evaluation of Neuropsychiatric Symptoms in Huntington's Disease.
J Neuropsychiatry Clin Neurosci. 2012;24(1):53–60.
[PubMed: 22450614]
+
Van Liew
C, Santoro
MS, Goldstein
J, Gluhm
S, Gilbert
PE, Corey-Bloom
J. Evaluating Recall and Recognition Memory Using the Montreal Cognitive Assessment: Applicability for Alzheimer's and Huntington's Diseases.
Am J Alzheimers Dis Other Demen. 2016;31(8):658–663.
[PubMed: 27678491]
+
Williams
S, Heron
L, France
K, Mulrooney
P, Edmondston
SJ. Huntington's Disease: Characteristics of Fallers. Physiother Res Int. 2014: doi:10.1002/pri.1577.
+
Zinzi
P, Salmaso
D, De Grandis
R, Graziani
G, Maceroni
S, Bentivoglio
A, Zappata
P, Frontali
M, Jacopini
G. Effects of an intensive rehabilitation programme on patients with Huntington's disease: a pilot study.
Clinical Rehabilitation. 2007; 21:603–13.
[PubMed: 17702702]
+++
Ischemic Encephalopathy (Near Drowning)
++
Damage to the central nervous system (CNS), regardless of cause, due to insufficient oxygen
Primary processes
At birth--Altered placental exchange (decreased maternal O2 saturation and/or reduced maternal blood flow to placenta): post maturity, prolapsed umbilical cord, umbilical cord around neck, placental insufficiency, placenta previa (abnormal implantation of placenta causing maternal hemorrhage)
ND: severe O2 deprivation from submersion in H2O. Survival beyond 24 hours of submersion. More common in males, adult and pediatric populations (Suominen, 2012)
Pathophysiology of IE in ND:
Gasping, hyperventilation, laryngospasm, hypoxemia (can lead to CNS ischemia and cardiac arrest).
Aspiration of 1-3mL/kg fluid -impaired gas exchange. Hypoxic injury to other systems.
Irreversible neuronal damage in 4 to 6 minutes.
Heart and lungs can persist for up to 30 minutes.
The duration of the oxygen deprivation undergone by the cells determines the severity of the damage. The greater the duration of oxygen deprivation, the greater the potential for cell damage. Cold water submersion- Neuroprotective Mammalian Diving Reflex more pronounced in children than adults: slows HR, redirects blood flow to the heart and brain (Near Drowning, 2003; Signorini, 2009)
CNS damage is potentially less when hypothermia is present due to the cold water lowering the amount of oxygen required by the brain tissue. (Batra, 2009)
Major sites affected: cerebral cortex (3rd, 5th, and 6th cortical layers), basal ganglia (particularly head and body of caudate nuclei and putamen), thalamus, brainstem, and cerebellum (Suominen, 2012)
Areas most vulnerable to ischemic injury include the hippocampus, insular cortex, and the basal ganglia (Suominen, 2012). In addition to the brain, the lungs and kidneys are also commonly affected (Suominen, 2012)
++
Signs and Symptoms
Alert, lethargic, comatose, not breathing, gasping for breath, vomiting, coughing, wheezing, dyspnea, bradycardia, tachycardia, anxiety, or hypothermia.
Respiratory problems may show hours after ND
Elderly population has been known to complain of dizziness for no apparent reason prior to the accident (Gregorakos, 2009)
Imaging
MRI: valuable in diagnosis (normal-appearing MRI after ND may show cerebral atrophy months after injury) (Kreis, 1995)
Diffusion-weighted MRI: identifies edematous areas of the brain with edema; superior to other imaging (Dag, 2006; Chau, 2009; Suominen, 2012)
Near-infrared spectroscopy (NIRS) is an effective bedside tool to monitor and understand brain perfusion changes in full-term asphyxiated newborns, which in conjunction with precise measurements of cerebral blood flow (CBF) obtained by MRI at particular times, may help tailor neuroprotective strategies in term newborns with hypoxic-ischemic encephalopathy (Wintermark, 2013)
Diagnosis of near drowning
Based on: events, signs and symptoms, analysis of arterial blood gases, and chest radiograph.
Some lab values can indicate a long submersion time: severe acidic pH, and high blood sugar and lactate values (Suominen, 2012)
Diagnosis of Neonate Hypoxic Ischemic Encephalopathy
American Academy of Pediatrics and the American College of Obstetrics and Gynecology guidelines: Fetal bradycardia or persistent late labor decelerations, Apgar score < 5 @ 5 min of age, positive pressure ventilation for at least 2 min after delivery, and Acidosis (pH of < 7.1) first hour after life. (Badawi, 1996)
Detecting the S100β protein levels in umbilical artery blood may be important value in early diagnosis and grading of Hypoxic-Ischemic Encephalopathy (HIE) (Jing Qian, 2009)
Severity of damage varies with severity of hypoglycemia (Bassu, 2009)
Most have bilateral diffuse infiltrates in the lungs on radiographic imaging (Miyake, 2000)
++
Non-Progressive
Neonate Hypoxic Ischemic Encephalopathy
Factors at birth: cause, location, and brain maturity at time of insult.
15-20% of infants die during newborn period; 25% -permanent deficits: CP, mental retardation, learning disabilities, and epilepsy. (Vannucci, 1997)
3 poor prognostic predictors within first 4 hours of birth: 1) administration of chest compressions >1 minute, 2) onset of breathing > 30 minutes, 3) base deficit value > 16 in blood gas analysis. Worsened prognosis with increased predictive factors. Early predictive prognosis determines rapid therapeutic measures (Prakesh, 2006).
ECG changes and serum Troponin I level at 72 hours after birth --predictive value in assessment of mortality in HIE (Kanik, 2009)
Near Drowning
Relevant factors/ND: age, submersion time (the longer the period of oxygen deprivation, the greater the extent of damage to the brain cells and tissues will be), H20 temp and degree of contamination, symptoms, associated injuries (C-spine/head), type of rescue, other medical conditions, and response to initial resuscitation
Indicators of poor outcome following cardiac arrest: immersion time > 10 minutes, time to basic life support > 10 minutes, time to first respiratory effort > 40 minutes, core temp > 33 C, persisting coma with GCS < 5. (Austin, 2013)
Survival: possible as long as 40 minutes after ND.
Full recovery -almost all who are alert and conscious upon arrival at emergency room or many with resuscitation (Suominen, 2012)
Sequelae: disturbances of consciousness, seizures, hypotonia, oculomotor-vestibular abnormalities, altered coordination, and feeding. Movement disorders: rigidity, dystonia, chorea, ataxia, dysarthria, dysphagia, and ocular apraxia. Deficits: isolated, associated, or diffuse (Pierro, 2005)
20% of all ND victims will have permanent sequelae, resulting in fatality.
Severe ischemic encephalopathy: vegetative state (Pierro, 2005)
Infections can follow the problems of the initial hypoxic damage to organs and have a very detrimental effect (Ecklund, 2012)
Risk of infection increased by shallow water drowning, arrhythmias, seizures, greater volume of fluid aspirated, and possible struggle that stirred up particles in the water in which the patient drowned (Gregorakos, 2009: Ecklund, 2012).
Inhalation of the bacteria Aeromonas hydrophila may cause necrotizing pneumonia (Miyake, 2000)
Fast improvement in condition in non-intubated patients with the use of antibiotics (Gregorakos, 2009)
Prognosis notably worse if patient aspirates gastric contents or contaminated water (Gregorakos, 2009)
+++
IV. Medical/Surgical Management
++
Initial Management
Early cardiopulmonary resuscitation is essential in reviving the patient, clearing the airway, and restoring circulation.
Patients admitted to the ICU receive neuro- protective management protocol
Sedation, nursing at 15° head up tilt with head in neutral alignment, controlled mechanical ventilation (Austin, 2013)
Pharmaceuticals
Treatment
Ventilation and perfusion, blood glucose homeostasis, neuroresuscitative measures for cerebral edema and seizures (fluid restriction, hyperventilation, and anticonvulsants).
Hypothermia at 33.5°C for 72 hours for neonatal hypoxic ischemic encephalopathy reduces death or disability to 44% to 55% (Shankaran, 2014); full body cooling is better than selective head cooling alone when creating moderate hypothermia; however, more research needs to be done on this topic (Allen, 2014)
Current research
Neonatal cryotherapy within 6 hours cools brain 3-4 0 C below normal temperature, reducing the neuromotor developmental delay and risk of death. (Glass, 2007); therapeutic hypothermia is beneficial in term and late preterm newborns with hypoxic ischemic encephalopathy. Cooling reduces mortality without increasing major disability in survivors. (Jacobs, 2013)
+++
V. Implications for Therapeutic Management
++
Practice Patterns (APTA, 2017)
Non-pulmonary components
5B: Impaired neuromotor development
5C: Impaired Motor Function and Sensory Integrity Associated with Non-progressive Disorders of the Central Nervous System-Congenital Origin or Acquired in Infancy or Childhood.
5D: Impaired Motor Function and Sensory Integrity Associated with Non-progressive Disorders of the CNS-Congenital Origin Acquired in Adolescence or Adulthood.
5I: Impaired arousal, range or motion, and motor control associated with coma
7A: Primary prevention/risk reduction for integumentary disorders
Pulmonary components
6C: Impaired ventilation, respiration/gas exchange, and aerobic capacity/endurance associated with airway clearance dysfunction
6F: Impaired ventilation and respiration/gas exchange associated with respiratory failure
Instruction, education, and training of prevention or treatment of oculomotor-vestibular abnormalities, muscle atrophy & contractures. Address all developing components of UMN syndrome. Improve motor control (stability and movement) and development of isolated movements.
Bedside care similar to TBI or SCI: management of airway and cardiopulmonary status, prevention of pressure ulcers
85% of the children status-post near drowning achieved age appropriate functioning in ADLs, but most of those children retained cognitive issues such as learning disorders and intellectual deficits (Austin, 2013).
Some of the most commonly reported long-term deficits include memory impairment, learning difficulty, visual defects, impaired visuospatial integration, and problems with executive function (Wolstenholme, 2012)
+++
VI. Consumer and Professional Resources
++
+
Allen
KA. Moderate hypothermia: is selective head cooling or whole body cooling better?
Adv Neonatal Care. 2014;14(2):113–118.
[PubMed: 24675631]
+
Austin
S, Macintosh
I. Management of drowning in children. Paediatr Child Health. 2013;23(9):397–401.
+
Badawi
N, Kurinczuk
J,
et al. Early prediction of the development of microcephaly after hypoxic-ischemic encephalopathy in the full-term newborn.
Pediatrics. 1996;97:151–152.
[PubMed: 8545217]
+
Batra
R, Paddle
J. Therapeutic hypothermia in near drowning induced hypoxic brain injury: a case report.
Cases J. 2009;2:9103.
[PubMed: 20062680]
+
Basu
P, Som
S, Choudhuri
N, Das,
H. Contribution of the blood glucose level in perinatal asphyxia.
Eur J Pediatr. 2009;168(7):833–838.
[PubMed: 18843505]
+
Chau
V, Poskitt
KJ, Sargent
MA,
et al. Comparison of computer tomography and magnetic resonance imaging scans on the third day of life in term newborns with neonatal encephalopathy.
Pediatrics. 2009;123(1):319–326.
[PubMed: 19117898]
+
Dag
Y, Firat
A, Karakas
H,
et al. Clinical outcomes of neonatal hypoxic ischemic encephalopathy evaluated with diffusion-weighted magnetic resonance imaging. Diagn Intervent Radiol. 2006;12:109–114.
+
Ecklund
M, Wahl
G, Yamshchikov
A, Smith
M. Journey of a survivor of near drowning, polymicrobial pneumonia, and acute respiratory distress syndrome. Crit Care Nurs Clin Am. 2012;24:601–623.
+
Glass
H, Ferriero
D. Treatment for hypoxic-ischemic encephalopathy in newborns.
Curr Treat Options Neurol. 2007;9(6):414–423.
[PubMed: 18173941]
+
Gregorakos
L, Markou
N, Psalida
V,
et al. Near drowning: clinical course of lung injury in adults.
Lung. 2009;187:93–97.
[PubMed: 19132444]
+
Jacobs
SE, Berg
M, Hunt
R, Tarnow-Mordi
WO, Inder
TE, Davis
PG. Cooling for newborns with hypoxic ischaemic encephalopathy. Cochrane Database Syst Rev 2013, Issue 1. Art. No.: CD003311.
+
Jing
Qian, Dong
Zhou, Yu-Wei
Wang. Umbilical artery blood S100β protein: a tool for the early identification of neonatal hypoxic-ischemic encephalopathy. Eur J Pediatr. 2009, Vol. 168 Issue 1:71–77.
+
Kanik
E, Ozer
EA, Bakiler
AR,
et al. Assessment of myocardial dysfunction in neonates with hypoxic-ischemic encephalopathy: is it a significant predictor of mortality?
J Matern Fetal Neonatal Med. 2009;22(3):239–242.
[PubMed: 19330708]
+
Kreis
R, Arcinue
E, Ernst
T,
et al. Hypoxic encephalopathy after near-drowning studied by quantitative h-magnetic resonance spectroscopy. Clinical Invest. 1995; 97: 1142–1154.
+
Miyake
M, Iga
K, Izumi
C, Miyagawa
A, Kobashi
Y, Konishi
T. Rapidly progressive pneumonia due to
Aeromonas hydrophila shortly after near-drowning.
Intern Med. 2000;39:12:1128–1130.
[PubMed: 11197807]
+
Pierro
M, Bollea
L, Di Rosa
G,
et al. Anoxic brain injury following near drowning in children rehabilitation outcomes: three case reports.
Brain Inj. 2005;19:1147–1155.
[PubMed: 16286328]
+
Ross
F, Elliot
E, Lam
L,
et al. Children under 5 presenting to paediatricians with near-drowning. J Pediatr. 2003;39:446–450.
+
Signorini
C,
et al. Free iron, total F
2-isoprostanes and total F
4-neuroprostanes in a model of neonatal hypoxic–ischemic encephalopathy: neuroprotective effect of melatonin.
J Pineal Res. 2009;46(2):148–154.
[PubMed: 19141088]
+
Suominen
PK, Vähätalo
R. Neurologic long term outcome after drowning in children.
Scand J Trauma Resusc Emerg Med. 2012;20:55.
[PubMed: 22894549]
+
Vannucci
R, Perlman
J. Interventions for perinatal hypoxic-ischemic encephalopathy.
Pediatrics. 1997;100:1004–1014.
[PubMed: 9374573]
+
Wintermark
P, Hansen
A, Warfield
SK, Dukhovny
D, Soul
JS. Near-infrared spectroscopy versus magnetic resonance imaging to study brain perfusion in newborns with hypoxic–ischemic encephalopathy treated with hypothermia.
NeuroImage. 2014;85, Part 1:287–293.
[PubMed: 23631990]
+
Wolstenholme
N, Moore
B. The clinical manifestations of anoxic brain injury. Prog Neurol. Psychiatry. 2010;14(4):8–13.
++
Most common reportable vector-borne disease affecting as many as 300,000 people in the United States per year (Nathavitharana, 2015).
Pathogenesis
A multi-system inflammatory disease caused by the bacterial spirochete Borrelia burgdorferi (Knisley, 2004). Although pathophysiology is not completely understood, tick saliva suppresses spirochete antimicrobial peptide production, which facilitates survival of spirochete and diminished chemotaxis of leukocytes (Hovius, 2009).
Contracted through infected deer tick bites (Ixodes scapularis or Ixodes pacificus), which often occur in groin, axilla, midriff or behind the knee (Knisley, 2004).
Bite may be painless/undetected (Knisley, 2004).
Bacteria not transmitted for 36-48 hours after bite; early removal of tick is vital as risk of bacterial transmission increases substantially with greater length of attachment (Knisley, 2004; Schub, 2015).
Once inside the subcutaneous tissue, the bacteria localize in sensory ganglia, endothelial, or glial cells, depending on the symptoms present in the patient, where they produce a toxin that acts upon the cell (Donta, 2012).
Areas most common in the United States:
Infection most common during spring and summer months in Northeast (Maryland to Maine), upper Midwest (Wisconsin and Minnesota), and Northern California (Knisley, 2004).
In a fourteen-year period (2000-2014), Virginia and the Appalachian mountain region showed an increase in disease incidence (Lantos, 2015).
Humans are most frequently infected between the months of May and August when human and tick activity increases in the outdoor environment (Chaaya, 2016).
Controlling deer tick density by self-application of acaricide decreases human exposure risk (Hoen, 2009).
Common risk factors include:
Exposure to rural wooded or grassy areas (Schub, 2015)
Exposure to animals often infested with ticks, such as deer and dogs (Basmatzoglou, 2015)
Hunters, rangers and other outdoor enthusiasts are at greater risk (Schub, 2015).
++
May mimic other conditions like a sports related injury (Jennings, 2009).
Early Localized Lyme Disease (stage 1) is characterized by flu-like symptoms: fatigue, chills and fever, headache, muscle and joint pain, swollen lymph nodes, erythema migrans (Mayo Foundation, 2007).
Erythema migrans
Occurs 3-30 days after bite
Begins as a single red papule and expands into a red "bull's eye"-like rash at site of bite. The rash is initially 5 cm or larger with a gradually expanding diameter that occurs 3-30 days after a bite. It is typically asymptomatic but may burn or itch. This rash occurs > 90% of infected individuals (Sanchez, 2016; Chaaya, 2016; Halperin, 2012; Hu, 2012; Zeller, 2007).
Multiple lesions of erythema migrans occur in 10-20% of cases (Chaaya, 2016).
Physical examination, medical history, and lab tests diagnose Lyme Disease (Mayo Foundation, 2007).
Diagnostic tests to confirm disease (testing for anti-Bb antibodies) (Biesiada, 2010):
Positive serological testing and presence of erythema migrans may present a false positive result in patients who do not live or have not recently travelled to a Lyme disease-endemic area. Therefore, exposure history reported by the patient is important to consider when determining whether to test for Lyme disease (Moore, 2016).
Enzyme-linked immunosorbent assay (ELISA) test (high specificity, low sensitivity) (Gomez- Solecki, 2007).
C6 peptide ELISA test is also available for diagnosis in some instances and is useful in detecting early IgG response. It is more reliable than looking at the IgM response which renders higher false-positives, and has higher sensitivity and specificity than the standard ELISA. It has decreased specificity compared to two-tiered test (Nathavitharana, 2015).
Western blot test (Mavin, 2007).
Western blot test is administered following a positive enzyme immunoassay test when testing for acute Lyme disease (Sanchez, 2016).
If symptoms of Lyme disease have been present for 4 weeks or greater, Western blot test alone is recommended due to its high sensitivity for Lyme disease of over 4 weeks duration (Sanchez, 2016).
Polymerase chain reaction (PCR) (Mayo Foundation, 2007).
Tests have high sensitivity, specificity and predictive value (Halperin, 2012).
Clinical differential diagnosis includes many skin conditions (e.g. tinea, nummular eczema and southern tick-associated rash illness (STARI)) (Sanchez, 2016).
++
Progressive if not detected and treated properly, but all symptoms typically resolve within period of treatment (Hu, 2012).
Progression
Differentiation of stages difficult--no clear endpoints between. Some skip stages (Knisley, 2004).
Duration of the disease may be up to 22 weeks (Halperin, 2015)
Early Localized Lyme Disease (Stage 1):
3-30 days after initial tick exposure (Hu, 2012)
Antibiotics given at this stage are effective in almost 99% of patients (Biesiada, 2010).
Early Disseminated Lyme Disease (Stage 2)
Can occur weeks to months after onset and can include: Numbness and pain in extremities from inflammatory radiculopathy, Bell's Palsy (occurs in 5-10% of untreated individuals), diplopia, vertigo, hearing loss, meningitis (occurs in 10-15% of untreated patients), abnormal heart rhythm due to conduction block, opsoclonus-myoclonus (a condition that involves multidirectional saccadic eye movements associated with myoclonus and ataxia) (Halperin, 2000; Peter, 2006; Knisley, 2004).
Late/Chronic Lyme Disease (Stage 3)
Can occur weeks, months, or years after onset
The "classic triad" of symptoms for chronic Lyme Disease include lymphocytic meningitis, painful radiculoneuritis, and cranial neuritis (Halperin, 2015)
Can include:
Chronic joint inflammation (Lyme Arthritis) and joint swelling in 32.5% of the cases – particularly in the knees of untreated patients (Hatchette, 2015; Zeller, 2007)
Cognitive defects such as memory loss and/or difficulty concentrating (Mayo Foundation, 2007)
Changes in mood and sleep patterns (Mayo Foundation, 2007)
Chronic headache and/or blindness due to increased intracranial pressure (Moses, 2003)
A sub-category of Chronic Lyme disease, known as Post-Lyme disease, is believed to be caused by persistent infection with B. burgdorferi, other tick-borne infections, antibiotic treatments, etc., and is commonly associated with fatigue, arthralagias, myalgias, neck stiffness, paresthesias, sleeplessness, irritability, and cognitive deficits (Marques, 2008).
These symptoms can last for weeks or years
Post-treatment Lyme Disease Syndrome is diagnosed when the symptoms of fatigue, neurocognitive deficits, musculoskeletal deficits myalgia and arthralgia (Oliveria, 2015), and insufficient memory and coordination lasts six months after the treatment of Lyme Disease (Adrion, 2015).
Lyme neuroborreliosis (LNB) may present as meningitis, cranial neuropathy, acute radiculoneuropathy or, rarely, as encephalomyelitis (Ramesh et al, 2009). After antibiotics, 95% of cases have no long-term effects (Nau, 2009).
Cardiac conduction abnormalities can be associated with Lyme disease, but they are usually self-limiting (Medical, 2016).
++
Lyme disease is curable and responds well to oral/IV antibiotic therapy. Type of antibiotics depends on the stage of disease.
Early – oral antibiotics
Late – IV antibiotics
Pregnant women and children <8 years old should not be treated with Doxycycline (Hu, 2012). Amoxicillin or cefuroxime should be used instead (Nathavitharana, 2015).
Long-term antibiotic regimens have not proven effective for patients with persistent symptoms attributed to Lyme disease (Berende, 2016).
Prophylactic treatment with doxycycline has been shown to be 87% effective at preventing erythema migrans at the bite (Sanchez, 2016).
Others treatment includes:
NSAIDs or corticosteroids for inflammation, temporary pace-maker for complete heart block, synovectomy for chronic joint inflammation (Knisley, 2004).
Patients discouraged from taking supplemental antioxidants and vitamin B, C and E, but encouraged to take extra doses of vitamin D (Donta, 2012)
Previous vaccine (Lymerix) discontinued 2002 due to FDA concerns about drug's safety and effectiveness (Mayo Foundation, 2007)
Patients with Post-Treatment Lyme Disease Syndrome do not benefit from the use of antibiotic therapy (Oliviera, 2015).
+++
V. Implications for Therapeutic Management
++
+++
VI. Consumer and Professional Resources
++
+
Adrion
ER, Aucott
J, Lemke
KW, Weiner
JP. Health Care Costs, Utilization and Patterns of Care following Lyme Disease.
PLoS One. 2015;10(2): e0116767. doi: 10.1371/journal.pone.0116767.
[PubMed: 25650808]
+
Basmatzoglou
T, Vgenopoulou
I, Saridi
M. LYME Disease: Prevention and Treatment of Recurrent Disease. Int J Caring Sci. 2015;8(1):221–230.
+
Berende
A, ter Hofstede
HJ, Vos
FJ,
et al. Randomized trial of longer-term therapy for symptoms attributed to Lyme disease.
N Engl J Med. 2016; 374(13): 1209–20.
[PubMed: 27028911]
+
Biesiada
G, Czepeil
J, Lesniak
MR, Garlicki
A, Mach
T. Lyme disease: review. Arch Med Sci. 2010;8(6):978–982.
+
Chaaya
G, Jaller-Char
J, Ali
S. Beyond the bull's eye: recognizing Lyme disease.
J Fam Pract. 2016; 65(6): 373–379.
[PubMed: 27474818]
+
Donta
ST. Issues in the diagnosis and treatment of Lyme disease.
The Open Neurology Journal. 2012;6:140–145.
[PubMed: 23248715]
+
Gomes-Soleki
M, Meirelles
L, Glass
J, Dattwyler
R. Epitope length, genospecies dependency, and serum panel effect in the IR6 immunosorbent assay for detection of antibodies to Borellia burgdorferi.
Clinical and Vaccine Immunology. 2007; 14: 875–879.
[PubMed: 17538122]
+
Halperin
JJ. Chronic Lyme disease: misconceptions and challenges for patient management.
Infect Drug Resist. 2015;8:119–128.
[PubMed: 26028977]
+
Halperin
JJ. Lyme disease: a multisystem infection that affects the nervous system. Continuum Lifelong Learning Neurol. 2012;18(6):1338–1350.
+
Halperin
J. Nervous system Lyme disease. Infect Med. 2000;17:556–560.
+
Hatchette
TF, Johnston
BL, Schleihauf
E, Mask
A, Haldane
D, Drebot
M, Baikie
M, Cole
T, Fleming
S, Gould
R, Lindsay
R. Epidemiology of Lyme Disease, Nova Scotia, Canada, 2002–2013.
Emerg Infect Dis. 2015;21(10):1751–1758.
[PubMed: 26401788]
+
Hoen
AG
et al.. Effects of tick control by acaricide self-treatment of white-tailed deer on host-seeking tick infection prevalence and entomologic risk for Ixodes scapularis-borne pathogens. Vector Borne Zoonotic Dis. 2009 Aug;9(4):431–8.
+
Hovius
JW. Spitting image: tick saliva assists the causative agent of Lyme disease in evading host skin's innate immune response.
J Invest Dermatol. 2009;129(10):2337–9.
[PubMed: 19749783]
+
Jennings
F, Lambert
E, Frederickson
M. Rheumatic diseases presenting as sports-related injuries.
Sports Med. 2008;38(11):917–30.
[PubMed: 18937522]
+
Karosi
T, Rácz
T, Szekanecz
E, Tóth
A, Sziklai
I. Recurrent laryngeal nerve paralysis due to subclinical Lyme borreliosis. J Laryngol Otol. 2009;10:1–3.
+
Knisley
J, Johnson
M. Lyme disease: knowledge is the best prevention.
The Nurse Practitioner. 2004;29: 34–43.
[PubMed: 15553916]
+
Lantos
PM, Nigrovic
LE, Auwaerter
PG, Fowler
Jr V, Ruffin
F, Brinkerhoff
J, Reber
J, Williams
C, Broyhill
J, Pan
W, Gaines
D. Geographic Expansion of Lyme Disease in the Southeastern United States, 2000–2014.
Open Forum Infect Dis. 2015;2(4):143. doi: 10.1093/ofid/ofv143.
[PubMed: 27909696]
+
Mavin
S, Milner
M, Evans
R, Chatterton
J, Joss
A, Ho-Yen
D. The Use of Local Isolates in Western Blots Improves Serological Diagnois of Lyme disease in Scotland.
Journal of Medical Microbiology. 2007; 56: 47–51.
[PubMed: 17172516]
+
Marques
A. Chronic Lyme Disease: An appraisal.
Infect Dis Clin North Am. 2008;22(2):341–360.
[PubMed: 18452806]
+
The Medical Letter. Treatment of Lyme disease. Med Lett Drugs Ther. 2016; 1494: 57–58.
+
Moore
A, Nelson
C, Molins
C, Mead
P, Schriefer
M. Current guidelines, common clinical pitfalls, and future directions for laboratory diagnosis of Lyme disease, United States. Emerg Infect Dis. 2016; 22(7): 1169–1177.
+
Moses
JM, Riseberg
RS, Mansbach
JM. Lyme disease presenting with persistent headache.
Pediatrics. 2003;112:e477–e479.
[PubMed: 14654649]
+
Nau
R, Christen
HJ, Eiffert
H. Lyme disease--current state of knowledge. Dtsch Arztebl Int. 2009 Jan;106(5):72–81.
+
Nathavitharana
RR, Mitty
JA. Diseases from North America: focus on tick-borne infections. Clin Med. 2015;15(1):74–77.
+
Oliveira
CR, Shapiro
ED. Update on persistent symptoms associated with Lyme disease.
Curr Opin Pediatr. 2015;27(1):100–104.
[PubMed: 25490690]
+
Peter
L, Jung
J, Tilikete
C, Ryvlin
P, Mauguiere
F. Opsoclonus-myoclonus as a manifestation of lLyme disease.
J Neurol Neurosurg Psychiatry. 2006; 77: 1090–1091.
[PubMed: 16914760]
+
Ramesh
G, Borda
JT, Gill
A, Ribka
EP, Morici
LA, Mottram
P, Martin
DS, Jacobs
MB, Didier
PJ, Philipp
MT. Possible role of glial cells in the onset and progression of Lyme neuroborreliosis. J Neuroinflammation. 2009 Aug 25;6:23.
+
Souayah
N, Ajroud-Driss
S, Sander
HW, Brannagan
TH, Hays
AP, Chin
RL. Small fiber neuropathy following vaccination for rabies, varicella or Lyme disease. Vaccine. 2009 Dec 9;27(52):7322–5.
+
Vazquez
M, Muehlenbein
C, Cartter
M, Hayes
E, Starr
E, Shapiro
E. Effectiveness of Personal Protective Measures to Prevent Lyme disease. Emergency Infectious Disease Journal. 2008; 14:2:210–216.
++
A common form of endolymphatic hydrops of the inner ear where endolymph accumulates excessively in the endolymphatic space and compromises perilymphatic space
Incidence is higher among females (84/100,00) compared to males (56/100,000) and highest in Caucasians (91/100,000) among other races (Simo, 2015). Incidence is also greater with major histocompatibility complexes like antigens B8/DR3 and Cw7.
7-15% report a family history of similar symptoms, suggesting an autosomal dominant inheritance (Eppsteiner, 2011).
Peak age of onset from 40-60 years (Yew, 2014).
While the terms are often used synonymously, "Meniere's disease" is used if the cause is unknown; "Meniere's syndrome" is used if the cause can be identified, such as an autoimmune disorder, allergies, etc. (Syed, 2015).
Causes
Most likely a combination of fibrosis, atrophy of sac, obstruction, infection, vascularity of ear, overproduction of endolymph, posttraumatic, migraines, allergic responses, and genetic abnormalities (Le, 2013). Migraines appear in 50% of cases (Foster, 2013).
Hydrops alone is insufficient to cause Meniere's disease. Additional cofactors, possibly vascular risk factors, must be present in order for asymptomatic hydrops to become symptomatic Meniere's disease (Foster, 2013; Gürkov, 2016)
In Meniere's disease, the endolymph buildup in the labyrinth interferes with the normal balance and hearing signals between the inner ear and the brain, causing vertigo.
Autoimmunity may contribute to the development of Meniere's disease (Kim, 2014).
Presence of autoimmune disorders such as Rheumatoid Arthritis, Systemic Lupus Erythematosus, and Ankylosing Spondylitis have been strongly linked to Meniere's disease (Gazquez, 2011).
Patients with Meniere's disease have been shown to have elevated levels of: immune complexes, autoimmune responses to type II collagen and to other inner ear antigens, local inflammation of the endolymphatic sac, IgG deposits within the endolymphatic sac, autoantibodies to the endolymphatic sac, and other identifiers of immune-mediated disease (Syed, 2015)
Associated problems
Increased risk of falls, the inability to drive or opporate machinery, and the inability to work.
Somatic symptoms of Meniere's disease (particularly vertigo) leads to emotional disturbances which can provoke further somatic symptoms. More severe vertigo in a patient with Meniere's disease can result in decreased quality of life (Orji, 2014).
Patients with Meniere's disease are three times more likely to have a diagnosed allergy when compared to the general population (Weinreich, 2014).
Pathogenesis
"The exact cause of Meniere's disease is unknown." It may occur when the pressure of the fluid in part of the inner ear gets too high. In some cases, Meniere's disease may be related to head injury, middle ear infection, and syphilis. Other risk factors include: allergies, alcohol use, family history, fatigue, recent viral illness, respiratory infection, smoking, stress, and the use of certain medications (Crane, 2010).
Distention of otoliths puts pressure on the ampulla creating spinning sensation.
+++
II. Diagnostic Procedures
++
Symptoms
The American Academy of Otolaryngology – Head and Neck Surgery (AAO-HNS) classified Meniere's disease into 4 stages according to the clinical symptoms and exclusion of identifiable other causes (Le, 2013):
Certain, definitive, probable, and possible
Definitive Meniere's disease required: 2 or more spontaneous rotational vertiginous episodes lasting at least 20 min up to 24 hours, low frequency sensorineural hearing loss documented by audiometry, tinnitus (ringing) or aural fullness (increased pressure) in the affected ear, and all other causes excluded (Le, 2013).
Acute episodes can occur in clusters of about 6–11 a year, although remission may last several months (Friberg, 1984).
Balance Impairment: Patients have greater dependence on visual and somatosensory input in maintaining balance and upright posture. Acute stages after a vertigo attack have greater somatosensory dependence than in the chronic stages (Fujimoto, 2013)
More recently, motion sensitivity has been identified as a common symptom (Sharon, 2014).
Bilateral symptoms have been reported in 24% of cases (Lee 2012).
Classification
By AAO-HNS classification standards, diagnosing Meniere's disease can be difficult due to the latency of "presentation of the cardinal complaints", and variable symptoms can occur at separate times with hearing loss occasionally recovering before diagnostic tests can be made (Pyykko, 2013; AAO-HNS, 1995).
Diagnostic Testing
Meniere's disease is most often diagnosed and treated by an otolaryngologist (commonly called an ear, nose, and throat doctor, or ENT). However, there is no definitive test or single symptom that a doctor can use to make the diagnosis. Diagnosis is based upon your medical history and the presence of symptoms.
Diagnostic tests might include
Autoantibodies and antigen have been theorized and are being sought through reasearch to serve as diagnostic biomarkers for Meniere's disease (Kim, 2014).
++
Although there is currently no cure, more than 85% of patients with Meniere's disease are helped by either changes in lifestyle and medical treatment, or minimally invasive surgical procedures such as intratympanic steroid therapy, intratympanic gentamicin therapy, and endolymphatic sac surgery.
Trends of Progression
Meniere's disease is at first progressive but fluctuates unpredictably. It is difficult to distinguish natural resolution from the effects of treatment. Significant improvement in vertigo is usually seen in the placebo arm of RCTs (Schmidt, 1992; Moser, 1984).
Acute attacks of vertigo often increase in frequency during the first few years after presentation and then decrease in frequency in association with sustained deterioration in hearing (Moffat, 1997).
Symptoms other than hearing loss improve in 60–80% of people irrespective of treatment (Jacobson, 1990).
Meniere's disease is said to "burn out" over time. The spontaneous remission rate is high: over 50% within 2 years and over 70% after 8 years.
Meniere's disease may be considered as one of the causes of persistent vertigo in patients with BPPV, posing difficulties in obtaining the right diagnosis and aggravating the prognosis (Riga, 2011).
In relation to Meniere's disease and the ICF model, 70% of responders reported impairments, 39% reported activity limitations, 47% reported participation restrictions, 16% reported interference with environmental factors, and 28% reported interference with personal factors (Tassinari, 2015).
Can lead to social phobic reactions.
+++
IV. Medical and Surgical Management
++
Pharmaceuticals
Diuretics control vertigo and stabilize hearing.
Gentamycin injections or sac decompression makes disease more complex.
Gentamycin injections are effective in relieving symptoms of vertigo, but are ineffective in relieving aural fullness (Zhai, 2013).
Intratympanic gentamicin carries the risk of inducing hearing loss, due to its dose-dependent ototoxicity. The titration method in administration of gentamicin has been shown to relieve vertigo symptoms while reducing the risk of damage to vestibular and cochlear structures (Syed, 2015).
Antidepressive medications if reactive depressive disorder occurs.
Intratympanic dexamethasone and methyl prednisolone are the two most common steroid preparations and may be effective in meaningful symptomatic control of hearing loss, tinnitus, and vertigo (Memari, 2014; Syed, 2015).
Oral betahistine is effective in preventing, treating and reversing the symptoms of Meniere's disease over time (Tootoonchi, 2015).
Symptoms from acute vertigo attacks can be mitigated with benzodiazepines, meclizine, antihistamines such as promethazone or diphenhydramine, and transdermal scopolamine patches. However, these medications have no effect on the progression of the disease (Foster, 2015).
Lifestyle modifications
Restrict salt, stress, caffeine, alcohol, and nicotine, and avoid MSG.
Avoid migraine food triggers such as MSG, chocolate, red wine, fermented dairy products, and aged or pickled foods.
Regular sleeping and eating patterns.
Remain physically active, but avoid excessive fatigue.
Sit or lie down immediately when dizziness occurs.
Avoid driving a car or operating heavy machinery.
Awareness of the possibility of a fall.
Surgical technique
Surgery may be recommended when all other treatments have failed to relieve dizziness. Some surgical procedures are performed on the endolymphatic sac to decompress it. Another possible surgery is to cut the vestibular nerve, although this occurs less frequently
Studies show that decompression surgery significantly improves vertigo and hearing in patients with Meniere's disease who have no psychological distress in comparison with those who displayed signs of mental disturbances, but is found to be effective regardless of a patient's psychological condition (Furukawa, 2013).
Vestibular neurectomy is very effective at vertigo control and is available for patients with good hearing who have failed all other treatments (Sajjadi, 2008).
Labyrinthectomy is undertaken as a last resort and is best reserved for patients with unilateral disease and deafness (Sajjadi, 2008).
For those with Meniere-related deafness, cochlear implantation represents the only viable option to restore auditory perception (Hansen, 2013).
Nonsurgical devices
+++
V. Implications for Therapeutic Management
++
Physical therapy preferred practice patterns (APTA, 2017)
Pattern 5A: Primary Prevention/Risk Reduction for Loss of Balance and Falling
Pattern 5D: Impaired Motor Function and Sensory Integrity Associated with Nonprogressive Disorders of the Central Nervous System – Aquired in Adolescence or Adulthood.
Pattern 5E: Impaired Motor Function and Sensory Integrity Associated with Progressive Disorders of the Central Nervous System.
Pattern 5F: Impaired Peripheral Nerve Integrity and Muscle Performance Associated With Peripheral Nerve Injury
Optimal treatment would be directed towards preventing vertigo attacks, reducing tinnitus, and reversing hearing loss. However, the nature of the disease's remissions and exacerbations can make evaluation of effectiveness of treatment difficult (Syed, 2015).
Physical therapy combined with behavioral therapy helps with coping through stress reduction and relaxation exercises.
Clinical studies suggest that increased levels of vasopressin and cortisol, stress-modulating hormones, are a result of this chronic disease and correlate with inner ear fluid homeostatic disorder and consequent endolymphatic hydrops. This new bodily homeostasis after stress may lead to wear and tear of organs and tissues and make individuals more vulnerable to illness (Van Cruijsen, 2005; Kirby, 2008).
Therapeutic techniques such as tinnitus retraining therapy, sound therapy (masking), cognitive behavioral therapy, and vestibular rehabilitation are more effective at improving quality of life over any current pharmaceutical option (Tassinari, 2015).
When evaluating postural stability of patients with Meniere's disease in the clinic, the therapist should realize that the results are affected by the time elapsed since their last vertigo episode. Clinicians should take a thorough history that includes time since last vertigo episode (Fujimoto, 2013).
Role of PT
+++
VI. Consumer and Professional Resources
++
+
AAO-HNS American Academy of Otolaryngology-Head and Neck Foundation, Inc. Committee on hearing and equilibrium guidelines for the diagnosis and evaluation of therapy in Meniere's disease.
Otolaryngol Head Neck Surg. 1995;113(3):181–185
[PubMed: 7675476]
+
Agrawal
Y, Ward
BK, Minor
LB. Vestibular dysfunction: Prevalence, impact and need for targeted treatment. J Vestib Res Equilib Orientat. 2013;23(3):113–117.
+
Ahsan
SF, Standring
R, Wang
Y. Systematic review and meta-analysis of meniett therapy for Meniere's disease.
The Laryngoscope. 2015;125(1):203–208. doi:10.1002/lary.24773.
[PubMed: 24913022]
+
American Physical Therapy Association (APTA). Guide to Physical Therapist Practice. Alexandria, VA, 2015.
+
Chen
EW, Fu
ASN, Chan
KM, Tsang
WWN. The effects of Tai Chi on the balance control of elderly persons with visual impairment: a randomised clinical trial.
Age Ageing. 2012;41(2):254–259.
[PubMed: 22180415]
+
Crane
BT, Schessel
DA, Nedzelski
J, Minor
LB. Peripheral vestibular disorders. In: Cummings
CW, Flint
PW, Haughey
BH
et al.. Otolaryngology: Head & Neck Surgery. 5th ed. Philadelphia, Pa: Mosby Elsevier; 2010:chap 165.
+
Eppsteiner
RW, Smith
RJH. Genetic disorders of the vestibular system.
Curr Opin Otolaryngol Head Neck Surg. 2011;19(5):397–402.
[PubMed: 21825995]
+
Foster
CA, Breeze
RE. Endolymphatic Hydrops in Ménière's Disease: Cause, Consequence, or Epiphenomenon?. Otology & Neurology. 2013;34(7):1210–1214.
+
Foster
CA, Breeze
RE. The Meniere attack: an ischemia/reperfusion disorder of inner ear sensory tissues. Med Hypotheses. 2013;81(6):1108–15.
[PubMed: 24199949]
+
Foster
CA. Optimal management of Ménière's disease. Dove Press:
Therapeutics and Clinical Risk Management. 2015;11:301–307.
[PubMed: 25750534]
+
Friberg
U, Stahle
J, Svedberg
A. The natural course of Menière's disease.
Acta Otolaryngol Suppl. 1984; 406: 72–77.
[PubMed: 6591717]
+
Fujimoto
C, Egami
N, Kinoshita
M, Sugasawa
K, Yamasoba
T, Iwasaki
S. Factors Affecting Postural Instability in Meniere's Disease.
Otolaryngology -- Head and Neck Surgery. 2013;149(5):759–765.
[PubMed: 23980034]
+
Furukawa
M, Kitahara
T, Horii
A, Uno
A, Imai
T, Ohta
Y, Morihana
T, Inohara
H, Mishiro
Y, Sakagami
M. Psychological condition in patients with intractable Meniere's disease,
Acta Oto-Laryngologica. 2013;133:6, 584–589.
[PubMed: 23675809]
+
Gazquez
I, Soto-Varela
A, Aran
I,
et al. High prevalence of systemic autoimmune diseases in patients with Meniere's disease.
PloS One. 2011;6(10):e26759. doi:10.1371/journal.pone.0026759.
[PubMed: 22053211]
+
Gottshall
KR, Hoffer
ME,
et al. The role of vestibular rehabilitation in the treatment of Meniere's disease. Otolaryngology: Head & Neck Surgery. 2005;133(3): 326–8.
+
Gürkov
R, Pyykö
I, Zou
J, Kentala
E. What is Menière's disease? A contemporary re-evaluation of endolymphatic hydrops. Journal of Neurology. 2016;263(S1):71–81.
+
Hansen
MR, Gantz
BJ, Dunn
C. Outcomes following cochlear implantation for patients with single-sided deafness, including those with recalcitrant Meniere's disease.
Otol Neurotol. 2013;34(9):1681–1687
[PubMed: 24232066]
+
Hornibrook
J, Flook
E, Greig
S,
et al. MRI Inner Ear Imaging and Tone Burst Electrocochleography in the Diagnosis of Meniere's Disease.
Otol Neurotol. 2015;36(6):1109–1114.
[PubMed: 25985318]
+
Jacobson
GP, Newman
CW. The development of the Dizziness Handicap Inventory. Otolaryngology: Head & Neck Surgery. 1990;116:424–427.
+
Kim
SH, Kim
JY, Lee
HJ, Gi
M, Kim
BG, Choi
JY. Autoimmunity as a Candidate for the Etiopathogenesis of Meniere's Disease: Detection of Autoimmune Reactions and Diagnostic Biomarker Candidate. PLoS One. 2014;9(10). doi:10.1371/journal.pone.0111039.
+
Kirby
S, Yardley
L. Understanding psychological distress in Ménière's disease: A systematic review.
Psychology, Health & Medicine. 2008;13(3):257–273.
[PubMed: 18569895]
+
Lamounier
P, Gobbo
DA, Souza
TSA de, Oliveira
CACP de, Bahmad
F. Electrocochleography for Meniere's disease: is it reliable?
Braz J Otorhinolaryngol. 2014;80(6):527–532.
[PubMed: 25443316]
+
Le
CH, Truong
AQ, Diaz
RC. Novel techniques for the diagnosis of Ménière's disease.
Curr Opin Otolaryngol Head Neck Surg. 2013;21(5):492–6.
[PubMed: 23995329]
+
Lee
ATH. Diagnosing the cause of vertigo: a practical approach.
Hong Kong Med J. 2012;18(4):327–332.
[PubMed: 22865178]
+
Memari
F, Hassannia
F. Effect of Intratympanic Dexamethasone on Controlling Tinnitus and Hearing loss in Meniere's Disease.
Iran J Otorhinolaryngol. 2014;26(76):129–133.
[PubMed: 25009802]
+
Moffat
DA, Ballagh
RH. Menière's disease. In: Kerr
AG, Booth
JB. Scott Brown's otolaryngology. 6th ed. Oxford: Butterworth-Heinemann, 1997.
+
Moser
M, Ranacher
G, Wilmot
TJ,
et al. A double-blind clinical trial of hydroxyethylrutosides in Menière's disease.
J Laryngol Otol. 1984; 98:265–272.
[PubMed: 6366100]
+
Nyabenda,
A., Briart,
C.,
et al. (2003). Benefit of rotational exercises for patients with Meniere's syndrome, method used by the ENT department of St-Luc University clinic.
Annales de Readaptation et de Medecine Physique. 46(9), 607–614.—this is a French journal.
[PubMed: 14642673]
+
Orji
F. The Influence of Psychological Factors in Meniere's Disease.
Ann Med Health Sci Res. 2014;4(1):3–7.
[PubMed: 24669323]
+
Pyykko
I, Nakashima
T, Yoshida
T, Zou
J, Naganawa
S. Meniere's disease: a reappraisal supported by a variable latency of symptoms and the MRI visualisation of endolymphatic hydrops. BMJ Open. 2013;3(2).
+
Requena
T, Espinosa-sanchez
JM, Lopez-escamez
JA. Genetics of dizziness: cerebellar and vestibular disorders. Curr Opin Neurol. 2014;27(1):98–104.
[PubMed: 24275721]
+
Riga
M, Bibas
A, Xenellis
J, Korres
S. Inner ear disease and benign paroxysmal positional vertigo: a critical review of incidence, clinical characteristics, and management. International Journal of Otolarygology. 2011. 7 pages.
+
Sajjadi
H, Paparella
MM. Meniere's disease. Lancet. 2008 Aug 2;372(9636):406–14.
+
Schmidt
JT, Huizing
EH. The clinical drug trial in Menière's disease with emphasis on the effect of betahistine.
Acta Otolaryngol Suppl. 1992;497:1–189.
[PubMed: 1496890]
+
Sharon
JD, Hullar
TE. Motion sensitivity and caloric responsiveness in vestibular migraine and Meniere's Disease.
The Laryngoscope. 2014;124(4):969–973.
[PubMed: 23818082]
+
Simo
H, Yang
S, Qu
W, Preis
M, Nazzal
M, Baugh
R. Meniere's disease: importance of socioeconomic and environmental factors.
Am J Otolaryngol. 2015;36(3):393–398.
[PubMed: 25771842]
+
Socher
DD, Socher
JA, Azzi
VJB. Evaluation of quality of life pre- and post-vestibular rehabilitation in patients with benign paroxysmal positional vertigo associated with Meniere's disease.
Int Arch Otorhinolaryngol. 2012;16(4):430–436.
[PubMed: 25991970]
+
Syed
M, Ilan
O, Nassar
J, Rutka
J. Intratympanic therapy in Meniere's syndrome or disease: up to date evidence for clinical practice.
Clinical Otolaryngology. 2015;40(6):682–690.
[PubMed: 25916787]
+
Tassinari
M, Mandrioli
D, Gaggioli
N, Roberti
di Sarsina P. Meniere's Disease Treatment: A Patient-Centered Systematic Review. Audiol Neurotol. 2015;20(3):153–165.
+
Tootoonchi
SJ, Ghiasi
S, Shadara
P, Samani
SM, Fouladi
DF. Hearing function after betahistine therapy in patients with Meniere's disease. Braz J Otorhinolaryngol. 2016;S1808-8694(15):234–237.
+
Van Cruijsen
N, Dullaart
R, Wit
H, Albers
F. Analysis of Cortisol and Other Stress-Related Hormones in Patients with Meniere's Disease. Otology & Neurotology. 2005;26(6):1214–1219..
+
Weinreich
HM, Agrawal
Y. The Link Between Allergy and Meniere's Disease.
Curr Opin Otolaryngol Head Neck Surg. 2014;22(3):227–230.
[PubMed: 24573125]
+
Yew
KS. Diagnostic approach to patients with tinnitus.
Am Fam Physician. 2014;89(2):106–113.
[PubMed: 24444578]
+
Zhai
F, Zhang
R, Zhang
T, Steyger
PS, Dai
C-F. Preclinical and Clinical Studies of Unrelieved Aural Fullness following Intratympanic Gentamicin Injection in Patients with Intractable Meniere's Disease. Audiol Neurotol. 2013;18(5):297–306.
++
Inflammatory demyelinating disease of the central nervous system (Calabresi, 2006; Evans, 2002).
Pathogenesis includes white matter inflammation, demyelination, and subsequent gliosis (plaque formation) representing scar formation in the white matter (Calabresi, 2006; Evans, 2002)
Leading cause of non-traumatic neurological disability in young adults (Abarony, 2015)
Onset usually between ages 15 and 50; Average age at diagnosis is 29 years for women and 31 years for men (Tsang, 2011)
Prevalence
Has increased over the past few decades, with a greater prevalence among females (Nischwitz, 2011), ratio of women: men is 2-3:1 (Tintore, 2015; Scalfari, 2011)
Incidence higher in African Americans versus Caucasians with Hispanics having a lower incidence than either (Wallin, 2012).
It is estimated that 2.5 million people are affected by MS worldwide with distribution of the disease varying significantly among geographic regions (Marrie, 2015)
Lower prevalence of MS near the equator and Asian countries (Nischwitz, 2011)
Higher prevalence in increasing socioeconomic status societies (Nischwitz, 2011)
Higher in individuals that smoke (Handel, 2011), have decreased levels of vitamin D (Kampman, 2008), and have obesity in adolescence (Tur, 2015).
Precise etiology remains unknown but probably a combination of genetic, autoimmune, and environmental factors (Calabresi, 2005; Granieri, 2001).
Genetic and Autoimmune factors:
30% concordance rate in monozygotic twins (Milo, 2014)
First degree relatives have a 20-40 times greater risk of developing MS; (Multiple sclerosis - PubMed Health, 2011)
May be linked to specific genes (MHC alleles) (Prat, 2002; Wallin, 2012).
HLA-DRB1 chromosomal locus has been consistently linked with MS prevalence (Nischwitz, 2011).
HLA-C*05 allele is believed to protect against MS (Faguy, 2016) - direct
Autoimmune response in which T cells react to myelin basic protein and proteolipid protein (Evans, 2002; Granieri, 2005; Prat, 2002)
"Having another autoimmune disease, such as thyroid disease, type 1 diabetes, or inflammatory bowel disease, is known to slightly raise the likelihood of developing MS" (Multiple Sclerosis – Mayo Clinic Website, 2015)
Increased concentrations of IL-33 (a protein called Interleukin-33) in patients with both treated and untreated MS, as well as patients with RRMS (Relapsing-Remitting MS), SPMS (Secondary-progressive MS), and PPMS (Primary-progressive MS) forms, suggest IL-33 may be involved in pathogenesis (Jafarzadeh, 2016).
Infectious origin by common microbial infections such as rubella, (Granieri, 2001), Epstein-Barr virus, (Bowen, 2005; Calabresi, 2005), or human herpesvirus (Calabresi, 2005; Evans, 2002; Granieri, 2001)
Environmental factors:
Country of origin, socioeconomic status, personal habits, vitamin deficiency, and obesity
A 2009 study found that patients with MS, who smoked, demonstrated the following: increased Expanded Disability Status Scale scores, unfavorable correlations with lesion measures (including increased number of CE lesions), decreased brain parenchymal fraction, and lateral ventricle volume and third ventricle width increases (Zivadinov, 2009).
Environmental risk factors have a dominant role in the pathogenesis of MS and can be affected by life style changes. High level of free fatty acids in the blood among a South Indian population with MS could indicate that high free fatty acids in blood may have a role in MS pathogenesis (Dhanya, 2016).
Geographic risk of developing MS seems to be set early in life and does not change after adolescence (Multiple Sclerosis Practice Essentials – Medscape, 2015)
People with HIV have a decreased chance of developing MS over their lifetime (Gold, 2015)
Researchers have disproven the following suspected causes: aspartame, increased exposure to heavy metals, and history of traumatic injury (NMSS, 2015)
+++
II. Diagnostic Procedures
++
Diagnosed by evaluation of clinical presentation and disease markers (Multiple sclerosis - PubMed Health, 2011; Bowen, 2005; Calabresi, 2005; Lublin, 2005).
Signs and Symptoms:
Asymmetric, variable distribution of symptoms depending on plaque location (Multiple sclerosis - PubMed Health, 2011; Bowen, 2005; Calabresi, 2005; Lublin, 2005)
Motor: Upper motor neuron symptoms including paresis, spasticity, hyperreflexia, Babinski sign, muscle spasms (sometimes painful), difficulty with small movements, and extremity weakness; in late stages may have progressive dysarthria and trouble with swallowing (Rovaris, 2006).
Cerebellar: Ataxia, intention tremor, dysphagia, dysarthria, loss of balance
Sensory: Paresthesias, dysesthesias, trigeminal neuralgia, Lhermitte's sign, hearing loss, abnormal nerve reflexes
Visual: Optic neuritis (visual loss), diplopia, nystagmus, eye discomfort, abnormal pupil responses, changes in visual fields or eye movements, decreased visual acuity, some experience intermittent visual blurring from hyperthermia (Balcer, 2015), internuclear opthalmoplegia, skew deviation, retrochiasmal visual pathway demyelination, intermediate uveitis, CN III, IV or VI palsies (Francis, 2013)
Autonomic: Bladder dysfunction (urgency, hesitancy, frequency, incontinence); bowel (constipation, urgency, stool leakage); sexual dysfunction (erection and vaginal lubrication issues); Female Sexual Dysfunction (FSD) is prevalent among with MS; often underdiagnosed (Scheepe, 2017)
Other: Dizziness, vertigo, heat sensitivity, cognitive impairment, fatigue (often worse in the late afternoon), depression, gait difficulties, facial pain, decreased attention span, poor judgment, memory loss, difficulty with reasoning/solving problems, depression, pain and cognitive dysfunction (Miller, 2012; Rovaris, 2006; Miller, 2007).
Diagnostic Tests
MRI: of the brain and spinal cord is the most sensitive tool for confirming diagnosis; detects presence of demyelinating white matter plaques in CNS, and allows the clinician to follow the disease progression.
In working memory task performance, functional MRI demonstrated increased activation of the right dorsolateral prefrontal cortex and anterior cingulate cortex, as well as during non-dominant hand movements. It is hypothesized that "activation of cognitive control brain regions, particularly in the right hemisphere, may be an important mechanism allowing patients with MS to accommodate to the neural disruption caused by this disease." It is predicted that this may also allow identification of at risk groups as well as interventions to help improve cognitive control. (Aharony, 2015; Colorado, 2011).
Common lesion locations include periventricular and juxtacortical white matter regions, brainstem, cerebellum, and spinal cord, especially cervical; and high lesion loads are associated with a greater risk of a second relapse (in patients with clinically isolated syndrome (CIS)), and future progression (Tintore, 2015).
MRI-derived atrophy measures are the most frequently used measure of the neurodegenerative process in MS. Gd + activity and T2 lesion volume accumulation can provide indications of the 'inflammatory' component of MS (Sicotte, 2011). Rate of atrophy ranges from 0.6% to 1.35% per year, 4 times greater than that of normal aging (Sahraian, 2010; Rovira, 2010)
The preferred method for confirming an MS diagnosis and monitoring disease progression; However, abnormalities seen do not always correlate with clinical signs and reported symptoms (Multiple Sclerosis Workup – Medscape, 2015)
CT: useful to detect large lesions; limited ability to detect small plaques; contrast enhancement increases sensitivity
CSF analysis: not sensitive or specific for MS; elevated IgG and total protein; presence of oligoclonal bands
Visual evoked potential (VEP): prolonged latency and conduction disturbances indicate optic nerve involvement
Blood work: performed to rule out systemic conditions that mimic MS
Optical coherence tomography (OCT): picks up changes associated with the retinal nerve fiber layer thickness. It is validated, non-invasive, and inexpensive (Bock, 2013).
The 2010 McDonald Criteria lists the additional data needed for MS diagnosis based on five different clinical presentations which are defined by number of attacks, objective clinical evidence of a certain number of lesions, or by an insidious neurological progression suggestive of MS. Additional data needed may include demonstration of dissemination in time and space and positive CSF. See table four in Polman, 2011 for full criteria. (Polman, 2011).
Diagnosis of MS: clinical presentation has no better explanation and criteria are fulfilled
Diagnosis of "possible MS": criteria are not completely fulfilled and suspicious
Diagnosis is "not MS": clinical presentation is better explained by another diagnosis during evaluation
High sensitivity and specificity allows for an early diagnosis of Multiple Sclerosis in patients who have experienced a typical clinically isolated syndrome
Dissemination in space can be shown by clinical or imaging evidence of lesions in two or more of four key areas of the CNS: the periventricular, juxtacortical, and infratentorial areas, as well as the spinal cord (Selchen, 2012)
++
MS is most commonly a progressive disease; it is random in terms of frequency and severity of exacerbations, rate of progression, and accrual of disability; average survival rate is 35 to 40 years from time of diagnosis; overall life span limited by ~ 7 yrs (Lublin, 2005). MS may reduce life-span, possibly related to comorbidities, especially psychiatric disorders (Scalfari, 2013).
Categories of MS: (Bowen, 2005; Calabresi, 2005; Evans, 2002; Lublin, 2005)
Relapsing-remitting MS (RRMS): relapses with full or near full recovery; lack of disease progression between relapses; 85% of patients with MS start with an acute isolated episode, often the first clinical episode (Miller, 2012). Disease usually starts in late 20s (Tur, 2015). Most common form of the disease (Lövblad, 2010)
Secondary-progressive MS (SPMS): initial relapsing-remitting course, followed by irreversible progression with or without occasional relapses, minor remissions, and plateaus; SPMS occurs about 15-20 years after symptom onset (Compston, 2008).
Primary-progressive MS (PPMS): disease progression from onset without plateaus and remissions or with occasional plateaus and temporary minor improvements; symptoms usually occur around age 40 (Compston, 2008; Scalfari, 2011). Affects 10-15% of MS patients; management focuses on symptoms (Lövblad, 2010)
Progressive-relapsing MS (PRMS): progressive disease from onset with clear acute relapses, with or without recovery, with continued progression between relapses; commonly seen in people who develop MS after age 40. Relatively rare affecting approximately 5% of patients (Faguy, 2016)
Clinically Isolated Syndrome: single symptomatic episode lasting a minimum of 24 hours; can be a precursor to developing relapsing-remitting MS or another type of the disease; 88% of patients with this develop MS within 14 years (Costello, 2010; Lövblad, 2010)
Benign MS: "considered to be relapsing-remitting MS with a disability score of less than 3 on the Expanded Disability Status Scale for a period of at least 10 years" (Selchen, 2012)
Asymptomatic MS: lesions detected incidentally on an MR examination or during an autopsy with no clinical indications of the disease; can also be known as preclinical, subclinical or radiologically isolated MS (Faguy, 2016)
Fulminant MS: rare, rapidly progression of disease that can lead to severe disability or death within weeks or months of onset (Milo, 2014; Selchen, 2012)
Unfavorable prognostic indicators include:
Male sex, later age of onset, progressive course from onset frequent exacerbations, poor recovery from exacerbations, involvement of cerebellar or motor functions (Lublin, 2005), color vision impairment (Martínez-Lapiscina, 2014), and short inter-attack interval. (Aharony, 2015)
Duration of the disease is associated with disability; inverse relationship between disease duration and serum levels of vitamin D (Hatamian, 2013)
Favorable prognostic indicators:
2 or more pregnancies, later onset of menarche (D'Hoogie, 2011) female gender, younger age of onset, and relapsing onset instead of progressive onset (Baghizadeh, 2013),
Exacerbating factors: viral and bacterial infections (Granieri, 2001), major organ diseases, major and minor life stress events.
Expected sequelae (structural and functional) (Multiple sclerosis - PubMed Health, 2011)
Left untreated: after onset, 30% of MS patients will develop significant physical disability within 20-25 years
Life expectancy: only slightly less than healthy individuals (with survival rate linked to disability). Death usually results from secondary complications (50-66%), but can also be due to primary complications (such as suicide).
Marburg variant of MS: an acute and sudden developmental form of the disease that can lead to coma or death within days (Elenein, 2011).
With disease progression greater loss of function is observed. After attacks, most patients return to near-normal function but less improvement is seen during remittance. With progression, many must use assistive devices including wheelchairs; however, patients with caretakers can usually remain at their home.
A study comparing MS groups with controls found that patients with MS are at an increased risk of cardiovascular diseases, specifically: the incidence rate ratio for MI was 1.85 (95% confidence interval (CI) 1.59 to 2.15), stroke was 1.71 (95% CI 1.46 to 2.00), heart failure was 1.97 (95% CI 1.52 to 2.56) and for Atrial Fibrillation/Flutter was 0.63 (95% CI 0.46 to 0.87) (Jadidi, 2013).
+++
IV. Medical and Surgical Management:
++
No cure for MS
Treatment is to decrease the frequency and severity of relapses and manage symptoms (Anon., 2011; Calabreski, 2006; Evans, 2002; NMSS, 2006).
Disease-modifying medications:
interferon-B-1a (Avonex, Rebif) and interferon-B-1b (Betaseron) are FDA approved for RRMS and reduce brain lesion development and decrease the frequency and severity of relapses; glatiramer acetate (Copaxone) is FDA approved for RRMS and reduces the severity of relapses; Mitoxantrone (Novantrone) is a chemotherapeutic agent used to decrease frequency of relapses in patients with SPMS, PRMS, or worsening RRMS but can only be used for 2-3 years due to cardiotoxicity
In January 2013, the FDA approved self-injection of interferon beta-1a (Rebif) in the treatment of relapsing forms of multiple sclerosis due to its ease of use (Jeffrey, 2013). Main side effect of this first-line disease modifying drug (DMD) is flu-like symptoms (Tur, 2015).
Natalizumab (Tysabri®) reduced relapse rate by 68% as a monotherapy and by 55% when used in combination with weekly intramuscular (IM) injections of interferon beta-1a (IFNβ-1a). "Responders walked 25 feet an average of 24%–45% faster than nonresponders." (Cadavid, 2013).
Disease-modifying therapies of either interferon beta or glatiramer acetate in patients with RRMS are not associated with depression status, development of depression or exacerbation of depressive symptoms (Kirzinger, 2013).
With glatiramer acetate, main side effects include immediate post-injection reaction including vasodilation, chest pain, dyspnea, palpitations, and tachycardia (Tur, 2015).
Delayed disease progression was found with the use of interferon beta-1b (Khatri, 2011).
Fingolimod (Gilenya): once daily oral medication with 54% relative risk reduction of clinical relapses. Mechanism of action: interferes with the ability of lymphocytes to leave nodes and enter the CNS (Wingerchuk, 2014).
Teriflunomide (Aubagio): once daily oral medication with 31% relative risk reduction of clinical relapses.
Contraindications: pregnancy (Wingerchuk, 2014)
Main side effect(s) of this first-line DMD are hair loss, peripheral neuropathy, and elevated blood pressure (Tur, 2015).
Dimethyl fumarate (Tecfidera): twice-daily oral medication with 51-53% relative risk reduction of clinical relapses. (Wingerchuk, 2014) Main side effect of this first-line DMD is flushing and gastrointestinal events (Tur, 2015).
Methotrexate, azathioprine (Imuran), intravenous immunoglobulin (IVIg) and cyclophosphamide (Cytoxan) may also be used if the above drugs are not working well (Multiple sclerosis - PubMed Health, 2011).
Mitoxantrone and natalizumab are used as the last resort for medications due to their adverse side effects (Derwenskus, 2013).
Second line DMDs such as IV natalizumab, oral fingolimod, and IV alemtuzumab have proved effective in reducing the number of relapses by around 55-70% (Polman, 2006; Miller, 2003; Kappos, 2010; Cohen, 2010).
If a patient with active MS has had at least two relapses over the past year together with MRI evidence of disease activity, natalizumb and alemtuzumab can be prescribed even if they are not on any DMDs (NHS England, 2014).
In patients with progressive multifocal leukoencephalopathy (PML), oral fingolimod can be prescribed as an alternative to natalizumab NHS England, 2014; Tur, 2014).
Reduction in active gadolinium-enhanced lesions and annual relapse rate in patients RRMS was found with the use of a myelin peptide skin patch (Walczak, 2013).
Management of MS: Dalfampridine (Ampyra): is potassium blocker to help improve walking (Ampyra, 2010).
Management of spasticity: pharmacological includes Baclofen, Dantrolene, Clonazepam, Diazepam, Tizanidine, Botox injections; surgical includes neurosurgical ablative procedures and tendonotomy for fixed contractures
Management of bladder dysfunction: pharmacological: cholinergic medications, intermittent self-catheterization, indwelling or suprapubic catheter
Multiple medications for symptom management of erectile dysfunction, tremor, constipation, depression, fatigue (Amantadine), and pain.
Steroids may be used to decrease the severity of attacks.
Management of fatigue: Modafinil (Provigil), and its (R)-enantiomer Armodafinil (Nuvigil) are FDA approved for narcolepsy, shift-work sleep disorder, and obstructive sleep apnea (Braley, 2010).
Educational programs, especially cognitive-behavioral therapy (CBT) based approaches, have a positive effect on reducing fatigue (Wendebourg, 2017)
Management of tremor: disabling tremor may be reduced by deep brain stimulation of ventral intermediate nucleus. Respondents showed improvements in quality of life and function (Zakaria, 2013).
Plasmapharesis is a medical method shown to significantly reduce autoimmune inflammatory "installed" syndrome; key to effective treatment is starting the procedure as close to the beginning of illness as possible (Vlaic, 2017)
Patients treated with Autologous Hematopoietic Stem Cell Transplantation (AHSCT) shown to have remission of disease progression for 5 years after transplant (Muraro, 2017)
Patients taking a disease-modifying drug and who have relapsing-remitting MS should have a follow-up scan 6 to 12 months following treatment (Klawiter, 2013)
+++
V. Implications for Therapeutic Management:
++
Preferred Practice Patter ns (APTA, 2017)
Physical, Occupational, and Speech Therapy aimed at restoring or maximizing function (National Multiple Sclerosis Society, 2006)
Management of MS requires a multi-disciplinary approach and is based on education, prevention, and self-management (Tur, 2015). The importance of healthy eating and regular exercise are important, especially for long-term mobility in MS (Tur, 2015).
Physical Therapy Implications:
Addresses impairments in:
Gait, balance, mobility, strength, endurance, and ROM
Interventions for
Pain (T.E.N.S., moist heat packs, massage, and exercise), ataxia (Frenkel's exercises), maintaining ROM, strengthening muscles, improving coordination, and spasticity; recommendation of appropriate assistive aids (WalkAide and Bioness provide functional electrical stimulation for utilization during walking) and seating options (for non-ambulatory patients); patient education on energy conservation and HEP (National Multiple Sclerosis Society, 2006), and prevention of cardiovascular patients (especially among women) (Jadidi, 2013).
Disease Stages and Intervention Strategies (Bello-Haas, 2002)
Early
Functional Limitations: visual impairments, sensory impairments, fatigue, unilateral muscle weakness, mild gait disturbance, mild balance impairments, mild cognitive impairments
Restorative Interventions: Balance training, ROM and strengthening exercises, endurance exercises
Compensatory Interventions: Energy conservation, education about the disease, support groups, compensatory strategies for visual, cognitive, sensory impairments, assess potential need for adaptive or assistive devices, ergonomic modifications
Preventative Interventions: Stretching exercises, energy conservation, stress management, and relaxation techniques
Middle
Functional Limitations: Increasing muscle weakness, progressive loss of mobility and ADLs, impaired balance, spasticity, ataxia, tremor, sensory loss, cognitive impairments
Restorative Interventions: See early stage
Compensatory Interventions: Compensatory strategies for cognitive impairments (e.g. memory aids, cueing devices, environmental structuring and labeling), orthoses, supportive/assistive/adaptive devices, compensatory strategies for sensory loss, proprioceptive loading, evaluate need for mobility devices (e.g. scooter or wheelchair)
Preventative Interventions: Monitor for affective disorders, stretching and ROM exercises
Late
Functional Limitations: Severe muscle weakness, wheelchair bound or restricted to bed, respiratory compromise, dysarthria, dysphagia, dependent in mobility, self-care, and ADLs, bladder dysfunction, incontinence, severe visual impairments, cognitive deficits
Compensatory Interventions: Education of caregivers on lifting, transfers, bed mobility
Preventative Interventions: Pulmonary hygiene, skin care hygiene, education of caregivers on prevention of secondary complications
Functional status, balance, fatigue, spasticity and quality of life was improved with PT supervised group exercise training in patients with MS (Tarakci, 2013). There are also beneficial effects in walking speed, stepping endurance, stair climbing, and timed up and go test in MS patients who are gaining muscle strength due to exercise (Döring, 2011).
Increased Physical Activity correlates to an increase in the quality of life for patients with MS (Marck, 2014).
In order to improve spasticity, PT should include active and passive exercises using either a partner or equipment such as theraband (Döring, 2011).
Aquatic therapy and a standing frame can be used to train torso, limb, and respiratory muscles to maintain cardiovascular fitness in those who are unable to stand (Döring, 2011).
Functional e-stim has improved the following in patients with MS: Ankle dorsiflexion at initial contact, knee flexion at initial contact, and peak knee flexion during swing were significantly greater. Significant improvements were also made in the 10-minute walk test (Scott, 2013).
Diet, exercise, omega 3 supplementation, BMI, alcohol intake, fish consumption, Vitamin D supplementation and DMD, have been shown to decrease fatigue in MS patients. (Weiland, 2015)
Exercise in general has been suggested to potentially reduce MS fatigue (Andreasen, 2011)
Resistance training with moderate intensity is well tolerated and provides beneficial effects for moderately impaired MS patients (Dalgas, 2008)
Rhythmic Auditory Stimulation proven to be an effective rehabilitation method to improve gait kinetic parameters such as stride length, stride time, cadence and gait speed (Shahraki, 2017)
Cooling suit worn by individuals with MS significantly improved patients' levels of fatigue and independence in activities of daily living (Ozkan Tuncay, 2017)
Hydrotherapy, including cold baths, has become more prevalent in treatment with MS due to its ability to promote local and systemic hypothermia (Corvillo, 2017)
Precautions to therapy:
Uhthoff phenomenon (Fraser, 2012) -- patients with MS may experience an exacerbation in symptoms when overheated. General guidelines to avoid overheating include alternating periods of exercise with equal periods of rest, exercising in the am when it is cooler and the patient is more rested, avoiding exercise in hot environments, performing aquatic exercise in water temperature between 80 – 84°F, and wearing a cooling vest, with circulating liquid, during exercise (National Multiple Sclerosis Society, 2006; Nilsagard, 2006; White, 2004).
Symptoms of being overheated include: transient visual obscurations, dyschromatopsia, and contrast sensitivity changes. Symptoms usually disappear once normal body temperature has been restored -- anywhere from 60 minutes to 24 hours
If full recovery is not attained within 2 months, this may indicate that re-myelination will not occur (Fraser, 2012).
+++
VI. Consumer and Professional Resources
++
Mellen Center for Multiple Sclerosis
MS Connection
MS Trust
MS World
Multiple Sclerosis Foundation
National Multiple Sclerosis Society
NeedyMeds
The Consortium of MS Centers
The Multiple Sclerosis Association of America: MSAA
+
Aharony
S, Lam
O, Lapierre
Y, Corcos
J. Multiple sclerosis (MS) for the urologist: What should urologists know about MS? Neurourol Urodynam. 2015. doi:10.1002/nau.22713.
+
Ampyra [package insert]. Hawthorne, NY: Acorda Therapeutics, Inc.; 2010.
+
Baghizadeh
S, Sahraian
MA, Beladimoghadam
N. Clinical and demographic factors affecting disease severity in patients with multiple sclerosis.
Iran J Neurol. 2013;12(1):1–8.
[PubMed: 24250889]
+
Balcer
LJ, Miller
DH, Reingold
SC, Cohen
JA. Vision and vision-related outcome measures in multiple sclerosis.
Brain. 2015;138(1):11–27.
[PubMed: 25433914]
+
Blumenthal
S. Multiple sclerosis.
Radiol Technol. 2006;77(4):309–321, quiz 322-325.
[PubMed: 16543484]
+
Bock
M, Paul
F, Dörr
J. [Diagnosis and monitoring of multiple sclerosis : The value of optical coherence tomography.]. Nervenarzt. 2013 Feb 21. [Epub ahead of print] German. PubMed PMID: 23423630.
+
Bowen
J. Diagnosing multiple sclerosis and its imitators.
Phys Med Rehabil Clin N Am. 2005;16:359–81.
[PubMed: 15893677]
+
Braley
TJ, Chervin
RD. Fatigue in multiple sclerosis: mechanisms, evaluation, and treatment.
Sleep. Aug 2010;33(8):1061–7.
[PubMed: 20815187]
+
Cadavid
D, Jurgensen
S, Lee
S. Impact of natalizumab on ambulatory improvement in secondary progressive and disabled relapsing-remitting multiple sclerosis.
PLoS One. 2013;8(1):e53297. doi: 10.1371/journal.pone.0053297. Epub 2013 Jan 4. PubMed PMID: 23308186; PubMed Central PMCID: PMC3537666.
[PubMed: 23308186]
+
Calabresi
PA. Multiple sclerosis: approaches to early diagnosis and new treatment options. Adv Stud Med. 2005;5(7):368–75.
+
Cohen
JA, Barkhof
F, Comi
G
et al.. Oral fingolimod or intramuscular interferon for relapsing multiple sclerosis.
N Engl J Med 2010;362:402–15
[PubMed: 20089954]
+
Colorado
RA, Shukla
K, Zhou
Y, Wolinsky
JS, Narayana
PA. Multi-task functional MRI in multiple sclerosis patients without clinical disability. Neuroimage. 2012 Jan 2;59(1):573–81. doi: 10.1016/j.neuroimage.2011.07.065. Epub 2011 Aug 5. PubMed PMID: 21840409; PubMed Central PMCID: PMC3195884.
+
Cook
SD, Dhib-Jalbut
S, Dowling
P,
et al. Use of Magnetic Resonance Imaging as Well as Clinical Disease Activity in the Clinical Classification of Multiple Sclerosis and Assessment of Its Course: A Report from an International CMSC Consensus Conference, March 5–7, 2010.
Int J MS Care. 2012;14(3):105–114.
[PubMed: 24453741]
+
Corvillo
I, Varela
E, Armijo
F, Alvarez-Badillo
A, Armijo
O, Maraver
F. Efficacy of aquatic therapy for multiple sclerosis: a systematic review. Eur J Phys Rehabil Med. February 2017 doi:10.23736/S1973-9087.17.04570-1.
+
Cotsapas
C, Voight
B, Todd
J,
et al. Pervasive Sharing of Genetic Effects in Autoimmune Disease. Plos Genetics [serial online]. August 2011;7(8):1–8. Available from: Academic Search Premier, Ipswich, MA. Accessed February 02, 2013.
+
Dalgas
U, Stenager
E, Ingemann-Hansen
T. Review: Multiple sclerosis and physical exercise: recommendations for the application of resistance-, endurance- and combined training. Mult Scler J. 2008;14(1):35–53.
+
Derwenskus
J. Evolving Treatment Strategies in the Management of Multiple Sclerosis. Journal of Managed Care Medicine. 2013;16(3):16–19.
+
D'hooghe
MB, Haentjens
P, Nagels
G, D'Hooghe
T, De Keyser
J. Menarche, oral contraceptives, pregnancy and progression of disability in relapsing onset and progressive onset multiple sclerosis.
J Neurol. 2012;259(5):855–861.
[PubMed: 21993617]
+
Dhanya
J, Suresh
K. Identifying clues to molecular etiology of multiple sclerosis in South Indian patients.
Mult Scler Relat Disord. 2016;5:7–11.
[PubMed: 26856937]
+
Döring
A, Pfueller
CF, Paul
F, Dörr
J. Exercise in multiple sclerosis -- an integral component of disease management.
EPMA J. 2011;3(1):2.
[PubMed: 22738091]
+
Elenein
RG, Sharer
LR, Cook
SD, Pachner
AR, Michaels
J, Hillen
ME. A second case of Marburg's variant of multiple sclerosis with vasculitis and extensive demyelination. Mult Scler. 2011 Dec;17(12):1531–8. doi: 10.1177/1352458511414042. Epub 2011 Aug 3. PubMed PMID: 21816761.
+
Evans
CF, Shriver
LP. Multiple sclerosis: emerging opportunities for therapeutic intervention. Current Drug Targets – CNS & Neurological Disorders. 2002;1:17–30.
+
Faguy
K. Multiple Sclerosis: An Update.
Radiol Technol. 2016;87(5):529–550.
[PubMed: 27146176]
+
Francis
CE. Visual issues in multiple sclerosis.
Phys Med Rehabil Clin N Am. 2013;24(4):687–702.
[PubMed: 24314686]
+
Fraser
C, Davagnanam
I, Radon
M, Plant
G. The time course and phenotype of Uhthoff phenomenon following optic neuritis. Multiple Sclerosis (Houndmills, Basingstoke, England) [serial online]. July 2012;18(7):1042–1044. Available from: MEDLINE with Full Text, Ipswich, MA. Accessed February 22, 2013.
+
Gold
J, Goldacre
R, Maruszak
H, Giovannoni
G, Yeates
D, Goldacre
M. HIV and lower risk of multiple sclerosis: beginning to unravel a mystery using a record-linked database study. J Neurol Neurosurg Psychiatry. 2014:jnnp - 2014-307932. doi:10.1136/jnnp-2014-307932.
+
Granieri
E, Casetta
I, Tola
MR, Ferrente
P. Multiple sclerosis: infectious hypothesis.
Neurol Sci. 2001;22:179–85.
[PubMed: 11603624]
+
Hatamian
H, Bidabadi
E, Saadat
SM, Saadat
NS, Kazemnezhad
E, Ramezani
H, Bakhshayesh
B. Is serum vitamin D levels associated with disability in patients with newly diagnosed multiple sclerosis?.
Iran J Neurol. 2013;12(2):41–6.
[PubMed: 24250900]
+
Jadidi
E, Mohammadi
M, Moradi
T. High risk of cardiovascular diseases after diagnosis of multiple sclerosis. Mult Scler. 2013 Jan 30. [Epub ahead of print] PubMed PMID: 23364857.
+
Jafarzadeh
A, Mahdavi
R, Jamali
M, Hajghani
H, Nemati
M, Ebrahimi
H-A. Increased Concentrations of Interleukin-33 in the Serum and Cerebrospinal Fluid of Patients with Multiple Sclerosis.
Oman Medical Journal. 2016;31(1):40–45.
[PubMed: 26813806]
+
Kampman
M, Brustad
M. Vitamin D: a candidate for the environmental effect in multiple sclerosis - observations from Norway.
Neuroepidemiology [serial online]. 2008;30(3):140–146. Available from: MEDLINE with Full Text, Ipswich, MA. Accessed February 16, 2013.
[PubMed: 18382112]
+
Kappos
L, Radue
EW, O'Connor
P
et al.. A placebo-controlled trial of oral fingolimod in relapsing multiple sclerosis
N Engl J Med 2010;362:387–401
[PubMed: 20089952]
+
Khatri
B, Barkhof
F, Comi
G, Hartung
HP, Kappos
L, Montalban
X, Pelletier
J, Stites
T, Wu
S, Holdbrook
F, Zhang-Auberson
L, Francis
G, Cohen
JA; TRANSFORMS Study Group. Comparison of fingolimod with interferon beta-1a in relapsing-remitting multiple sclerosis: a randomised extension of the TRANSFORMS study. Lancet Neurol. 2011 Jun;10(6):520–9. doi: 10.1016/S1474-4422(11)70099-0. Epub 2011 May 13. PubMed PMID: 21571593.
+
Kirzinger
SS, Jones
J, Siegwald
A, Crush
AB. Relationship between disease-modifying therapy and depression in multiple sclerosis.
Int J MS Care. 2013;15(3):107–12.
[PubMed: 24453772]
+
Klawiter
EC. Current and new directions in MRI in multiple sclerosis. Contin Minneap Minn. 2013;19(4 Multiple Sclerosis):1058–1073.
+
Karussis
D. The diagnosis of multiple sclerosis and the various related demyelinating syndromes: A critical review.
J Autoimmun. 2014;48:134–42.
[PubMed: 24524923]
+
Lövblad
KO, Anzalone
N, Dörfler
A, Essig
M, Hurwitz
B, Kappos
L, Lee
SK, Filippi
M. MR imaging in multiple sclerosis: review and recommendations for current practice.
AJNR Am J Neuroradiol. 2010;31(6):983–9.
[PubMed: 20019103]
+
Lublin
FD. Clinical features and diagnosis of multiple sclerosis.
Neurol Clin. 2005;23:1–15.
[PubMed: 15661085]
+
Marck
CH, Hadgiss
EJ, Weiland
TJ, van der Meer
DM, Periera
NG, Jelinek
GA. Physical activity and associated levels of disability and quality of life in people with multiple sclerosis: a large international survey.
BMC Neurology 2014;14:143. doi: 10.1186/1471-2377-14-143
[PubMed: 25016312]
+
Marrie
RA, Elliott
L, Marriott
J,
et al. Effect of comorbidity on mortality in multiple sclerosis.
Neurology. 2015;85(3):240–247.
[PubMed: 26019190]
+
Martínez-lapiscina
EH, Ortiz-pérez
S, Fraga-pumar
E, Martínez-Heras
E, Gabilondo
I, Llufriu
S, Bullich
S, Figueras
M, Saiz
A, Sánchez-Dalmau
B, Villoslada
P. Colour vision impairment is associated with disease severity in multiple sclerosis. Mult Scler. 2014;XX(X): 1–10.
+
Miller
DH, Chard
DT, Ciccarelli
O. Clinically isolated syndromes.
Lancet Neurol 2012;11:157–69
[PubMed: 22265211]
+
Miller
DH, Khan
OA, Sheremata
WA
et al.. A controlled trial of natalizumab for relapsing multiple sclerosis.
N Engl J Med 2003;348:15–23
[PubMed: 12510038]
+
Miller
DH, Leary
SM. Primary-progressive multiple sclerosis.
Lancet Neurol 2007;6:903–912
[PubMed: 17884680]
+
Milo
R, Miller
A. Revised diagnostic criteria of multiple sclerosis.
Autoimmun Rev. 2014;13(4-5):518–524.
[PubMed: 24424194]
+
Muraro
PA, Pasquini
M, Atkins
HL,
et al. Long-term Outcomes After Autologous Hematopoietic Stem Cell Transplantation for Multiple Sclerosis. JAMA Neurol. February 2017. doi:10.1001/jamaneurol.2016.5867.
+
NHS England. Clinical Commissioning Policy: Disease Modifying Therapies for Patients with Multiple Sclerosis.2014
+
Nilsagård
Y, Denison
E, Gunnarsson
L-G. Evaluation of a single session with cooling garment for persons with multiple sclerosis--a randomized trial.
Disabil Rehabil Assist Technol. 2006;1(4):225–233.
[PubMed: 19260170]
+
Nischwitz
S, Muller-Myhsok
B, Weber
F. Risk conferring genes in multiple sclerosis.
FEBS Lett. Dec 1 2011;585(23):3789–97.
[PubMed: 21439284]
+
Özkan
Tuncay F, Mollaoğlu
M. Effect of the cooling suit method applied to individuals with multiple sclerosis on fatigue and activities of daily living. J Clin Nurs. March 2017. doi:10.1111/jocn.13788.
+
Polman
CH, O'Connor
PW, Hardova
E
et al.. A randomized, placebo-controlled trial of natalizumab for relapsing multiple sclerosis.
N Engl J Med 2006;354:899–910
[PubMed: 16510744]
+
Polman
CH, Reingold
SC, Banwell
B, Clanet
M, Cohen
J, Filippi
M, Fujihara
K, Havdova
E, Hutchinson
M, Kappos
L, Lubin
F, Montalban
X, O'Connor
P, Sandberg-Wolheim
M, Thompson
A, Waubant
E, Weinshenker
B, Wolinsky
J. Diagnostic criteria for multiple sclerosis: 2010 revisions to the McDonald criteria.
Ann Neurol. 2011;69(2):292–302.
[PubMed: 21387374]
+
Prat
E, Martin
R. The immunopathogenesis of multiple sclerosis.
J Rehabil Res Dev. 2002;39(2):187–200.
[PubMed: 12051463]
+
Rovaris
M, Confavreux
C, Furlan
R
et al.. Secondary progressive multiple sclerosis: current knowledge and future challenges.
Lancet Neurol 2006;5:343–54
[PubMed: 16545751]
+
Rovira
A, Tintoré
M, Alvarez-Cermeño
JC, Izquierdo
G, Prieto
JM. [Recommendations for using and interpreting magnetic resonance imaging in multiple sclerosis]. Neurol Barc Spain. 2010;25(4):248–265.
+
Sahraian
MA, Eshaghi
A. Role of MRI in diagnosis and treatment of multiple sclerosis.
Clin Neurol Neurosurg. 2010;112(7):609–615.
[PubMed: 20417027]
+
Scalfari
A, Knappertz
V, Cutter
G
et al.. Mortality in patients with multiple sclerosis.
Neurology 2013;81:184–92
[PubMed: 23836941]
+
Scalfari
A, Neuhaus
A, Daumer
M
et al.. Age and disability accumulation in multiple sclerosis.
Neurology 2011;77:1246–52
[PubMed: 21917763]
+
Scheepe
JR, Alamyar
M, Pastoor
H, Hintzen
RQ, Blok
BFM. Female sexual dysfunction in multiple sclerosis: Results of a survey among Dutch urologists and patients.
Neurourol Urodyn. 2017;36(1):116–120.
[PubMed: 26493281]
+
Scott
SM, van der Linden
ML, Hooper
JE, Cowan
P, Mercer
TH. Quantification of gait kinematics and walking ability of people with multiple sclerosis who are new users of functional electrical stimulation. J Rehabil Med. 2013 Feb 14. doi: 10.2340/16501977-1109.
+
Shahraki
M, Sohrabi
M, Taheri
Torbati HR, Nikkhah
K, NaeimiKia
M. Effect of rhythmic auditory stimulation on gait kinematic parameters of patients with multiple sclerosis.
J Med Life. 2017;10(1):33–37.
[PubMed: 28255373]
+
Sicotte
NL. Neuroimaging in Multiple Sclerosis: Neurotherapeutic Implications.
Neurotherapeutics. 2011;8(1):54–62.
[PubMed: 21274685]
+
Sumelahti
ML, Hakama
M, Elovaara
I, Pukkala
E. Causes of death among patients with multiple sclerosis. Mult Scler. 2010 Dec;16(12):1437–42. doi: 10.1177/1352458510379244. Epub 2010 Sep 8. PubMed PMID: 20826526.
+
Tarakci
E, Yeldan
I, Huseyinsinoglu
BE, Zenginler
Y, Eraksoy
M. Group exercise training for balance, functional status, spasticity, fatigue and quality of life in multiple sclerosis: a randomized controlled trial.
Clin Rehabil. 2013;27(9):813–22.
[PubMed: 23543341]
+
Tintore
M, Rovira
A, Rio
J
et al.. Defining high, medium and low impact prognostic factors for developing multiple sclerosis.
Brain 2015;138:1863–74
[PubMed: 25902415]
+
Traboulsee
A, Simon
JH, Stone
L,
et al. Revised Recommendations of the Consortium of MS Centers Task Force for a Standardized MRI Protocol and Clinical Guidelines for the Diagnosis and Follow-Up of Multiple Sclerosis.
AJNR Am J Neuroradiol. 2016;37(3):394–401.
[PubMed: 26564433]
+
Tsang
BK-T, Macdonell
R. Multiple sclerosis- diagnosis, management and prognosis.
Aust Fam Physician. 2011;40(12):948–955.
[PubMed: 22146321]
+
Tur
C, Montalban
X. Natalizumab: risk stratification of individual patients with multiple sclerosis.
CNS Drugs 2014;28:641–48
[PubMed: 24942634]
+
Tur,
C, Thompson,
A. Early accurate diagnosis crucial in multiple sclerosis. Practitioner. September 2015;259(1785):21–21.
+
Vlaic
S, Poalelungi
V, Bălăeţ
M, Purcărea
V, Bălăeţ
C, Coculescu
B. Pathophysiological changes of inflammatory syndrome in multiple sclerosis after instituting therapeutic plasmapheresis.
J Med Life. 2017;10(1):50–53.
[PubMed: 28255377]
+
Walczak
A, Siger
M, Ciach
A, Szczepanik
M, Selmaj
K. Transdermal application of myelin peptides in multiple sclerosis treatment.
JAMA Neurol. 2013;70(9):1105–9.
[PubMed: 23817921]
+
Wallin
MT, Culpepper
WJ, Coffman
P,
et al. The Gulf War era multiple sclerosis cohort: age and incidence rates by race, sex and service.
Brain. 2012;135(6):1778–1785.
[PubMed: 22628389]
+
Weiland
TJ, Jelinek
GA, Marck
CH, Hadgkiss
EL, van der Meer
DM, Pereira
NG, Taylor
KL. Clinically Significant Fatigue: Prevalence and Associated Factors in an International Sample of Adults with Multiple Sclerosis Recruited via the Internet. PLoS One. 2015;10(2). doi:10.1371/journal.pone.0115541.
+
Wendebourg
MJ, Heesen
C, Finlayson
M, Meyer
B, Pöttgen
J, Köpke
S. Patient education for people with multiple sclerosis-associated fatigue: A systematic review.
PloS One. 2017;12(3):e0173025. doi:10.1371/journal.pone.0173025.
[PubMed: 28267811]
+
White
LJ, Dressendorfer
RH. Exercise and multiple sclerosis.
Sports Medicine. 2004;34(15):1077–1100.
[PubMed: 15575796]
+
Wingerchuk
DM, Carter
JL. Multiple sclerosis: Current and Emerging Disease-Modifying Therapies and Treatment Strategies.
Mayo Clin Proc. 2014;89(2):225–40.
[PubMed: 24485135]
+
Zakaria
R, Vajramani
G, Westmoreland
L, Fletcher
N, Eldridge
P, Alusi
S, Osman-Farah
J. Tremor reduction and quality of life after deep brain stimulation for multiple sclerosis-associated tremor.
Acta Neurochir (Wien). 2013;155(12):2359–64.
[PubMed: 23975649]
+
Zivadinov
RM, Weinstock-Guttman
BM, Hashmi
KM,
et al. Smoking is associated with increased lesion volumes and brain atrophy in multiple sclerosis.
Neurology. 2009;73(7):504–510.
[PubMed: 19687451]
++
Muscular dystrophy (MD) is heterogeneous group of inherited disorders characterized by progressive muscle weakness and degeneration (Lovering, 2005). The focus of this diagnostic outline will be on the most common type, Duchenne muscular dystrophy (DMD).
Classifications of MD: (1) Duchenne, (2) Becker (BMD), (3) congenital, (4) myotonic, (5) Emery-Dreifus, (6) facioscapulohumeral, (7) limb-girdle, (8) oculopharyngeal , and (9) distal (Emery 2002)
Classification based on: mode of inheritance, age at onset, rate of progression, muscles involved, morphology of muscle, and genetic markers.
The first six types are the most prevalent and exhibit initial symptoms in infancy, childhood, or adolescence. The remaining three occur less often and are often characterized by adult onset (Emery, 2002).
++
++
The two most common forms of the disorder are DMD and BMD.
Both involve X-linked recessive traits with mutations in the dystrophin gene (Lovering, 2005; Zhou, 2006).
Most mutations in the dystrophin gene are deletions, which are identified in 65% of patients with DMD and 85% of patients with BMD (Bushby, 1992).
BMD is the result of abnormal quality or quantity of dystrophin (Lovering, 2005; Zhou, 2006).
DMD is caused by the absence of dystrophin on the cytoplasmic surface of the plasma membrane of the sarcolema of skeletal and cardiac muscle cells. DMD is the most common and severe childhood MD affecting 1 in every 3500 live male births (Kinali, 2007).
Many clinically similar muscular dystrophies are linked to a variety of distinct single-gene disorders. Molecular genetic mapping techniques have identified at least 29 different genetic loci that give rise to at least 34 varied clinical disorders (Lovering, 2005).
++
++
In DMD, the dystrophin-associated protein complex (DAPC) is destabilized when dystrophin is absent leading to diminished levels of member proteins, which leads to progressive fiber damage and membrane leakage (Straub, 1997; Nowak, 2004).
The exact mechanism leading to degeneration of muscle fibers due to dystrophin deficiency is not clear, however, cytoskeletal disruption, sarcolemmal instability and abnormal calcium homeostasis are believed to play a role (Deconinck, 2007).
Diminished or absent dystrophin causes fragility in muscle membrane with resulting necrosis of muscle.
Aberrant vascular response to exercise causes hypoxia of muscle tissue.
Protein (creatine kinase) leaks out of muscle into extra-cellular matrix and calcium leaks in.
Necrotic muscle tissue is replaced with adipose and fibrous tissue which makes muscles more susceptible to contracture.
The absence/disruption of dystrophin also influences central nervous system (CNS) function. In boys with DMD, evidence points to a "disordered CNS architecture, abnormalities in dendrites, and loss of neurons, all associated with neurons that normally express dystrophin" (Anderson, 2002).
++
++
Family history and physical examination findings are crucial to a complete diagnostic process (Richman 2011). The first recognizable signs of DMD usually appear by 2-3 years of age; symptoms of BMD usually begin later in childhood or adolescence (Kinali, 2007).
Early stage of DMD (generally from diagnosis to age 7 years) (Case, 2006)
Gross motor delays
Frequent falls and clumsiness
Poor endurance
Generalized and symmetrical muscle atrophy with weakness progressing proximal to distal
Range of motion (ROM) generally within normal limits (WNL)
Muscle tightness or contractures
Predictable patterns of muscle weakness
Hip extensors, abductors, adductors and flexors
Knee extensors
Ankle dorsiflexors
Abdominals
Neck flexors (sternocleidomastoid)
Shoulder depressors, extensors and abductors
Elbow extensors – weakness noted on testing but not usually functionally limiting
Large calf muscles secondary to adipose and fibrous replacement of muscle tissue (pseudohypertrophy)
Difficulty rising from the floor and compensatory pattern of "walking" up legs with hands referred to as Gower's sign.
Characteristic abnormal gait pattern:
Trandelenburg (waddling) – weak hip abductors
Increased lumbar lordosis because boys compensate for weakness by resting on anterior pelvic ligaments
Drop foot – weak dorsiflexors and tight G-S
Toe walking or decreased heel strike
Increased hip flexion to clear foot
In-toeing due to substitution of TFL for weak iliopsoas
Scapular retraction and shoulder extension to compensate for anterior shift of pelvis; decreased or absent arm swing
Possible scoliosis
Weakness of hip extensors and ankle dorsiflexors, and decrease in G-S ROM tend to predict cessation of ambulation and wheelchair use (Bakker, 2002).
Delayed speech/language skills
Cognitive abilities typically below, or at lower end of average IQ range (Richman, 2011; Wicksell, 2004)
Middle stage of DMD (7 years to early teens) (Case, 2006)
Progressive weakness in previously involved muscle groups
Add posterior tibialis to tight muscles
Functional limitations: difficulty rising from floor or chair, climbing stairs or managing slopes; frequent falls
Maximally compensated gait: increase in all previously mentioned compensatory strategies plus wide base of support and extreme neck flexion (chin tuck)
Adult stage of DMD (late teens into adulthood) (Case, 2006)
Function: loss of ambulation, shift to power wheelchair or electric scooter (usually by age 12 years), and dependence in most activities of daily living (ADL)
Progressive muscle weakness: impacts function in upper extremities; small muscles required for writing and computer use not as affected
Involvement of involuntary muscles
Loss of shoulder and elbow stability
Deformity – scoliosis develops within two years of being in wheelchair fulltime in about 90% of boys and will progress rapidly in about 60%
Death usually from respiratory failure/infection or cardiac abnormalities
Cognitive abilities (Wicksell, 2004; Busby, 2010b)
Dystrophin present in cortex, cerebellum and hippocampus
Most boys have IQ in normal range (average IQ=85)
About 30% have IQ less than 70
Increased risk for delays in speech/language development, motor planning and fine motor dexterity
Decreased storage for verbal information; instructions should be broken down into small chunks
No deterioration in function over time
++
++
During screening for suspected DMD based on child's history and physical presentation, serum muscle enzymes are tested: creatine kinase levels are raised significantly in patients with DMD (secondary to leakage through muscle membrane) and are 10-100 times normal ranges before the age of 5 (Daras, 2015).
Diagnosis confirmed by laboratory procedures including: dystrophin deletion/duplication testing (deletion or duplication mutation is found), muscle biopsy (dystrophin protein is absent), and genetic sequencing (mutation is found) (Bushby, 2010a).
Genetic testing for specific gene mutations in blood has emerged as the confirmatory test for DMD leaving electromyography (EMG) and muscle biopsy to be performed only in the small number of patients who are negative for dystrophin mutations (Wong, 2006; Manzor, 2009).
EMG studies can be used for differential diagnostic testing of MD from myopathies or other muscle disease. EMG characteristics specific to MD are termed "muscle membrane irritability" and include increased insertional activity, fibrillation potentials, and positive sharp waves. However, sometimes insertional activity can be decreased, which is often seen in chronic, end-stage muscular dystrophies (Paganoni, 2012).
EEG abnormalities: correlation between synaptic function and dystrophin absence (Anderson, 2002).
++
Progressive disease; currently no cure (Lovering, 2005).
Signs and symptoms of DMD are similar but more severe than those associated with BMD (Richman, 2011).
The onset of symptoms for DMD is usually between 2–5 years of age. Prognosis is poor; most patients lose the ability to walk by age 12 and experience cardiac or respiratory failure in their mid-30's (Richman, 2011).
Median life expectancy for DMD has increased from less than 30 years to 35.5 years when patients receive adjunctive ventilation therapies as compared to patients not receiving ventilation therapy (Kohler, 2009; Calvert, 2006).
Approximately 80% of DMD deaths are related to respiratory muscular failure, while 20% succumb to dilated cardiomyopathy (enlargement and weakening of left ventricle) (Kinali, 2007).
Secondary complications of MD: joint contractures, scoliosis, decreased functional mobility and independence with ADL's, cardiomyopathy, and respiratory impairment. (Richman 2011)
+++
IV. Medical/Surgical Management
++
Pharmacological management
Steroids
Corticosteroids and creatine monohydrate have been shown to improve strength in MD patients (Lovering, 2005; Tarnopolsky, 2004)
Long term use of corticosteroids (prednisone and deflazacort) improves outcomes: prolonged walking (independent or assisted) by up to 3 years; improved isometric muscle strength by 60% in arms and 85% in legs; improved pulmonary function; delayed start of problems from age 10 years to age 15 years; delayed onset of scoliosis; delayed start of cardiac problems for several years (Bushby, 2010b; Gloss, 2016; Kinali, 2007; Wong, 2006)
Side effects: weight gain, osteoporosis (compensatory high doses of Vitamin D and calcium), hypertension, cataracts (tiny… do not impair functional vision), decrease in height, and behavioral changes (especially in 7-9 year old age group)
In research comparing intermittent (i.e., 10 days on, 10 days off) versus daily dosing, daily dosing prolonged gait 2.5 years longer than intermittent but resulted in more side effects (Ricotti, 2013; Gloss, 2016)
Creatine – "high quality evidence" that short- and medium-term creatine increases muscle strength and functional performance in individuals with muscular dystrophy (Kley, 2013)
Tadafil – preliminary support for role of tadafil in alleviating muscle hypoxia (Nelson, 2014)
Long term Gentamicin antibiotic therapy has been tried experimentally in a subgroup of patients (about 13%) who have a nonsense mutation in the dystrophin gene leading to premature stop codon based on positive outcomes in mice (Malik, 2010). Results inconclusive to date.
Ace inhibitors and beta blockers are often prescribed for treatment of cardiomyopathies (Muntoni, 2003; Kinali, 2007)
Continuous noninvasive ventilation (NIV) and mechanically assisted coughing have been shown to prolong life and obviate tracheotomy increase survival in patients with respiratory failure (Bach, 2011; Gomez-Merino, 2002; Kinali, 2007).
Bone health a concern secondary to long term steroid use and progressive loss of weightbearing activity; requires multidisciplinary assessment and management (Bushby, 2010b; McDonald, 2002)
Surgical management focuses on control of lower extremity contractures, fracture management, use of orthoses in conjunction with surgery to prolong ambulation, and spinal stabilization for control of scoliosis; decision making informed by understanding of long term prognosis and psychosocial issues (Bushby, 2010b).
Nutritional status (Bushby, 2010b)
Gastrointestinal – constipation and gastroesophageal reflux the most common problems (Bushby, 2010b).
Gene therapy and stem cell therapy treatment continue to be advanced, but have not been proven safe for human use (Lovering, 2005; Kinali, 2007).
Bone morphogenetic protein (BMP) inhibitors are a proven treatment for muscle regeneration in mice, not yet tested in humans (Shi, 2013).
Cardiac transplantation has been used successfully (Muntoni, 2003; Kinali, 2007; Papa, 2017).
+++
V. Implications for Therapeutic Management
++
Possible preferred practice patterns from the Guide to Physical Therapist Practice: 4C: impaired muscle performance; 5a: primary prevention/risk reduction for loss of balance and falling, 5b: impaired neuromotor development; 6b: impaired aerobic capacity/endurance associated with deconditioning and 6e: impaired ventilation and respiration/gas exchange associated with ventilatory pump dysfunction or failure (APTA, 2014)
The physical therapist collaborates as a team member with other specialists including pediatric neurologists, orthopedists, genetic counselors, dieticians, occupational therapists, speech and language pathologists, orthotists, pulmonologists, respiratory therapists, cardiologists, teachers, and physical education teachers/coaches.
Child to adult progression of MD is characterized by loss of mobility, which results in increasing dependence on assistive devices to maintain independence, the need to prevent excess fatigue, and to ease caregiver responsibilities (Parker, 2005).
The Vignos Functional Rating Scale for Duchenne Muscular Dystrophy (Vignos, 1963) can be used to classify the functional level of a child with MD (Berard, 2005).
A modified 6 Minute Walk Test (6MWT) has been validated for children with DMD. Modifications include standardized instructions, a "safety chaser" that follows the participant, and standardized modes of verbal encouragement. Although validation has been obtained, further normative data for the 5 to 12 year-old age group is needed due to small cohort samples. The 5 to 12 year-old age group is significant in DMD children because this is typically the time in which the most profound progression of the disease takes place (Goemans, 2013).
Outcome measures for ambulatory boys with DMD: Vignos Lower Extremity Scale, North Star Ambulatory Assessment, Motor Function Measure, 6MWT (Bushby, 2010b).
Outcome measures for non-ambulatory boys/men with DMD: Brooke Upper Extremity Functional Scale, Egan Klassifikation Scale, Hammersmith Motor Scales, Motor Function Measure, Quantitative Myometry, Jebsen-Taylor Hand Function Test, Individualized Neuromuscular Quality of Life Questionnaire (Bushby, 2010b; Connolly, 2015; Schreiber-Bontemps, 2015).
Physical therapy treatment options for MD
Provide support for function and participation based on patient's goals. Provision of appropriate wheelchair and seating equipment, and aids and adaptations to allow maximum independence in ADL, function, and participation at various stages of disease progression. (Bushby, 2010)
Exercise
Remains controversial with lack of substantive research on optimum type, intensity and frequency of exercise (Gianola, 2013; Voet, 2010; Skura, 2005; Bushby, 2010b)
Balance needed to address progressive weakness and decreased endurance without risking muscle injury (related to underlying pathology of disorder)
High intensity resistive exercise, eccentric exercise or repeated practice of challenging physical activities (e.g., stair climbing) are thought to increase risk of contraction-induced muscle injury (Bushby, 2010b)
Through all stages of DMD, resistance and aerobic exercise can be beneficial to patients by improving their endurance and work capacity with emphasis on sub-max performance to prevent fatigue and muscle tissue damage (Lovering, 2005; Sveen, 2007; Ansved, 2007; Gianola, 2013; Voet, 2010).
In a relatively small population of boys with BMD, high and low intensity exercise programs were shown to be beneficial to increase muscle strength and endurance (Sveen, 2013).
Aquatic therapy provides opportunities for maintaining mobility and providing low-intensity exercise, especially in later stages (Lovering, 2005; Bushby, 2010b).
Assisted bicycle training (for upper and lower extremity), 15 minutes, 5 days a week for 24 weeks, resulted in better maintenance of function and ROM than in control group in a relative small number of participants (Jansen, 2013)
Prevention of joint contractures – a crucial aspect of management
Positioning with appropriate use of assistive technology
Passive manual stretching and self-stretches – at least twice a day
Ankle foot orthoses (AFO's) – useful for static control of ankle ROM, typically used during night (see below) or during day when no longer ambulating
Regular passive stretches and the use of night splints can help reduce the prevalence of joint contractures (Kinali, 2007; Eagle, 2002; Wong, 2006).
Serial casting – not recommended except in limited circumstances
Supported standing (see below)
Exercises/intervention to increase and maintain cardiorespiratory capacity and endurance.
Monitoring for progressive scoliosis
Patient and caregiver education in all stages and all settings
Referral to speech and language pathologist as appropriate for speech/language delays, dysphagia or augmentative communication (Cyrulnik, 2007; Bushby, 2010b)
Psychosocial support; social groups have been shown to increase self-development, independence, and a sense of belonging, however it many not carry forward into typical-population communities; palliative care/hospice may be beneficial to patient and caregivers in end stages (Arias, 2011; Bushby, 2010a; Parkyn, 2011).
++
+
American Physical Therapy Association. Guide to Physical Therapist Practice 3.0. Alexandria, VA: American Physical Therapy Association; 2014. Available at:
http://guidetoptpractice.apta.org/. Accessed December 29, 2017.
+
Anderson
JL, Head
SI, Rae
C, Morley
JW. Brain function in Duchenne muscular dystrophy.
Brain, Part 1. 2002;125:4–13.
[PubMed: 11834588]
+
Ansved
T. Muscular dystrophies: Influence of physical conditioning on the disease evolution.
Curr Opin Clin Nutr Metab Care. 2003;6(4):435–9.
[PubMed: 12806218]
+
Arias
R, Andrews
J, Pandya
S,
et al. Palliative care services in families of males with Duchenne muscular dystrophy.
Muscle Nerve. 2011;44(1):93–101.
[PubMed: 21674523]
+
Bach
JR, Chaudhry
SS. Standards of care in MDA clinics.
Am J Phys Med Rehabil. 2000;79:193–196,
[PubMed: 10744195]
+
Bach
JR, Martinez
D. Duchenne muscular dystrophy: continuous noninvasive ventilator support prolongs survival.
Respir Care. 2011;56(6):744–750.
[PubMed: 21333078]
+
Bakker
JPJ, de Groot
IJM, Beelen
A,
et al. Predictive factors of cessation of ambulation in patients with Duchenne muscular dystrophy.
Am J Phys Med Rehabil. 2002;81:906–912.
[PubMed: 12447089]
+
Berard
C, Payan
C, Hodgkinson
I, Fermanian
J. A motor function measure for neuromuscular diseases: construction and validation study.
Neuromuscul Disord. 2005;15(7):463–470.
[PubMed: 16106528]
+
Bushby
K. Recent advances in understanding muscular dystrophy.
Arch Dis Child. 1992; 67:1310–1312.
[PubMed: 1444538]
+
Bushby
K, Finkel
R, Birnkrant
DJ,
et al. DMD Care Considerations Working Group. Diagnosis and management of Duchenne muscular dystrophy: part 1: diagnosis, and pharmacological and psychosocial management. Lancet Neurol. 2010a;9(1):77–93.
+
Bushby
K, Finkel
R, Birnkrant
DJ,
et al.
DMD Care Considerations Working Group. Diagnosis and management of Duchenne muscular dystrophy, part 2: implementation of multidisciplinary care.
Lancet Neurol. 2010b;9(2):177–189.
[PubMed: 19945914]
+
Calvert
LD, McKeever
TM, Kinnear
WJM, Britton
JR. Trends in survival from muscular dystrophy in England and Wales, and impact on respiratory services.
Respir Med. 2006; 100;1058–1063.
[PubMed: 16257521]
+
Case
L. Physical therapy management of dystrophinopathies (Duchenne and Becker muscular dystrophy). Presentation at Annual Parent Project Conference; July 2006; Cincinnatti, OH.
+
Case
LE, Coats
J, Draper
RM, Individual with DMD. Commentary on "Supported standing in boys with Duchenne muscular dystrophy."
Pediatr Phys Ther. 2016;28(3):330–331.
[PubMed: 27341582]
+
Connolly
AM, Malkus
EC, Mendell
JR,
et al. Outcome reliability in non-ambulatory boys/men with Duchenne muscular dystrophy.
Muscle Nerve. 2015;51(4):522–532.
[PubMed: 25056178]
+
Cyrulnik
SE, Fee
RJ, De Vivo
DC, Golstein
E, Hinton
VJ. Delayed developmental language milestones in children with Duchenne's muscular dystrophy.
J Pediatr. 2007;150:474–478.
[PubMed: 17452219]
+
Darras
BT, Menache-Starobinski
CC, Hinton
V, Kunkel
LM. Dystrophinopathies. In: Darras
BT, Jones
H, Ryan
M, De Vivo
DC. Neuromuscular Disorders of Infancy, Childhood, and Adolescence: A Clinicians Approach. 2nd ed. London: Elsevier; 2015: 551–92.
+
Deconinck
N, Dan
B. Pathophysiology of Duchenne muscular dystrophy: current hypotheses.
Pediatr Neurol. 2007;36:1–7.
[PubMed: 17162189]
+
Gianola
S, Pecoranro
V, Lambiase
S, Gatti
R, Banfi
G, Moja
L. Efficacy of muscle exercise in patients with muscular dystrophy: a systematic review showing a missed opportunity to improve outcomes.
PLoS One. 2013;8(6):e65414.
[PubMed: 23894268]
+
Gloss
D, Moxley
RT, Ashwal
S, Oskoui
M. Practice guidelines update summary: corticosteroid treatment of Duchenne muscular dystrophy: report of the Guideline Development Subcommittee of the American Academy of Neurology.
Neurology. 2016;86(5):465–472.
[PubMed: 26833937]
+
Goemans
N, Klingels
K, Van den Hauwe
M,
et al. Test-retest reliability and developmental evolution of the 6-min walk test in Caucasian boys aged 5-12 years. Neuromuscur Disord. 2013;23(1):19–24.
+
Gomez-Merino
E, Bach
JR. Duchenne muscular dystrophy: prolongation of life by noninvasive ventilation and mechanically assisted coughing.
Am J Phys Med Rehabil. 2002; 81:411–415.
[PubMed: 12023596]
+
Jansen
M, van Alfen
N, Geurts
AC, de Groot. Assisted bicycle training delays functional deterioration in boys with Duchenne dystrophy: the randomized controlled trial "no use is disuse".
Neurorehabil Neural Repair. 2013;27(9):816–827.
[PubMed: 23884013]
+
Kinali
M, Main
M, Ellahoo
J,
et al. Predictive factors for the development of scoliosis in Duchenne muscular dystrophy.
Eur J Paediatr Neurol. 2007;11(3): 160–166.
[PubMed: 17257866]
+
Kley
RA, Tarnopolsky
MA, Vorgerd
M. Creatine for treating muscle disorders. Cochrane Database Syst Rev. 2013;5(6):CD004760.
+
Kohler
M, Clarenbach
C, Bahler
C,
et al. Disability and survival in Duchenne muscular dystrophy. J Neurol Neurosurg Psychiatry. 2009; 80:300–325.
+
Lovering
RM, Porter
NC, Bloch
RJ. The muscular dystrophies: from genes to therapies.
Phys Ther. 2005; 85(12):1372–1388.
[PubMed: 16305275]
+
Malik
V, Rodino-Klaac
LR, Viollet
L,
et al. Gentamicin-induced readthrough of stop codons in Duchenne muscular dystrophy.
Ann Neurol. 2010;67(6):771–780.
[PubMed: 20517938]
+
Manzur
A, Muntoni
F. Diagnosis and new treatments in muscular dystrophy.
Postgrad Med J. 2009;85:622–630.
[PubMed: 19892898]
+
McDonald
DG, Kinali
M, Gallagher
AC,
et al. Fracture prevalence in Duchenne muscular dystrophy.
Dev Med Child Neurol. 2002;44(10):695–698.
[PubMed: 12418795]
+
Muntoni
F. Cardiac complications of childhood myopathies.
J Child Neurol. 2003;18(3):191–202.
[PubMed: 12731645]
+
Nelson
MD, Rader
F, Tang
X,
et al. PDE5 inhibition alleviates functional muscle ischemia in boys with Duchenne muscular dystrophy.
Neurology. 2014;82(23):2085–2091.
[PubMed: 24808022]
+
Nowak
KJ, Davies
KE. Duchenne muscular dystrophy and dystrophin: pathogenesis and opportunities for treatment.
EMBO Reports. 2004;5(9):872–876.
[PubMed: 15470384]
+
Paganoni
S, Amato
A. Electrodiagnostic evaluation of myopathies.
Phys Med Rehabil Clin N Am. 2013;24(1):193–207.
[PubMed: 23177039]
+
Paleg
GS, Smith
BA, Glichman
LB. Systematic review and evidence-based clinical recommendations for dosing of pediatric supported standing programs.
Pediatr Phys Ther. 2013;25:232–247.
[PubMed: 23797394]
+
Papa
AA, D'Ambrosio
P, Petillo
R, Palladino
A, Politano
L. Heart transplantation in patients with dystrophinopathic cardiomyopathy: review of the literature and personal series.
Intractable Rare Dis Res. 2017;6(2):95–101.
[PubMed: 28580208]
+
Parker
AE, Robb
SA, Chambers
J,
et al. Analysis of an adult Duchenne muscular dystrophy population. Quarterly J Med. 2005;98:729–736.
+
Parkyn
H, Coveney
J. An exploration of the value of social interaction in a boys' group for adolescents with muscular dystrophy.
Child Care Health Dev. 2011;39(1):81–89.
[PubMed: 22168921]
+
Richman
S, Schub
T. Muscular Dystrophy, Duchenne's. [serial online]. September 2, 2011; Available from: CINAHL Plus with Full Text, Ipswich, MA. Accessed April 17, 2012.
+
Ricotti
V, Ridout
DA, Scott
E,
et al. NorthStar Clinical Network. Long-term benefits and adverse effects of intermittent versus daily glucocorticoids in boys with Duchenne muscular dystrophy.
J Neurol Neurosurg Psychiatry. 2013;84:698–705.
[PubMed: 23250964]
+
Rodillo
E, Fernandez-Bermejo
E, Heckmatt
J,
et al. Prevention of rapidly progressive scoliosis in Duchenne muscular dystrophy by prolongation of walking with orthoses. J Child Neurol. 1988; 269–274.
+
Schreiber-Bontemps
A, Brochard
S, Fontaine-Carbonnel
S,
et al. Promoting the use of Motor Function Measure (MFM) as outcome measure in patients with Duchenne muscular dystrophy (DMD) treated by corticosteroids. Ann Phys Rehabil Med. 2015;58(Supplement 1):e139–e140.
+
Shi
ST, de Gorter
DJ, Hoogaars
WM, 't Hoen
PA, Dijke
PT. Overactive bone morphogenetic protein signaling in heterotopic ossification and Duchenne muscular dystrophy.
Cell Mol Life Sci. 2013;70:407–423.
[PubMed: 22752156]
+
Stevens
PM. Lower limb orthotic management of Duchenne muscular dystrophy: a literature review. J Prosth Orthotics. 2006;18(4):111–119.
+
Skura
CL, Padden
TM, Fowler
EG. The effect of concentric exercise for two children with Duchenne muscular dystrophy. Peds Phys Ther. 2005;17(1):84.
+
Stuberg
W. Home accessibility and adaptive equipment in Duchenne muscular dystrophy: A case report.
Pediatr Phys Ther. 2001;13(4):169–174.
[PubMed: 17053635]
+
Sveen
ML, Jeppesen
TD, Hauerslev
S, Krag
TO, Vissing
J. Endurance training: an effective and safe treatment for patients with LGMD2I.
Neurology. 2007;68(1):59–61.
[PubMed: 17200494]
+
Sveen
ML, Andersen
SP, Ingelsrud
LH, Blichter
S,
et al. Resistance training in patients with limb–girdle and Becker muscular dystrophies.
Muscle Nerve. 2013;47:163–169.
[PubMed: 23169433]
+
Straub
V, Campbell
KP. Muscular dystrophies and the dystrophin-glycoprotein complex.
Curr Opin Neurol. 1997;10(2):168–75.
[PubMed: 9146999]
+
Tarnopolsky
MA, Mahoney
DJ, Vajsar
J,
et al. Creatine monohydrate enhances strength and body composition in Duchenne muscular dystrophy.
Neurology. 2004; 62:1771–77.
[PubMed: 15159476]
+
Townsend
E, Bibeau
C, Holmes
T. Supported standing in boys with Duchenne muscular dystrophy.
Pediatr Phys Ther. 2016;28(3):320–329.
[PubMed: 27008581]
+
Townsend
EL, Tamhane
H, Gross
KD. Effects of AFO use on walking in boys with Duchenne muscular dystrophy: a pilot study.
Pediatr Phys Ther. 2015;27(1):24–29.
[PubMed: 25401456]
+
Vignos
PJ, Spencer
GE, Archibald
KC. Management of progressive muscular dystrophy. J Am Med Assoc. 1963;184:103–112.
+
Voet
NB, van der Kooi
EL, Riphagen,
II, Lindeman
E, van Engelen
BG, Geurts
AC. Strength training and aerobic exercise training for muscle disease. Cochrane Database Syst Rev. 2010; CD003907.
+
Wicksell
RK, Kihlgren
M, Melin
L, Eeg-Ologsson
O. Specific cognitive deficits are common in children with Duchenne muscular dystrophy. Dev Med Child Neuro. 2004;46(3):154–159.
+
Zhou
GQ, Xie
HQ, Zhang
SZ, Yang
ZM. Current understanding of dystrophin-related muscular dystrophy and therapeutic challenges ahead.
Chin Med J (Engl). 2006;119(16):1381–1391.
[PubMed: 16934185]
++
A chronic, progressive, autoimmune response.
Epidemiology:
Incidence rates of 5.3 per million persons per year (Carr 2010).
Prevalence is between 14 and 20 per million in the United States (Berrih-Aknin, 2013). A prevalence rate of 77.7 cases per million (Carr 2010)
18,000 people in the US (Bershad, 2008)
In Alabama, 79% of patients were Caucasian and 21% were African American. There was a 1:1 and 1:2 male to female ratio respectively (Oh, 2009).
Myasthenia gravis is the most common primary disorder of neuromuscular transmission, and the incidence of myasthenia gravis increases with age in females and males. (Sathasivam, 2015)
Age of onset:
Women between 20 – 40, and men between 40 – 60 (Kothari, 2004)
Distribution of age of MG onset shows a female-dominant peak in early-onset myasthenia gravis (EOMG) and a male-dominant peak in late-onset myasthenia gravis (LOMG). (Cavalcante, 2013)
In recent years, the incidence of MG in the elderly (over 50 years old) population is increasing. (Cavalcante, 2013)
Pathogenesis:
Excessive production of auto-antibodies disrupts neuromuscular junction (NMJ) transmission by blocking, destroying, or altering acetylcholine receptors (AChR). (Pinto, 2007; Simpson, 1960)
Sensitized T-helper cells and immunoglobulin antibody (IgG) directed attacks on the Acetylcholine receptors at the neuromuscular junction (Kothari, 2004).
Ocular onset occurs in up to 85% of patients with IgG1-dominant antibodies to AChRs that leads to fatigable weakness (Merriggioli, 2009).
In other instances, components of the neuromuscular junction besides ACh receptors, such as the muscle-specific receptor tyrosine kinase (MuSK), can be targeted (Meriggioli, 2009).
Two main pathologies of the thymus in patients with MG:
Enlarged thymus glands, often associated with early-onset MG (Vincent, 2008).
Epithelial tumors known as thymomas that affect approximately 10% of patients with MG (Vincent, 2008).
Genetics (Berrih-Aknin, 2013):
MG families are very rare, and few studies have focused on familial or twin cases., but the association of human leukocyte antigen (HLA) class I and class II genes with MG is clearly established.
Most genes associated with MG are involved in regulating the immune system. It could therefore be proposed that subjects with specific alleles that are associated with lower immunoregulatory capacity may be more susceptible to autoimmune diseases, particularly MG.
Types:
++
++
The prognosis for patients with MG is less than 5% mortality (Juel, 2007).
Life expectancy—may be shortened due to secondary disorders. (Kothari, 2004)
Within two years of onset, 30-80% progress to generalized MG (Grob, 2008).
Disease course is usually variable and patients will experience remissions and exacerbations, especially during the first year after onset.
In general, symptoms are most severe during the first two years, and symptoms usually improve somewhat after this time period (Grob, 2007).
Approximately 20% of patients with MG will experience a myasthenic crisis typically within the first two years following their diagnosis. A myasthenic crisis is defined as the need for intubation or mechanical ventilation due to weakness of the respiratory muscles (Juel, 2004).
The frequency of a second autoimmune disease is 15% in patients with MG.
Autoimmune thyroiditis is the most common, followed by systemic lupus erythematosus (SLE) and rheumatoid arthritis.
It is important to monitor patients closely for any changes in signs/symptoms. (Gilhus, 2014)
Prognosis by Type:
Seropositive and Seronegative MG: Outcome and long-term prognosis is good with proper treatment, especially early thymectomy for seropositive MG. (Romi, 2006)
Complete stable remission can be achieved in nearly 25% of patients suffering from anti-AChR or double-negative MG. Unfortunately, treatment response is much less favorable in anti-MuSK MG, with a significantly increased risk of myasthenic crisis. (Sieb, 2014)
Thymoma MG: Less positive prognosis in thymoma MG with RyR antibodies and non-thymoma MG with titin/RyR antibodies. Patients with thymomatous MG tend to have a more severe form of the disease. (Romi, 2006)
Expected sequelae if untreated/improperly treated:
Respiratory Distress Syndrome/Respiratory Failure, known as a myasthenic crisis is due to weakening of the diaphragm and intercostals muscles and is often caused by other systemic illnesses or infections. If untreated, death within 10 years (Oosterhuis, 1989).
Other expected sequelae:
A small percentage of newborns of mothers with MG will have neonatal MG due to the ability of the maternal antibodies to cross the placenta (Vincent, 2008).
Symptoms of transient neonatal MG are a weak suck, cry, and hypotonia, which usually resolves within a few weeks.
+++
IV. Medical/Surgical Management
++
Goal: symptom-free remission with no maintenance medication (Kothari, 2004)
Aceytlcholinesterase inhibitors (first line defense):
Used to treat the symptom of muscle weakness but not the underlying autoimmune attack (Juel, 2007); provide temporary relief of symptoms but do not prevent progression of disease (Guptill 2016).
Drug of choice is pyridostigmine. (Gilhus, 2015; Kumar 2011; Guptill, 2016)
Immunosuppressants (i.e. corticosteroids):
Decreases immune response. The best results are seen when patients take an initial high dosage then gradually taper off as improvements in strength are noted. Many adverse side affects are associated with corticosteroids (Juel, 2007).
The side effects of immunosupressants lead to widespread metabolic repercussions that may increase medical co-morbidities and decrease quality of life (Conti-Fine, 2006)
Plasmapheresis:
For short-term management, removes circulating immune complexes with improvement lasting 6-8 weeks (Kothari, 2004; Massey, 1997)
Intravenous immunoglobulins (IgG):
Thymectomy:
In patients < 60 y.o., symptomatic relief from decrease in T-cell reactivity against disease-specific antigens. (Kothari, 2004 and Ahlberg, 1997)
Thymectomy is effective for early-onset MG if done shortly after symptoms present. It also can be safe and effective for children with MG down to 5 years of age. This procedure is not recommended for patients with MUSK, LRP4, or ocular types of MG because no therapeutic effect has been shown. (Gilhus, 2015)
Non-invasive ventilation (NIV) can be used in cases of myasthenic crisis to potentially avoid intubation.
Lower Acute Physiology and Chronic Health Evaluation (APACHE II) scores, lower PaCO2 values, and lower serum bicarbonate values correlated with NIV success (Wu, 2009).
Myasthenic crisis is the life-threatening exacerbation of MG due to weakness of respiratory muscles and swallowing difficulties. (Sieb, 2014)
Additional treatment specifically for ocular MG can include occlusive devices, prisms, eyelid supports, contact lenses, and strabismus surgery or eyelid elevation surgery (Haines, 2012).
+++
V. Implications for Therapeutic Management
++
Physical Therapy Preferred Practice Pattern (APTA, 2017):
There is limited research regarding exercise-related therapy and the management of MG.
One review found that generally patients believe activity is essential for them to improve symptoms and quality of life. Activity has also been shown to help reduce fatigue. There are currently no guidelines for determining when a patient with MG can participate in activity. Close monitoring and thorough assessment must be done before a patient resumes physical activity or sport. (Cass, 2013)
Aerobic activity at mild-mod levels on a regular basis helps increase functional status and may become a coping strategy for stress. (Cass, 2013) and maintain the individual identity (Grohar-Murray, 1998)
Breathing exercises are effective for reducing fatigability. (Cass, 2013)
Low impact aerobic exercise balanced with rest: swimming, walking, and running to increase endurance and reduce fatigue.
Low impact aerobic and anaerobic activities increase patient confidence (Leddy and Chutkow, 2000)
Lower limb strengthening, but not high reps, may benefit moderate MG or neuromuscular diseases
A balance exercise program increases functional ability and balance (Naumes, 2016)
Supervised physical training increases maximal muscle force and endurance (Naumes, 2016)
Inspiratory muscle training and diaphragmatic breathing therapy (Cup, 2007;Davidson, 2005)
A partial home treatment program consisting of interval-based inspiratory muscle training in combination with breathing retraining has shown to be effective for improvements in respiratory muscle strength, chest wall mobility, respiratory pattern, and respiratory endurance (Fregonezi et al, 2005).
Patients should have a customized exercise program to improve individual overall health and address concerns due to safety standards (Cass, 2013)
The PT should be aware of the side effects of medications when planning treatment sessions. (Gilhus, 2014)
Anticholinergics: side effects on the autonomic system, most commonly diarrhea and abdominal pain.
Corticosteroids: osteoporosis, weight gain, hypertension, glucose intolerance
Dietary referral for nutrition education (Pinto, 2007)
A patient with MG is able to exercise even competitively, but it is important to always monitor fatigue and to collaborate with the healthcare team to optimize patient safety.
In accordance with the ICF model, patients are limited in ADLs due to impairments of body function, but improvements in function can occur if they are provided facilitators in environmental factors (Leonardi, 2009).
Contraindications/precautions:
When eating, the patient should never speak with food in the mouth or swallow with the neck extended due to the risk of aspiration.
Avoid overwork weakness
High humidity and temperature will exacerbate fatigue
PT should be aware for signs of a myasthenic crisis.
Signs include strenuous breathing, increased muscle weakness, and increased difficulty with swallowing, chewing and speaking.
Patient education:
Decreasing stress (Pinto, 2007)
Effect of high humidity and temperature, increased activity levels, and infections on fatigue.
Mental and physical fatigue management strategies and rest should be used to reduce fatigue and increase quality of life (Grohar-Murrey, 1998).
Adequate pacing for activities and avoiding prolonged exercise (Naumes, 2016)
Health promotion and prevention measures:
+++
VI. Consumer and Professional Resources
++
Myasthenia Gravis Foundation of America, Inc
MDJunction
American Autoimmune Related Disease Association
National Institute of Neurological Disorders and Stroke
Muscular Dystrophy Association
+
Ahlberg
R, Pirskanen
R,
et al. The effect of thymectomy on autoreactive T-and B-lymphocytes in myasthenia gravis.
Journal of Neuroimmunology. 1997; 74:45–54
[PubMed: 9119978]
+
Armstrong
SM, Schumann
L. Myasthenia gravis: diagnosis and treatment.
Journal of the American Academy of Nurse Practitioners. 2003; 15:72–78
[PubMed: 12640942]
+
Barrons
RW. Drug-induced neuromuscular blockade and myasthenia gravis.
Pharmacotherapy. 1997; 17(6):1220–1232
[PubMed: 9399604]
+
Benatar
M. A systematic review of diagnostic studies in myasthenia gravis.
Neuromuscul Disord. 2006;16(7):459–467.
[PubMed: 16793269]
+
Berrih-Aknin
S, Le Panse
R. Myasthenia gravis: a comprehensive review of immune dysregulation and etiological mechanisms.
J Autoimmun. 2014; 52:90–100.
[PubMed: 24389034]
+
Bershad
EM, Feen
ES, Suarez
JI. Myasthenia gravis crisis.
Southern Medical Journal. 2008; 101 (1): 63–69
[PubMed: 18176295]
+
Carr
AS, Cardwell
CR, McCarron
PO, McConville
J. A systematic review of population based epidemiological studies in Myasthenia Gravis. BioMed Central Neurology. 2010; 10:46. doi: 10.1186/1471-2377-10-46.
+
Cass
S. Myasthenia gravis and sports participation.
Curr Sports Med Rep. 2013;12(1): 18–21.
[PubMed: 23314079]
+
Cavalcante
P, Cufi
P, Mantegazza
R, Berrih-Aknin
S, Bernasconi
P, Le Panse
R. Etiology of myasthenia gravis: Innate immunity signature in pathological thymus.
Autoimmun Rev 2013;12(9):863–874.
[PubMed: 23535157]
+
Ceremuga
TE, Yao
XL, McCabe
JT. Etiology, mechanisms, and anesthesia implications of autoimmune myasthenia gravis. AANA journal. 2002; 70:301–310
+
Chiu
H-C, Vincent
A, Newsom-Davis
J, Hsieh
K-H, Hung
T-P. Myasthenia gravis: population differences in disease expression and acetylcholine receptor antibody titers between chinese and caucasians.
Neurology 1987; 37: 1854–57
[PubMed: 3484357]
+
Conti-Fine
B, Kaminski
H, Milani
M. Myasenthia gravis: past, present, and future.
Journal of Clinical Investigation 2006; 116: 2843–2854.
[PubMed: 17080188]
+
Cosi
V, Longardi
M, Piccolo
G, Erbetta
A. Treatment of myasthenia gravis with high-dose intravenous immunoglobulin.
Acta Neurologica Scandinavica. 1991; 84:81–84
[PubMed: 1950455]
+
Cup
EH,
et al. Exercise therapy and other types of physical therapy for patients with neuromuscular diseases: a systematic review.
Arch Phys Med Rehabil, 2007;88 (11):1452–64.
[PubMed: 17964887]
+
Davidson
L, Hale
L, Mulligan
H. Exercise prescription in the physiotherapeutic management of myasthenia gravis: a case report. New Zealand Journal of Physiotherapy. 2005; 33:13–18
+
Elsais
A, Wyller
VB, Loge
JH, Kerty
E. Fatigue in myasthenia gravis: Is it more than muscular weakness? BioMed Central Neurology. 2013; 13: 132. doi: 10.1186/1471-2377-13-132.
+
Fakiri
MO, Tavy
DLJ, Hama-Amin
A, Wirtz
P. Accuracy of the ice test in the diagnosis of myasthenia gravis in patients with ptosis. Muscle & Nerve. December 2013; 48(6): 902–904.
+
Fregonezi
GA de F, Resqueti
VR, Güell
R, Pradas
J, Casan
P. Effects of 8-Week, Interval-Based Inspiratory Muscle Training and Breathing Retraining in Patients With Generalized Myasthenia Gravis.
Chest. 2005;128(3):1524–1530.
[PubMed: 16162753]
+
Gilhus
NE, Nacu
A, Andersen
JB, Owe
JF. Myasthenia gravis and risks for comorbidity.
Eur J Neurol. 2014; 22(1): 17–23.
[PubMed: 25354676]
+
Gilhus
NE, Verschuuren
JJ. Myasthenia gravis: subgroup classification and therapeutic strategies.
Lancet Neurol. 2015;14(10):1023–1036.
[PubMed: 26376969]
+
Grob
D. Natural history of myasthenia gravis. In: Engel
AG. Myasthenia gravis and myasthenic disorders. Oxford: Oxford University Press. Contemporary Neurology Series; 1999:131–45
+
Grob
D, Brunner
N, Namba
T, Pagala
M. Lifetime course of myasthenia gravis. Muscle & Nerve. February 2008;37(2):141–149.
+
Grohar-Murray
ME, Becker
A, Reilly
S, Ricci
M. Self-care actions to manage fatigue among myasthenia gravis patients.
J Neurosci Nurs. 1998;30(3):191–199.
[PubMed: 9689611]
+
Guptill
JT, Soni
M, Meriggioli
MN. Current Treatment, Emerging Translational Therapies, and New Therapeutic Targets for Autoimmune Myasthenia Gravis.
NeuroTherapeutics. 2016
;13:118–131.
[PubMed: 26510558]
+
Haines
SR, Thurtell
MJ. Treatment of ocular myasthenia gravis.
Curr Treat Options Neurol. 2012;14(1):103–112.
[PubMed: 22037997]
+
Juel
VC, Massey
JM. Myasthenia gravis.
Orphanet J Rare Dis. 2007;2:44.
[PubMed: 17986328]
+
Juel
VC. Myasthenia gravis: Management of Myasthenic Crisis and Perioperative Care.
Seminars in Neurology. 2004;24(1):75–81.
[PubMed: 15229794]
+
Kothari,
M. Myasthenia gravis. Journal of American Osteopathic Association. 1994; 104: 377–384.
+
Kumar
V, Kaminski
HJ. Treatment of myasthenia gravis.
Curr Neurol Neurosci Rep. 2011
;11(1):89–96.
[PubMed: 20927659]
+
Leddy
JJ, Chutkow
JG. Myasthenia gravis in a collegiate football player.
Med Sci Sports Exerc. 2000
;32:1975–1979.
[PubMed: 11128838]
+
Leonardi
M, Raggi
A, Antozzi
C, Confalonieri, Maggi
L, Cornelio
F, Mantegazza
R. Disability and functional profiles of patients with myasthenia gravis measured with ICF classification. Internation Journal of Rehabilitation Research. June 2009; 32(2): 167–172
+
Massy
JM. Acquired myasthenia gravis.
Neurol Clin. 1997; 15:577–559
[PubMed: 9227954]
+
Meriggioli
MN, Sanders
DB. Autoimmune myasthenia gravis: emerging clinical and biological heterogeneity.
Lancet Neurol. 2009;8(5):475–490.
[PubMed: 19375665]
+
Naumes
J, Hafer-Macko
C, Foidel
S. Exercise and Myasthenia Gravis: A Review of the Literature to Promote Safety, Engagement, and Functioning. Int J Neurorehabil. 2016;3(218):2376–0281.
+
Nicole
MW. Intravenous immunoglobulin improved clinical outcomes in patients with myasthenia gravis and worsening weakness.
ACP J Club. 2007; 147(2): 50
[PubMed: 17764141]
+
Oh
S, Morgan
M, Lu
L, Hatanaka
Y, Hemmi
S, Young
A, Claussen
G, Racial differences in myasthenia gravis in Alabama. Muscle & Nerve. March 2009; 39(3): 328–332.
+
Oosterhuis
HJGH. The natural course of myasthenia gravis: a long term follow up study.
J Neurol Neurosurg Psychiatry 1989; 52:1121–27
[PubMed: 2795037]
+
Penn
AS, Low
BW, Jaffe
IA, Luo
L, Jacques
JJ. Drug-induced autoimmune myasthenia gravis.
Ann. N. Y. Acad. Sci. 1998;841:433–449.
[PubMed: 9668273]
+
Pinto,
S. Myasthenia gravis. Clinical Information Systems. 2008, Nov 11.
+
Romi
F, Gillhus
NE, Aarli
JA. Myasthenia gravis: disease severity and prognosis.
Acta Neurologica Scandinavica. 2006; 183:24–25
[PubMed: 16637923]
+
Sathasivam,
S. Diagnosis and management of myasthenia gravis. Prog Neurol Psychiatry. 2014;18(1): 6–14.
+
Scheer
BV, Valero-Burgos
E, Costa
R. Myasthenia gravis and endurance exercise.
Am J Phys Med Rehabil. 2012;91(8):725–727.
[PubMed: 22311063]
+
Sieb
JP. Myasthenia gravis: an update for the clinician.
Clin Exp Immunol. 2014;175(3):408–418.
[PubMed: 24117026]
+
Simpson
JA. Myasthenia gravis: A new hypothesis. Scott Med J. 1960; 5: 419–39
+
Sitek
E, Bilinska
M, Wieczorek
D, Nyka
W. Neuropsychological assessment in myasthenia gravis. Neurological Sciences. February 2009; 30(1): 9–14.
+
Thanvi
BR, Lo
TCN. Update on myasthenia gravis.
Postgrad Med J. 2004;80(950):690–700.
[PubMed: 15579606]
+
Turner,
C. A review of myasthenia gravis: Pathogenesis, clinical features and treatment. Current Anaesthesia and Critical Care. 2007; 18(1): 15–23
+
Vincent
A, Palace
J, Hilton-Jones
D. Myasthenia gravis.
Lancet. 2001; 357: 2122–2128
[PubMed: 11445126]
+
Vincent
A. Autoimmune disorders of the neuromuscular junction.
Neurology India. 2008;56(3):305.
[PubMed: 18974557]
+
Wong
SH, Nitz
JC, Williams
K, Brauer
SG. Effects of balance strategy training in myasthenia gravis: A case study series.
Muscle Nerve. 2014
;49:654–660.
[PubMed: 23966018]
+
Wong
SH, Petrie
A, Plant,
GT. Ocular Myasthenia Gravis: Toward a Risk of Generalization Score and Sample Size Calculation for a Randomized Controlled Trial of Disease Modification:
J Neuroophthalmol. 2016;36:252–258.
[PubMed: 27031125]
+
Wu
J, Kuo
P, Fan
P, Wu
H, Shih
F, Yang
P. The Role of Non-invasive Ventilation and Factors Predicting Extubation Outcome in Myasthenic Crisis. Neurocritical Care. February 2009; 10(1): 35–42.
+++
Myelomeningocele / Spina Bifida
++
++
"Spina bifida (SB) is a neural tube defect (NTD) that occurs in the first month after conception [about 28 days gestation] and involves a defect of variable severity in the developing spinal cord. The term SB is an umbrella term and includes different types of spinal cord defects, of which myelomeningocele is the most frequent and the most involved" (Thibideau, 2017); can occur anywhere along the spine if neural tube does not close all the way; severity depends on size and location of the opening in the spine and whether the spinal cord and nerves are affected (National Center for Birth Defects and Developmental Disabilities, 2016).
Myelodysplasia is an alternate umbrella term: anomalies of the spinal cord; abnormalities of the spinal column or cranium that arise as a result of an embryologic developmental failure (i.e., neural tube closure).
Myelomeningocele (MMC) will be the focus of this diagnostic outline
Cause of NTD is unknown; however, non-genetic (environmental) and genetic factors are thought to influence
Environmental: decreased folate intake (most common), exposure to teratogens, maternal hyperthermia early in pregnancy, maternal use of seizure medications, pre-gestational diabetes, and obesity are thought to have a role (Copp, 2015).
Genetic: elevated for Irish and Celts (4.5 per 1000) and low among Japanese (0.3 per 1000); families with 1 child with spina bifida have 3 times greater chance of having second child born with defect (Apkon, 2014).
++
++
Spina bifida aperta (cystica) (SBA): exposed neural tissue is visible at birth and may or may not have a protruding sac at site of lesion (Mohd-Zin, 2017).
Anencephaly: lethal condition; vault of skull usually missing; exposed mass of undifferentiated vascular and dyplastic neural tissue
Encephalocele: most often lethal condition; protrusion of brain tissue in a sac typically in occipital area of skull
Myelomeningocele: spinal cord protrudes from the spinal canal into a sac; defect in lumbosacral area in 85% of cases; paralysis and loss of sensation below level of defect; may be boney abnormalities
Spina bifida occulta (SBO) or closed neural tube defect (NTD): non-fusion of vertebral arches; incidence 4.5% of general population and 21-25% of parents of children with spina bifida (Mohd-Zin, 2017)
Closed NTD with subcutaneous mass:
Lipomyelomeningocele involves a fatty subcutaneous mass that passes through the open bifid vertebrae and attaches to the spinal cord; children usually present with varying degrees of lower extremity paralysis, decreased sensation and neurogenic bowel and bladder; seldom associated with Chiari II malformation or hydrocephalus (Ross, 2007).
Meningocele – skin-covered sac filled with cerebral spinal fluid (CSF) that herniates through vertebral defect with meninges; occurs most often in lumbar or sacral area; neurological exam in neonate is usually normal but neurologic problems may develop with growth; hydrocephalus may be associated but rarely Chiari II malformation (Ross, 2007).
Closed NTD without subcutaneous mass; simple and complex dysraphic sites (Ross, 2007)
Incidence of SB is 1-10 per 1000 live births worldwide and .2 per 1000 births in the USA (Au, 2010).
Decline in the incidence of SB secondary to: increased prenatal screening, improved nutrition for pregnant women and folic acid supplementation.
US Department of Health and Human Services required addition of folic acid to all enriched cereal grain products in 1988 (decreased incidence by 26-31%)
Folic acid supplementation thought to prevent 50-70% of neural tube defects; recommendation 400ug prior to conception; 600ug during first trimester
++
++
Infant at birth (Danielsson, 2008; Ozaras, 2015):
Presence of a sac or skin lesion
Muscle weakness or paralysis below lesion; decreased deep tendon reflexes
Sensory impairment below lesion
Possibility of musculoskeletal deformities from lack of movement in utero: for example, hip dislocation (unilateral or bilateral), foot deformities (club foot most common)
Kyphosis, scoliosis (present in 15-25% of newborns)
Urinary/bowel dysfunction
Cranial ultrasonographic findings: The lemon sign (overlapping of the frontal bones); small cerebellum (transcerebellar diameter <10th percentile); effacement of the cisterna magna (width <2 mm on axial scan of posterior fossa); banana sign (small cerebellum hemispheres curling anteriorly and obliteration of the cisterna magna as a result of downward displacement of the hindbrain structures); ventriculomegaly (atrial width: severe: >14 mm; borderline: >10 mm); funneling of the posterior fossa (clivus-supra-occiput angle <72 degrees) (D'Addario, 2008).
Hydrocephalus – will develop in 80-90% of infants; about 25% have evidence at birth; an additional 55% will develop symptoms within the first several days; remaining 10% will need a shunt placed within first 6 months.
Arnold Chiari II malformation – primary cause of hydrocephalus
Pathology: small, flattened posterior fossa; downward displacement of cerebellum through foramen magnum; brainstem structures displaced in caudal direction with compression of medulla; elongated 4th ventricle with obstruction of CSF flow from 4th ventricle; and may get traction of cranial nerves
About 2-3% of children with spina bifida show significant effects on brainstem and cranial nerve function: stridor (especially with inspiration), apnea (when crying or at night), gastroesophageal reflux, paralysis of vocal cords, swallowing difficulty, bronchial aspiration
Some will outgrow; some require surgery (usually posterior fossa decompression and cervical laminectomy)
Hydrocephalus and Arnold Chiari II malformation are thought to be primary cause of global neurologic problems (e.g., generally low muscle tone, mild fine motor delays and characteristic learning disabilities)
++
++
Prenatal screening during pregnancy: measurement of maternal serum alpha-fetoprotein is part of the triple test (alpha -fetoprotein, human chorionic gonadotropin, and estradiol) and the quad screen (alpha -fetoprotein, human chorionic gonadotropin, estradiol, inhibin A) routinely done during pregnancy. Confirmation can be done via amniocentesis which is used to measure for elevated alpha-fetoprotein level in the amniotic fluid (Apkon, 2014).
Elevated alpha-fetoprotein will detect about 89% of neural tube defects; false negatives and positives
Prenatal ultrasound of the fetus at 18-20 weeks gestation (Phillips, 2017).
Cranial ultrasonography, electromyography, magnetic resonance imaging (MRI)
+++
III. Prognosis & Sequelae
++
++
MMC is non-progressive after surgical repair, however, musculoskeletal deformity, hydrocephalus, potential tethered cord syndrome and other potential secondary problems can cause neurological compromise (Phillips, 2017).
75% of children with MMC can expect to live into their early adult years, 85% will attend or graduate from high school and/or college (Bowman, 2001).
90% survival with aggressive treatment (i.e., surgical closure, antibiotics, clean intermittent catheterization, and shunting)
Quality of life (QOL) – self-perceived QOL not influenced primarily by medical issues; more strongly associated with individual's beliefs and experiences (influenced by family); many adults lack independence that is influenced by learning issues, negative attitudes and societal limits (Sawin, 2010)
++
Expected sequelae – at least somewhat variable based on level of lesion; risks include:
++
Neurogenic bladder - 80% of adults have social bladder incontinence; urinary tract infections; 6-11 times more likely to have renal failure
Bowel incontinence
Latex allergies - presumably from increased exposure to latex; typically mild allergic response (e.g., skin rashes) but may be more serious; important to be familiar with many everyday products that contain latex
Tethered cord
In typically developing children the spinal cord ascends within the boney spinal canal (at bottom of L3 at birth and at top of L2 by adolescence)
In children with myelodysplasia, the cord tends to get stuck in lower position; may get tethered to scar tissue or bony abnormalities; leads to stretching of cord and its blood vessels; may damage cord; and may develop hydromyelia (fluid cyst in the cord), cord lipomas or other problems
Typically with lesions above L3 will see symptoms of tethered cord before age 6 years and with lesions below L3 will see symptoms after 6 years.
Signs or symptoms may include: progressive deterioration of gait, loss of strength, back or leg pain, hypertonus, rapid progression in scoliosis, progressive foot deformity, deterioration in urologic function
Hydromyelia – excess CSF collects in pockets along spinal cord; creates areas of pressure and necrosisis of surrounding peripheral nerves
Scoliosis – causes: asymmetric muscle function, unilateral dislocated hip, paralytic scolios (80% of those with thoracic level lesion will develop; 50% at mid-lumbar level; and 20% at low lumbar level; typically gradual onset from 5-9 years of age; 49% develop scoliosis, 43% will require spinal fusion
Lordosis (often as a compensatory strategy for stability when standing/walking or from unopposed pull of iliopsoas)
Hydrocephalus – often treated routinely in neonatal period because prevalence so high; signs and symptoms of shunt malfunction may include bulging fontanelle, vomiting, change in appetite, "sunset sign" of eyes, edema or redness along shunt tract, headaches, irritability, lethargy, or seizures
Seizure disorder – 20-25% of children; increased incidence with repeated shunt infections; can usually be managed with medication(s)
Muscle imbalance around joints leads to contractures
Possible boney deformities: equinovarus, calcaneovalgus, hip subluxation/dislocation, scoliosis (above), lordosis
Impaired sensation at and below the level of lesion increases the risk of skin damage due to temperature changes, burns, accidents, pressure, and delayed wound healing (Phillips, 2017); children who had their first incidence of skin breakdown during adolescence had higher mortality than those with the first incidence during young adulthood (Cai, 2016)
Overweight/obese - relative lack of activity places at risk for abnormal weight gain that can decrease functional abilities; 50% overweight by 6 years of age.
Up to 85% have low muscle tone and developmental delay (Wolf, 1992)
Sexual dysfunction
Cognition and perception – normal distribution curve of average IQ similar to that of children without spinal bifida but shifted to the left (i.e., about 20-point lower IQ); may be cognitive and visual perceptual deficits especially in children who have shunts, delayed recall of recently acquired words and slowed acquisition of new words
Areas of relative strength: social cognition, phonological processing, long-term memory, expressive language, reading, spelling (Sandler, 1997)
Areas of relative weakness: attention/organizational deficits, abstract reasoning, visual-perceptual abilities, eye-hand coordination, visual-motor integration, writing, mathematics (Sandler, 1997)
Functional mobility and adaptive equipment needs (Apkon, 2014; Phillips, 2017)
+++
IV. Medical/Surgical Management
++
++
Intravesical botulinum toxin injections
Clean intermittent catheterization and anticholinergic medications for neurogenic bladder (Apkon, 2014)
Seizure medications
Medications for constipation
++
++
Neonatal surgery to close neural tube within 24-48 hours of birth; multi-layer repair (Northrup, 2000; Apkon, 2014).
In utero surgical repair of defect in fetus:
Became an option in 1990's; performed between 20-25 weeks gestation; survival rate 94% (Adzick, 2011).
Outcomes: significant reversal of hindbrain herniation; decreased need for shunting; improvement in lower extremity function; decreased physical and chemical damage of the exposed neurological tissue of the open lesion (Adzick, 2011)
Management of Myelomeningocele (MOM's) Study funded by National Institutes of Health (2003-2010)
Neurosurgical management:
Closure of MMC
Treatment of hydrocephalus, management of shunt malfunctions – typically placement of ventriculo-peritoneal shunt
Symptomatic Chiari II malformations
Tethered spinal cord if symptoms interfere with function or are progressive, may require surgery (microdissection)
Syringo-hydromelia
TOMAX-procedure to restore unilateral genital sensation in men for improved sexual health (Apkon, 2014; Overgoor, 2015)
Surgical management of neurogenic bladder: vesicostomy, refluxing ureterostomy, Mitrofanoff procedure.
Surgical management of neurogenic bowel: Chait tube, ACE (antegrade colonic enema) or Malone ACE, which involves a small opening (ostomy) for daily saline flush via catheter insertion.
Musculoskeletal surgeries: to improve ambulation, achieve better alignment for function and to protect skin (Apkon, 2014)
+++
V. Key Aspects of therapeutic management
++
Possible Preferred Practice Pattern: Pattern 5C: Impaired Motor Function and Sensory Integrity Associated with Non-Progressive Disorders of the Central Nervous System— Congenital Origin or Acquired in Infancy or Childhood (APTA, 2014)
A comprehensive team of providers who are specialists in orthopedics, urology, developmental pediatrics, rehabilitation medicine, orthoses management, physical, occupational, and speech therapy is optimal for the functional management of children with MMC (Ryan, 1991; Apkon, 2014).
The focus of therapy evolves throughout the individual's life span based on individual needs with an emphasis on function, independence, and self-esteem (Ryan, 1991). Independence with mobility strongly correlates with quality of life (Schoenmakers, 2005).
Patient and family education on precautions relative to individual sequelae including risk of skin breakdown and pressure sores, shunt malfunction, signs of tethered cord and Chiari malformation, bowel/bladder management, prevention of musculoskeletal deformity, latex allergies, and seizure management (Apkon, 2014).
Strength training: train active muscle groups 2-3 times per week, 1-3 sets of 8-12 repetitions using free weights, machines, or isokinetic exercise machines (O'Connell, 1992; O'Connell, 1995).
Interventions for gait training (Northrup, 2000; Brown, 2001; Apkon, 2014; Funderburg, 2017):
Orthoses/assistive devices: consideration should include energy efficiency, various gait parameters, ease of donning/doffing, transfers, need for assistive devices, home/school/work environment, ability to accommodate musculoskeletal problems (e.g., hip flexion contractures or scoliosis), and motivation of patient/family (Funderburg, 2017).
Solid AFO's recommended for children with lower lumbar and sacral level lesions; forearm crutches when more support needed; stresses on knee possible complication (Funderburg, 2017)
For thoracic or lumbar level lesions:
Swivel walker compared to parapodium: more freedom of upper extremities, less energy consumption, enhanced distance efficiency, and general preference by patients and families; but slower walking velocity (Funderburg, 2017)
Conflicting evidence whether RGO's or HKAFO's promoted faster, more energy-efficient gait (Funderburg, 2017)
Transition to wheelchair use typical; continuing ambulation as long as possible recommended
Electrical stimulation studied in children with spinal cord injury (mostly incomplete lesions) but limited research with children who have spina bifida (Funderburg, 2017).
Treadmill training program using 12-step protocol shown to increase walking speed, total distance, endurance and potential to use less restrictive assistive devices (deGroot, 2011); infant stepping on treadmill with sensory enhancement on treadmill done with limited number of subjects with spina bifida at lumbar or sacral levels may decrease the time to initiation of walking (Pantall, 2011; Moerchen, 2013; Christensen, 2014); and, based on a single subject case study, possibly increase bone mineral content (Lee, 2016).
Wheelchair
Habilitation goals for children with MMC are to promote overall wellness and encourage self- care management, self-care advocacy, independence, and mobility across all areas of participation in their homes, schools, and communities (Swanson, 2010).
Outcome measures (few with specific reliability and validity confirmed for children with spina bifida:
Shuttle Ride Test for aerobic fitness (Bloeman, 2017)
Pediatric Evaluation of Disability Inventory (Haley, 1992)
Functional Independence Measure for Children II (WeeFIM) (Uniform Data System for Medical Rehabilitation, 2009)
Trail Making Test B (Tombaugh, 2004)
Vineland Adaptive Behavior Scale-II
Pediatric Quality of Life Inventory
Pediatric Outcomes Data Collection Instrument (Damiano, 2005).
+
Adzick
NS, Thom
EA, Spong
CY,
et al. A randomized trial of prenatal versus postnatal repair of myelomeningocele. New Eng J Med. 2011;364(11):993–1004.
+
American Physical Therapy Association.
Guide to Physical Therapist Practice 3.0. Alexandria, VA: American Physical Therapy Association; 2014. Available at:
http://guidetoptpractice.apta.org/. Accessed December 29, 2017.
+
Apkon
SD, Grady
R, Hart
S,
et al. Advances in the care of children with spina bifida.
Adv Pediatr. 2014;61(1):33–74.
[PubMed: 25037124]
+
Au
KS, Ashley-Koch
A, Northrup
H. Epidemiologic and genetic aspects of spina bifida and other neural tube defects.
Dev Disabil Res Rev. 2010;16(1):6–15.
[PubMed: 20419766]
+
Bloemen
MAT, de Groot
JF, Backx
FJG, Benner
J, Kruitwagen
C, Takken
T. Wheelchair Shuttle Test for assessing aerobic fitness in youth with spina bifida: validity and reliability.
Phys Ther. 2017;97(10):1020–1029.
[PubMed: 29029556]
+
Bowman
EM, McLone
DG, Grant
JA, Tomita
T, Ito
JA. Spina bifida outcomes: a 25-year perspective.
Pediatr Neurosurg. 2001;34:114–120.
[PubMed: 11359098]
+
Brown
JP. Orthopaedic care of children with spina bifida: you've come a long way, baby!
Orthop Nurs. 2001;20(4):51–58.
[PubMed: 12025673]
+
Cai
B, McDermott
S, Wang
Y,
et al. Skin ulcers and mortality among adolescents and young adults with spina bifida in South Carolina during 2000-2010.
J Child Neurol. 2016;31(3):370–377.
[PubMed: 26239488]
+
Christensen
C, Lowes
LP. Treadmill training for a child with spina bifida without functional ambulation.
Pediatr Phys Ther. 2014;26(2):265–273.
[PubMed: 24675133]
+
Copp
AJ, Adzick
NS, Chitty
LS, Fletcher
JM, Holmbeck
GN, Shaw
GM. Spina bifida. Nat Rev Dis Primers. 2015;Apr 30;1:15007.
+
D'Addario
V, Rossi
AC, Pinto
V,
et al. Comparison of six sonographic signs in the prenatal diagnosis of spina bifida.
J Perinat Med. 2008;36(4):330–4.
[PubMed: 18598123]
+
Damiano
DL, Gilgannon
MD, Abel
MF. Responsiveness and uniqueness of the Pediatric Outcomes Data Collection Instrument compared to the Gross Motor Function Measure for measuring orthopaedic and neurosurgical outcomes in cerebral palsy.
J Pediatr Orthop. 2005;25:641–645.
[PubMed: 16199947]
+
Danielsson
AJ, Bartonek
A, Levey
E, McHale
K, Sponseller
P, Saraste
H. Associations between orthopaedic findings, ambulation and health-related quality of life in children with myelomeningocele.
J Child Orthop. 2008;2(1):45–54.
[PubMed: 19308602]
+
deGroot
JF, Takken
T, van Brussel
M,
et al. Randomized controlled study of home-based treadmill training for ambulatory children with spina bifida.
Neurorehabil Neural Repair. 2011;25(7):597–606.
[PubMed: 21415263]
+
Funderburg
SE, Josephson
HE, Price
AA, Russo
MA, Case
LE. Interventions for gait training in children with spinal cord impairments: a scoping review.
Pediatr Phys Ther. 2017;29(4):342–349.
[PubMed: 28953180]
+
Haley
SM, Coster
WJ, Ludlow
L, Haltiwanger
JT, Andrellos
PJ. The Pediatric Evaluation of Disability Inventory: Development Standardization and Administration Manual. Boston, MA: New England Medical Center Publications; 1992.
+
Lee
DK, Muraszko
K, Ulrich
BD. Bone mineral content in infants with myelomeningocele, with and without treadmill stepping practice.
Pediatr Phys Ther. 2016;28(1):24–32.
[PubMed: 27088680]
+
Moerchen
VA, Hodfakker
HL. Infants with spina bifida: immediate responses to contextual and manual sensory augmentation during treadmill stepping.
Pediatr Phys Ther. 2013;25(1):36–45.
[PubMed: 23288007]
+
Mohd-Zin
SW, Marwan
AI, Abou
Chaar MK, Ahmad-Annuar
A, Abdul-Aziz
NM. Review article: Spina bifida: pathogenesis, mechanisms, and genes in mice and humans.
Scientifica (Cairo). 2017;2017:Article ID 5364827. Available at:
https://www.hindawi.com/journals/scientifica/2017/5364827/. Accessed January 7, 2018.
+
National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention.
Spina Bifida. Available at:
https://www.cdc.gov/ncbddd/spinabifida/facts.html. Last updated October 16, 2016. Accessed January 7, 2018.
+
Northrup
H, Volcik
KA. Spina bifida and other neural tube defects.
Curr Probl Pediatr. 2000;30(10):313–332.
[PubMed: 11147289]
+
O'Connell
D, Barnhart
R. Improvement in wheelchair propulsion in pediatric wheelchair users through resistance training: a pilot study.
Arch Phys Med Rehabil. 1995;76:368–372.
[PubMed: 7717838]
+
O'Connell
D, Barnhart
R, Parks
L. Muscular endurance and wheelchair propulsion in children with cerebral palsy or myelomeningocele.
Arch Phys Med Rehabil. 1992;73(8):709–711.
[PubMed: 1642518]
+
Oeffinger
D, Bagley
A, Rogers
S,
et al. Outcome tools used for ambulatory children with cerebral palsy: responsiveness and minimum clinically important differences.
Dev Med Child Neurol. 2008;50(12):918–25.
[PubMed: 19046185]
+
Overgoor
ML, Braakhekke
JP, Kon
M, DeJong
TP. Restoring penis sensation in patients with low spinal cord lesions: the role of the remaining function of the dorsal nerve in a unilateral or bilateral TOMAX procedure.
Neurourol Urodyn. 2015;34(4):343–348.
[PubMed: 24481885]
+
Ozaras,
N. Spina bifida and rehabilitation. Turk J Phys Med Rehab. 2015;61:65–9.
+
Pantall
A, Teulier
C, Smith
BA, Moerchen
V, Ulrich
BD. Impact of enhanced sensory input on treadmill step frequency: infant born with myelomeningocele.
Pediatr Phys Ther. 2011;23(1):42–52.
[PubMed: 21266940]
+
Phillips
LA, Burton
JM, Evans
SH. Spina bifida management.
Curr Probl Pediatr Adolesc Health Care. 2017;47(7):173–177.
[PubMed: 28734746]
+
Ross
M, Brewer
K, Wright
FV, Agur
A. Closed neural tube defects: neurologic, orthopedic, and gait outcomes.
Pediatr Phys Ther. 2007;19(4):288–295.
[PubMed: 18004196]
+
Ryan
KD, Ploski
C, Emans
JB. Myelodysplasia--the musculoskeletal problem: habilitation from infancy to adulthood.
Phys Ther. 1991;71(12):935–946.
[PubMed: 1946627]
+
Sandler
A. Living with Spina Bifida: A Guide for Families and Professionals. 2nd ed. Chapel Hill, NC: University of North Carolina Press; 1997.
+
Sawin
KJ, Bellin
MH. 2010. Quality of life in individuals with spina bifida: a research update. Dev Disabilities Research Rev. 16:47–59.
+
Schoenmakers
MA, Uiterwaal
CS, Gulmans
VA, Gooskens
RH, Helders
PJ. Determinants of functional independence and quality of life in children with spina bifida.
Clin Rehabil. 2005;19(6):677–685.
[PubMed: 16180605]
+
Swanson
ME, Dicianno
BE. Physiatrists and developmental pediatricians working together to improve outcomes in children with spina bifida.
Pediatr Clin North Am. 2010;57(4):973–981.
[PubMed: 20883886]
+
Thibadeau
J, Reeder
MR, Andrews
J,
et al. Understanding the natural progression of spina bifida: prospective study.
JMIR Res Protoc. 2017;6(9):e180. doi: 10.2196/resprot.7739
[PubMed: 28912114]
+
Tombaugh
TN. Trail Making Test A and B: normative data stratified by age and education. Arch Clin Neuropsych. 2004;19(2):203–214.
+
Uniform Data System for Medical Rehabilitation. WeeFIM II System. Amherst, NY: University at Buffalo, The State University of New York; 2009.
+
Wolf
LS, McLaughlin
JF. Early motor development in infants with meningomyelocele. Pediatr Phys Ther. 1992;12–17.
++
Group of neuromuscular disorders characterized by delayed muscle relaxation after voluntary contractions (Makela, 2005).
Caused by prolonged bursts of action potentials due to muscle fiber hyperexcitability (Cannon, 2015)
Sustained contractions may last for several seconds to minutes and cause discomfort (Cannon, 2015)
Non-dystrophic Myotonias (Channelopathies) result from gene mutations affecting skeletal muscle voltage-gated ion channels (Meola, 2000), which specifically affect the sodium and chloride channels (Statland, 2013).
A rare group of diseases with a prevalence of 1:100,000 worldwide (Statland, 2013).
Sodium Channelopathies: mutations of chromosome 17 contribute to defects in SCN4A (Chrestian, 2006).
Dominant heredity
Onset first decade of life.
Mutations cause gain-of-function defects to sodium channels in muscles (Cannon, 2015)
Includes: Paramyotonia Congenita (PC), Potassium-aggravated myotonia (PAM), Periodic paralysis with myotonia (PPM) (Chrestian 2006).
Chloride Channelopathies: mutations of chromosome 7 contribute to defects in CLCN1 gene.
Mutations cause loss-of-function defects to chloride channels in muscles (Cannon, 2015)
Myotonia Congenita (MC) In one study of the contribution of CLCN1, three of four patients with CLCN1 were diagnosed with one of the types below (the one that was not categorized had not been diagnosed with a myotonia) (Vindas-Smith, 2015).:
Thomsen's MC - dominant variant, onset at infancy, less severe, with mutations that impact chloride channel function (Statland, 2013).
Becker's MC – recessive variant, onset in childhood (Chrestian 2006) with mutations that impair chloride conductance (Statland, 2013).
Both forms of myotonia congenita are more severe in men than in women (Platt, 2009).
Dystrophic Myotonia (DM1 and DM2): Splicing of chloride channel RNA forms defect in CLCN1 (Chrestian, 2006; Bermudez de Leon, 2007)). New research has neuro link: DMPK gene CUG repeats (Bermudez de Leon, 2007).
The most common adult muscular dystrophy (Meola, 2013).
Infancy to early adulthood (Miller, 2007)
Prevalence of DM is estimated at 1/20,000 (Smith, 2000).
DM1: most common form of dystrophy in muscles during pregnancy (Zaki, 2007).
Formerly known as Steinert's disease (Meola, 2015)
Caused by a CTG repeat expansion within the DMPK gene of chromosome 19 (Meola, 2015)
DM-1 is classified into three categories: mild, classical, or congenital (Smith, 2000).
DM-1 can be divided into four subgroups: congenital, early childhood, adult onset, and late onset/ asymptomatic (Meola, 2013).
Congenital form is most severe (Smith, 2000):
DM2: proximal myotonic myopathy (PROMM) (Smith, 2000): less severe.
Appears during adult life (Meola, 2013).
Caused by a CCTG repeat expansion within the CNBP gene of chromosome 3 (Meola, 2015)
Different from DM1 in that it is generally less severe and lacks a congenital form (Meola, 2015)
Genetics:
Dominant heredity:
Genetic significance:
++
++
Myopathy tends to increase with age (Matthews, 2010)
Myotonia Congenita:
Generally no progressive weakness.
Reaction times slowed-- inability to relax muscles after contraction (Kurhara, 2005)
Cardiac involvement may manifest (Finsterer, 2007).
Paramyotonia Congenita:
generally no progressive weakness or cardiac involvement.
Less severe form of myotonia may only be active in cold weather (Kurhara, 2005).
DM1 and DM2:
Progressive muscle weakness even if myotonia is reduced.
Systemic manifestations may occur (Kurhara, 2005)
The congenital forms that are present at birth have a drastically shorter lifespan
Milder forms that present much later in adulthood are characterized by normal lifespan (Smith, 2000).
Patients with Myotonic Dystrophy type 1 have an increased risk for spinal deformities, contractures, foot deformities, and fractures (Schilling, 2013).
"Cardiorespiratory disorders are responsible for 70% of the mortality in DM1" (Meola, 2013).
Expected sequelae:
Fractures due to falls and clumsiness, squinting, joint contractures, stridor, and aspiration pneumonia related to swallowing difficulties and cardiac involvement (Moxley, 1997; Finsterer, 2007).
Although not much is known about what effect these disorders have on quality of life, painful myotonia was found to be the best predictor of poor health and functioning (Matthews, 2010).
+++
IV. Medical/Surgical Management
++
The goal for treatment of nondystrophic myotonias is to reduce the sarcolemma excitability (Statland, 2013).
Sodium Channel Blockers
Mexiletine, tocainide, and flecainide decrease the severity of myotonia in individuals with non-dystrophic myotonia (Cannon, 2015)
Mexiletine relieves signs/symptoms of myotonia, including stiffness, weakness, and pain (Statland, 2012)
MC and PC-treated with Na channel blockers such as mexiletine, or phenytoin drugs decrease muscle stiffness (Kurhara, 2005).
For treating paramyotonia and the recessive form of myotonia, tocainide, mexiletine, and other class IB antiarrhythmic medications may be more successful than phenytoin (Statland, 2014). Myotonia in patients with myotonia congenita can improve with procainamide (Sharp 2014).
Mexiletine may be effective to calm NDM (non-dystrophic myotonia). In one study of multiple case trials, mexiletine was able to significantly reduce the "transitory depression" from average 62% to 28.8% (Lo Monaco, 2015)
First-line treatment choice of mycologist is class Ib anti-arryhthmic mexiletine (Matthews, 2010). Secondarily, class IC anti-arrhythmic drugs flecainide and propafenone are indicated. (Platt, 2009).
Mexiletine or carbamazepine can be used to treat myotonia in DM (Meola, 2015)
Mexiletine, gabapentin, NSAIDs, low-dose thyroid replacement, low-dose steroids, or tricyclic antidepressants can be used to manage pain in DM (Meola, 2015)
Diuretics acetazolamide and hydrochlorothiazide:
Carbonic Anhydrase Inhibitors, acetazolemide and dichlorphenamide
Used to prevent PPM (Platt, 2009). Carbamazepine beneficial in alleviating symptoms in MC (Savitha, 2006).
Risk of malignant hyperthermia during anesthesia with anesthesia-induced myotonic symptoms and prolonged recovery time in patients with Non-dystrophic Myotonias. (Rosenbaum, 2002).
For mild cases, only education of how to prevent episodes is needed.
In some patients with myotonia congenital, Acetazolamide is an effective treatment of myotonia (Sharp, 2014).
Tricyclic antidepressant drugs are beneficial in DM (Trip, 2006)
Clomipramine reduces grip relaxation time (Heatwole, 2012).
Defibrillators are important in the medical management of arrhythmias in patients with DM2 (Dalton, 2006).
Individuals with DM1 or DM2 can be given prokinetic drugs metochlopromide or tegaserod to ease gastric symptoms.
+++
V. Implications for Therapeutic Management
++
Physical Therapy Preferred Practice Pattern (APTA, 2017):
Muscle stiffness:
Education on proper "warming up" techniques before and during sports activities and the effect of cold temperature on symptoms.
Yoga or Pilates can be used for dynamic ROM exercises. Nutritionists: patient's diet (avoiding high potassium foods and consuming more carbohydrates) (Moxley, 1997).
Muscle Weakness:
Generalized strengthening exercises and specific hand training exercises (Aldehag, 2005).
Gait training and balance exercises for falls.
Too much or inappropriate exercise may increase the disease progression so finding the adequate amount and intensity is crucial (avoid overwork weakness)
NMES may be useful for improving muscle weakness and abnormal muscle activation (Chisari, 2013).
Proximal lower extremity muscle exercise of the knee extensors and hip flexors are beneficial to individuals with myotonia (Lindeman, 1995)
Systemic Manifestations:
Aerobic exercise to manage weight, increase cardiac health and overall fitness.
Patients with Dystrophic Myotonias should be monitored for diabetes mellitus and vision changes. Refer if necessary (Kurhara, 2005).
Pain Management:
NSAIDS or acetaminophen, ROM and strengthening exercises, and thermotherapy are the most common (Trip, 2006).
Patient Education:
Teach patients how to monitor respiratory cycle with pulse monitors, Borg scale, etc…
Frequent rest break need to be taken due to decreased exercise tolerance.
Avoid cold environments and strenuous exercise that could exacerbate symptoms (Matthews, 2010).
Avoid sudden movements after prolonged rest (Conravey, 2012).
Prevention: genetic counseling and prenatal ultrasound recommended with family history (Zaki, 2007).
Supports/devices:
Ankle-foot orthoses and neck braces may need to be prescribed to patients with dorsiflexors weakness (leading to foot drop) or neck muscle weakness.
Assistive devices, such as canes or walkers, may need to be fitted due to increased fall risk from vision problems and muscle weakness.
Relaxation techniques are beneficial as stress is can worsen myotonia (Conravey, 2012).
+++
VI. Consumer and Professional Resources
++
+
Aldehag
A, Jonsson
H, Ansved,
T. Effects of a hand training programme in five patients with myotonic dystrophy type 1.
Occupational Therapy International. 2005; 12(1): 14–27.
[PubMed: 15962697]
+
Bermúdez
de León M, Bulmaro
C. Myotonic dystrophy 1 in the nervous system: From the clinic to molecular mechanisms. Journal of Neuroscience Research. 2007. Vol 86 Issue 1, 18–26.
+
Cannon
SC. Channelopathies of skeletal muscle excitability.
Comprehensive Physiology. 2015;5(2):761–790.
[PubMed: 25880512]
+
Chisari
C, Bertolucci
F, Dalise
S, Rossi
B. Chronic muscle stimulation improves muscle function and reverts the abnormal surface EMG pattern in myotonic dystrophy: a pilot study.
J Neuroeng Rehabil. 2013;10:94. doi:10.1186/1743-0003-10-94.
[PubMed: 23938156]
+
Ciafaloni
E,
et al. The hypocretin neurotransmission system in myotonic dystrophy type 1.
Neurology. 2008; 70:226–230.
[PubMed: 18195268]
+
Chrestian
N,
et al. Myotonia congenita-a cause of muscle weakness and stiffness.
Nature Clinical Practice Neurology. 2006; 2(7): 393–399.
[PubMed: 16932590]
+
Conravey
A, Santana-Gould
L. Myotonia congenita and myotonic dystrophy: surveillance and management.
Curr Treat Options Neurol. 2010;12(1):16–28.
[PubMed: 20842486]
+
de Die-Smulders
CE, Höweler
CJ, Thijs
C, Mirandolle
JF, ANten
HB, Smeets
HJM, Chandler
KE, Geraedts
JPM. Age and causes of death in adult-onset myotonic dystrophy.
Brain. 1998;121 (Pt 8):1557–1563.
[PubMed: 9712016]
+
Drost
G, Stunnenberg
BC, Trip
J, Borm
G, McGill
KC, Ginjaar
I, van der Kooi
AW, Zwarts
MJ, van Engel
B, Faber
CG, Stegeman
DF, Lateva,
Z. Myotonic discharges discriminate chloride from sodium muscle channelopathies.
Neuromuscul Disord. 2015;25(1):73–80.
[PubMed: 25454733]
+
Fialho
D,
et al. Chloride channel myotonia: exon 8 hot-spot for dominant negative interactions.
Brain. 2007; 130:3265–3274.
[PubMed: 17932099]
+
Finsterer
J,
et al. Cardiac involvement over 10 years in myotonic and becker muscular dystrophy and mitochondrial disorder.
International Journal of Cardiology. 2007; 119: 176–184
[PubMed: 17258336]
+
Groh
W,
et al. Electrocardiographic abnormalities and sudden death in myotonic dystrophy 1. New England J of Med. 2008.Volume 358:2688–2697.
+
Heatwole
C, Bode
R, Johnson
N,
et al. Patient-reported impact of symptoms in myotonic dystrophy type 1 (PRISM-1).
Neurology. 2012;79(4):348–357.
[PubMed: 22786587]
+
Heatwole
CR, Moxley
RT III. The nondystrophic myotonias.
Neurotherapeutics. 2007; 4(2):238–51.
[PubMed: 17395134]
+
Heatwole
CR, Statland
JM, Logigian
EL. The diagnosis and treatment of myotonic disorders.
Muscle & Nerve. 2013; 47(5):632–648.
[PubMed: 23536309]
+
Hermans
MCE, Faber
CG, Bekkers
SCAM, Die-Smulders
CEM, Gerrits
MM, Merkies
ISJ, Snoep
G, Pinto
YM, Schalla,
S. Structural and functional cardiac changes in myotonic dystrophy type 1: a cardiovascular magnetic resonance study.
Journal of Cardiovascular Magnetic Resonance. 2012; 14(1):48–54.
[PubMed: 22827830]
+
Jakkani
R, Jyoti
S, Ahmed
M, Thomas
MM. Magnetic resonance imaging findings in adult-form myotonic dystrophy type 1.
Singapore Med J. 2012;53(7):e150–152.
[PubMed: 22815033]
+
Kurhara
T. New classification and treatment for myotonic disorders. Internal Medicine. 2005; 44(10): 1027–1032.
+
Lindeman
E, Leffers
P, Spaans
F, Drukker
J, Reulen
J, Kerckhoffs
M, Koke
A Strength training in patients with myotonic dystrophy and hereditary motor and sensor neuropathy: A randomized clinical trial.
Arch Phys Med Rehabil. 1995;76(7):612–620..
[PubMed: 7605179]
+
Lo Monaco
M, D'Amico
A, Luigetti
M, Desaphy
J-F, Modoni
A. Effect of mexiletine on transitory depression of compound motor action potential in recessive myotonia congenita.
Clin Neurophysiol. 2015;126(2):399–403.
[PubMed: 25065301]
+
Makela
JP, Somer
H. Myotonias and army personnel: symptoms and effects on service fitness.
Military Medicine. 2005; 170: 806–809.
[PubMed: 16261989]
+
Matthews
E, Fialho
D, Tan
SV,
et al. The non-dystrophic myotonias: molecular pathogenesis, diagnosis and treatment.
Brain. 2010; 133(1):9–22.
[PubMed: 19917643]
+
Meola
G, Sansone
V. Therapy in myotonic disorders and in muscle channelopathies. Neurological Science. 2000; 21: S953–S961
+
Meola
G, Cardani
R. Myotonic dystrophies: an update on clinical aspects, genetic, pathology, and molecular pathomechanisms. Neuromuscular Diseases: Pathology and Molecular Pathogenesis. 2015;1852(4):594–606.
+
Meola
G. Clinical aspects, molecular pathomechanisms and management of myotonic dystrophies. Acta Myologica. 2013;32(3):154–165.
+
Miller
TM. Differential diagnosis of myotonic disorders.
Muscle & Nerve. 2008; 37(3):293–299.
[PubMed: 18067134]
+
Moxley
III RT. Myotonic disorders in childhood: diagnosis and treatment.
Journal of Child Neurology. 1997: 12: 116–129.
[PubMed: 9075021]
+
Peter
S. Harper, Baziel
van Engelen, Bruno
Eymard, Douglas
E. Wilcox. Myotonic Dystrophy. Present management, future therapy. Oxford University Press 2004
+
Platt
D, Griggs
R. Skeletal Muscle Channelopathies: New insight into the periodic paralyses and nondystrophic myotonias.
Curr Opin Neurol. Oct 2009. 22(5): 524–531.
[PubMed: 19571750]
+
Ranum
LP, Day
JW. Myotonic dystrophy: RNA pathogenesis comes into focus.
Am J Hum Genet. 2004; 74(5): 793–804.
[PubMed: 15065017]
+
Rosenbaum
HK, Miller
JD. Malignant hyperthermia and myotonic disorders.
Anesthesiol Clin North America. 2002; 20(3): 623–664.
[PubMed: 12298310]
+
Savitha
MR, Krishnamurthy
B, Hyderi
A, Ul-Haque
F, Ramachandra
N. Myotonia Congenita – A Successful Response to Carbamazepine.
Indian J Pediatr. 2006. 73(5): 431–433.
[PubMed: 16741331]
+
Schilling
L, Forst
R, Forst
J, Fujak
A. Orthopaedic Disorders in Myotonic Dystrophy Type 1: descriptive clinical study of 21 patients.
BMC Musculoskeletal Disorders. 2013;14(1):1–17.
[PubMed: 23281775]
+
Sharp
L, Trivedi
JR. Treatment and management of neuromuscular channelopathies. Curr Treat Options Neurol. 2014; Oct;16(10):313. doi10.1007/s11940-014-0313-6.
+
Singh
RR, Tan
SV, Hanna
MG, Robb
SA, Clarke
A, Jungbluth
H. Mutations in SCN4A: A Rare but Treatable Cause of Recurrent Life-Threatening Laryngospasm.
Pediatrics. 2014;134(5):e1447–e1450.
[PubMed: 25311598]
+
Smith
C, Bennett
R, Bird
T. Myotonic Dystrophy: Making an Informed Choice About Genetic Testing.
Medical Geriatrics and Neurology, University of Washington Medical Center. Aug 2000.
http://www.rehabinfo.net. Accessed 7 Feb 2013.
+
Statland
J, Barohn
R. Muscle channelopathies: the nondystrophic myotonias and periodic paralyses. Contin Lifelong Learn Neurol. 2013;19(6):1598–1614.
+
Statland
J, Phillips
L, Trivedi
JR. Muscle channelopathies.
Neurol Clin. 2014;32(3): 801–815.
[PubMed: 25037091]
+
Strommen
J,
et al. Neuromuscular rehabilitation and electrodiagnosis.3. Diseases of muscles and neuromuscular junction. Archives Physical Medical Rehabilitation. 2005; 86(S1): S18–S27.
+
Sugie
K, Sugie
M, Taoka
T,
et al. Characteristic MRI findings of upper limb muscle involvement in myotonic dystrophy type 1.
PLoS ONE. 2015;10(4):e0125051. DOI#: 10.1371/journal.pone.0125051.
[PubMed: 25919300]
+
de Swart
BJM, van Engelen
BGM, van de Kerkhof
JPBM, Maassen
B. Myotonia and flaccid dysarthria in patients with adult onset myotonic dystrophy.
J Neurol Neurosurg Psychiatry. 2004;75(10):1480–1482.
[PubMed: 15377703]
+
Trip
J, Drost
G
et al.. Drug treatment for myotonia. Cochrane Database Syst. Rev. 2006; (1):CD004762.
+
Trivedi
JR, Cannon
SC, Griggs
RC. Nondystrophic myotonia: Challenges and future directions.
Exp Neurol. 2014;253(0):28–30.
[PubMed: 24361411]
+
Turner
C, Hilton-Jones
D. The myotonic dystrophies: diagnosis and management.
J Neurol Neurosurg Psychiatry. 2010; 81:358–367.
[PubMed: 20176601]
+
Udd
B, Krahe
R. The myotonic dystrophies: molecular, clinical, and therapeutic challenges.
The Lancet Neurology. 2012;11(10):891–905.
[PubMed: 22995693]
+
Udd
B, Meola
G, Krahe
R, Wansink
DG, Bassez
G, Kress
W, Schoser
B, Moxley
R. Myotonic dystrophy type 2 (DM2) and related disorders. Report of the 180
th ENMC workshop including guidelines on diagnostics and management.
Neuromusc disorders. 2011; 21(6): 443–450.
[PubMed: 21543227]
+
Vihola,
A
et al.. Histopathological differences of myotonic dystrophy type 1(DM1) and PROMM/DM2.
Neurology. 2003; 60 (11):1854–7.
[PubMed: 12796551]
+
Vindas-Smith
R, Fiore
M, Vásquez
M, Cuenca
P, Valle
G, Lagostena
L, Gaitan-Penas
H, Estevez
R, Pusch
M, Morales
F. Identification and Functional Characterization of CLCN1 Mutations Found in Nondystrophic Myotonia Patients.
Human Mutation. 2016;37(1):74–83.
[PubMed: 26510092]
+
Weinberg
J,
et al. Occult presentation of myotonia congenita in a 15-year old athlete.
The American Journal of Sports Medicine. 1999; 27: 529–531.
[PubMed: 10424227]
+
Zaki
M,
et al. Congenital myotonic dystrophy: prenatal ultrasound findings and pregnancy outcome. Ultrasound Obstetrics Gynecology. 2007; 29: 284–288.
++
++
A group of autosomal dominant genetic disorders that are associated with tumors affecting the central and peripheral nervous system encompassing the rare diseases of neurofibromatosis 1, neurofibromatosis 2, and schwannomatosis (Simone, 2011; Widemann, 2014)
++
++
++
++
NF 1:
Diagnostic Criteria- 2 or more of the following (Tonsgard, 2006):
6 or more café-au-lait macules >0.5 cm pre-puberty and >1.5 cm post-puberty; usually first sign of disease; present in >99% of NF1 (Shofty, 2015)
Axillary or inguinal freckling
2 or more cutaneous dermal neurofibromas; usually present at 5+ years; present in >99% of NF1 (Shofty, 2015)
1 plexiform neurofibroma
2 or more iris Lisch nodules
Optic glioma
Bony lesion
Primary relative with NF1
Approximately 50% of NF1 patients meet criteria by age 1 and 97% meet criteria by age 8 (Koontz,2013)
Other associated findings:
Cognitive deficits with low IQ, visual disturbances, ptosis, visual spatial difficulty, focal neurological deficits, decreased bowel/bladder function, weakness, seizures, headache, learning disabilities, , scoliosis, short stature, proptosis, precocious puberty, pseudarthrosis, tibial dysplasia, gynecomastia, renal artery stenosis, ataxia, brisk deep tendon reflexes, attention deficit hyperactivity disorder (ADHD), clonus, paresthesia of hands/feet, bony erosion or dysplasia, speech and language disorder, heart murmur, mood/anxiety disorder, executive dysfunction, cutaneous and subcutaneous neurofibromas causing itch and deformity, spatiotemporal gait deficits with impaired balance and impaired running speed in children (Ferner, 2007; Tonsgard, 2006; Gutmann, 1997; Simone, 2011; Champion, 2014; Descheemaeker, 2004)
Children with NF1 have 10 times prevalence of hypertension than healthy controls (Dubov, 2015).
Adults with NF1 have lower prevalence of fasting blood glucose levels (FBG) compared to controls (Martins, 2016)..
NF1 patients with paraspinal neurofibromas have a higher prevalence of morphological changes of vertebrae (Hu, 2016).
Pulmonary hypertension is an extremely rare occurrence with NF1.
Bony changes are classic features (Bamps, 2015)
Examples: rib-penciling (diagnosed when a rib is smaller in diameter than the midportion of the 2nd rib), vertebral scalloping, transverse process spindling, apical wedging, foraminal enlargement, and dural ectasia (widening or ballooning of the dural sac surrounding the spinal cord)
Brain volume is increased in children and adolescents with NF1.
NF 2:
Hallmark finding is:
Bilateral vestibular schwannomas (on radiographic imaging), primary relative with NF 2, and unilateral vestibular schwannoma (VS)
Or 2 of the following:
Meningioma, schwannoma, glioma, neurofibroma, juvenile posterior subscapular lens opacity; unilateral vestibular schwannoma and two of the following: meningioma, schwannoma, glioma, neurofibroma, juvenile posterior subscapular lens opacity; multiple meningiomas and unilateral vestibular schwannoma
Average age at diagnosis is 17-24 with an average delay in diagnosis of 7 years (Koontz, 2013; Hoa, 2012; Neff, 2005)
Presumptive/Possible NF 2:
Unilateral VS <30 with at least one of the following: meningioma, glioma, schwannoma, juvenile cortical cataract OR with more than 2 meningiomas, Unilateral VS <30, and one of the following: glioma, schwannoma, juvenile cortical cataract (Hoa, 2012)
Non-tumoral calcifications with or without tumoral calcifications can suggest a diagnosis of NF-2 in patients with no obvious lesion in cerebellopontine angles on CT (Aiyappan 2015)
Spinal tumors are found more commonly in the cervical spine in patients with NF2.
Spinal tumors are more common in more severe cases of NF2 (Nowak, 2016).
Other:
Hearing loss, tinnitus, dysequilibrium, HA, facial numbness/weakness, dysphagia, cranial nerve signs and symptoms, compressive myelopathy, and seizures 2° intracranial meningioma or vestibular schwannoma, ophthalmologic effects, tumor types of spinal, skin, meningioma, and bilateral vestibular schwannoma (Neff, 2005; Kulku, 2007; Abarrah, 2009; Hoa, 2012; Simone, 2011)
Schwannomatosis:
Diagnostic Criteria:
Age >30 years and two or more nonintradermal schwannomas (at least one with histological confirmation); one schwannoma confirmed to be pathological plus a primary relative meeting criterion; must not fit any criteria of NF2; no vestibular schwannoma on MRI scan; no primary relative with NF2 (Baser, 2006).
Possible Diagnosis:
Age <30 and two or more nonintradermal schwannomas with at least one with histological confirmation; no evidence of vestibular tumor on MRI; no known NF2 Mutation OR Age >45; two nonintradermal schwannomas, one with histological confirmation and no symptoms of cranial nerve VIII pathology; no NF2 OR Radiographic evidence of nonvestibular schwannoma and first-degree meeting of criteria.
Segmental Diagnosis:
Patients who have a diagnosis of schwannomatosis tend to be younger than those presenting with Schwannomas.
Spinal schwannomas in patients with schwannomatosis appear predominantly in the lumbar area (Peng, 2016).
Those presenting with Schwannomas but not presenting with NF2 criteria should be suspect for schwannomatosis (Gonzalvo, 2011).
In the absence of other characteristic NF2 manifestations, the absence of bilateral VS and presence of intra-dermal schwannomas are the main clinical criteria to differentiate schwannomatosis instead of NF2 (Castellanos, 2015)
Other signs and symptoms
Severe pain, weakness, numbness, and tingling in fingers or toes (MacCollin, 2005)
Depression is common in people with schwannomatosis (Merker, 2012).
++
Diagnostic Testing (Gutmann, 1997)
++
NF 1
NF 2: MRI, auditory brainstem evoked potentials, acoustic reflex testing, molecular testing (Neff, 2005; Hoa, 2012)
Schwannomatosis: MRI and CT scan of peripheral nerves/spinal cord
++
General:
NF1:
Progressive symptoms depending on age group
Learning disabilities/visual deficits
Malignancy most common cause of death
If malignancy occurs, life expectancy commonly decreased by 10-15 years with an increased mortality rate in young adults (Friedman, 1999; Tonsgard, 2006).
In virtually all adults with NF1, cutaneous neurofibromas occur (Jouhilahti, 2011).
Approximately 33% of patients with NF1 develop plexiform neurofibromas of which 2-5% develop MPNST
Scoliosis is the most frequent musculoskeletal manifestation in NF-1. It usually occurs in the low thoracic region and has a prevalence ranging from 2% to 69% in the literature (Bamps, 2015)
Patients with NF1 related osteopenia and an age ~40 tend to develop osteoporosis due to accelerated bone loss (Heerva, 2012).
NF2:
Variable
Progressive loss of hearing, ambulation, and balance
Chronic pain
Patients with NF2 are expected to have a decreased life expectancy (Koontz,2013)
Death may occur as early as 40 years of age in more severe cases like Wishart form. Wishart form is characterized by tumor growth with the disease showing a severe phenotype; therefore, increasing progression of NF2 (Simone, 2012).
Tumors compressing vital structures such as the brainstem or other cranial nerves can be fatal (Friedman, 1999).
The presence of spinal tumors in patients with NF 2 indicates poor prognosis. Regular monitoring for spinal tumors can assist in maintaining a more functional prognosis (Aboukais, 2013).
Schwannomatosis:
Pain or medications can cause disability in severe cases; no decreased life expectancy (NINDS, 2007).
Chronic pain was the most common symptom in 68% of cases and usually persisted despite aggressive surgery and medication use and often associated with anxiety and depression (Widemann, 2012)
+++
IV. Medical/Surgical management
++
General:
No known cure
Multi-disciplinary treatment (Tonsgard, 2006).
Therapies targeting mast cell function, different Ras signaling pathway components, as well as growth factor receptors, may prove successful (Bennett, 2009).
NF 1 & 2:
Chemotherapy (Vincristine and Carboplatin) for optic gliomas and surgical removal of dermal/plexiform neurofibromas (Friedman, 1999).
Surgery
Surgical management of bony defects, accompanied with bisphosphonate treatment to promote healing (Simone, 2011)
Age, tumor type, location, and depth are helpful to estimate the progression of plexiform neurofibromas after surgery in NF1 patients.
Patients benefit from elective surgery of small and completely removable plexiform neurofibromas (Nguyen 2013)
If extremity function is compromised, amputation may be necessary (Wong, 1998)
If plastic surgery is wanted/needed, patient should wait until after the age of 18 to establish dx progression (Lee, 2003)
The posterior-only surgical correction of dystrophic scoliosis in patients with NF1 using a multi anchor point method can have satisfactory clinical efficacy (Deng 2017).
Vitamin D3 Supplementation raises BMD (Schnabel, 2012)
Amitriptyline and Topiramate if migraines occur (Tonsgard, 2006)
Pirfenidone (antifibrotic)- decrease size and progression of tumor (Babovic-Vuksanovic, 2006)
Vestibular schwannoma:
Hearing screen; surgery with hearing loss; possible cochlear or auditory brainstem implant depending on functional status of cochlear nerve and facial nerve function (Neff, 2005; Otto, 2001)
Stereotactic radiation is good for primary tumor (Kida, 2000; Mathieu, 2007)
Middle fossa resection has high rate of hearing preservation in children with NF2 (Slattery, 2007)
Early surgical intervention via a translabyrinthine approach has been shown to be beneficial for vestibular schwannomas in patients with NF2 (Nowak, 2015).
Bevacizumab can improve hearing and reduce vestibular schwannoma size in patients with NF2, but possible severe side effects must be considered (Alanin, 2015; Goutagny, 2017)
NF1 gene therapies: Anti-RAS therapy, RAS antibodies and viruses, Rapamycin inhibiting NF1-MPNST mTOR pathway (Oren, 2009)
Radiotherapy for NF1 may be used as a palliative treatment or when progression or recurrence outweighs the risks
Radiation therapy can be effective for local and symptomatic control with well-tolerated toxicities (Kahn, 2014)
Radiation for NF2 may be considered if vestibular tumors are less than 3cm to avoid surgery (Simone, 2011)
Schwannomatosis:
Surgical removal of tumors if symptomatic unless compression of spinal cord occurs (MacCollin, 2005; Parsons, 2009)
If the lesions are symptomatic, surgical intervention is necessary
Asymptomatic lesions are usually monitored through serial MRI studies yearly with no surgical intervention. Patients with asymptomatic lesions are educated on catching early signs and symptoms of symptomatic schwannomas (Huang, 2004).
Pain management:
Na channel antagonist, gabapentin, opioids, NSAIDS, oxycodone formulations. (MacCollin, 2005; Merker 2012)
Antidepressants:
Early results suggest stereotactic radiation is safe and effective for those without tumor suppressor syndrome (Plotkin, 2013)
+++
V. Key Aspects of therapeutic management
++
Possible Preferred Practice Pattern's from the "Guide to Physical Therapist Practice: 5E Impaired Motor Function and Sensory Integrity Associated with Progressive Disorders of the Central Nervous System; Other possible- 5A, 5B, 5F, 5I
Contraindications/Precautions:
Chemotherapy effects to tissue
Balance and falls with vestibular neuroma
Referrals from physicians: ophthalmologist, neurologist, otorhinolaryngologist, orthopedics, psychologist
Age specific monitoring of disease manifestations (Ferner, 2007)
Treatment:
Gait, locomotion, and balance
Vestibular exercises-VOR (Herdman, 1995)
Muscle performance: plyometrics, muscle strength, neuromuscular reeducation, agility (Johnson, 2012)
Motor function: motor control-movement: gait training (kulku, 2007), motor control-coordination, motor control-stability
Joint integrity/mobility: passive range of motion exercises, train isolated movement patterns, spinal mobility
Speech and language: speech therapy (Simone, 2011)
Integumentary: scar management
Education: home exercise program, self-help skills, support groups, lip reading (Kulku, 2007)
Assistive/adaptive devices: hearing aids (Kulku, 2007), cochlear implants (Kulku, 2007), auditory brainstem implants (Maini, 2009), protective bracing (Simone, 2011)
+
Abbarah
T, Abbarah
MA. Neurofibromatosis type 1 causing conductive hearing loss.
Ear Nose Throat J. 2009;88(5):912.
[PubMed: 19444785]
+
Aboukais
R, Baroncini
M, Zairi
F, Bonne
N, Schapira
S, Vincent
C, Lejeune
J. Prognostic value and management of spinal tumors in neurofibromatosis type 2 patients. Acta Neurochir. 2013;155(5):771–777.
+
Aiyappan
SK, Ranga
U, Veeraiyan
S. Neurofibromatosis type 2: intracranial calcifications as a clue to diagnosis.
J Neurosci Rural Pract. 2015;6(1):120–121.
[PubMed: 25552869]
+
Alanin
MC, Klausen
C, Caye-Thomasen
P,
et al. The effect of bevacizumab on vestibular schwannoma tumour size and hearing in patients with neurofibromatosis type 2. Eur Arch Oto-Rhino-Laryngol. 2015;272(12):3627–3633.
+
Arden-Holmes
SL, North
KN. Therapeutics for childhood neurofibromatosis type 1 and type 2. current treatment options. Neurology. 2011;13(6):529–543.
+
Babovic-Vuksanovic
D, Ballman
K, Michels
V,
et al. Phase II trial of pirfenidone in adults with neurofibromatosis type 1.
Neurology. 2006;67(10):1860–1864.
[PubMed: 17035676]
+
Bamps
S, Calenbergh
FV, Loon
JV, Paesschen
RV, Vanderschot
P. Posterolateral approach in a neurofibromatosis type-I patient with severe dystrophic thoracic kyphoscoliosis: a case report, cadaver study, and literature review.
J Neurosci Rural Pract. 2015;6(1):84–86.
[PubMed: 25552858]
+
Bennett
E, Thomas
N, Upadhyaya
M. J Ped Neurol. 2009;7(2):105–15.
+
Baser
M, Friedman
J, Evans
D. Increasing the specificity of diagnostic criteria for schwannomatosis.
Neurology. 2006; 66:730–732.
[PubMed: 16534111]
+
Binder
DK, Smith
JS, Barbaro
NM. Primary brachial plexus tumors: imaging, surgical, and pathological findings in 25 patients.
Neurosurg Focus. 2004;16(5): E11.
[PubMed: 15174831]
+
Castellanos
E, Bielsa
I, Carrato
C,
et al. Segmental neurofibromatosis type 2: discriminating two hit from four hit in a patient presenting multiple schwannomas confined to one limb.
BMC Med Genomics. 2015;8:2.
[PubMed: 25739810]
+
Champion
JA, Rose
KJ, Payne
JM, Burns
J, North
KN. Relationship between cognitive dysfunction, gait, and motor impairment in children and adolescents with neurofibromatosis type 1.
Dev Med Child Neurol. 2014;56(5):468–74.
[PubMed: 24387687]
+
Deng
A, Zhang
H-Q, Tang
M-X, Liu
S-H, Wang
Y-X, Gao
Q-L. Posterior-only surgical correction of dystrophic scoliosis in 31 patients with neurofibromatosis type 1 using the multiple anchor point method.
J Neurosurg Pediatr. 2017;19(1):96–101.
[PubMed: 27739946]
+
Descheemaeker
M-J, Ghesquière
P, Symons
H, Fryns
JP, Legius
E. Behavioural, academic and neuropsychological profile of normally gifted neurofibromatosis type 1 children.
J Intellect Disabil Res. 2005;49(1):33–46.
[PubMed: 15634310]
+
Dubov
T, Toledano-Alhadef
H, Chernin
G, Constantini
S, Cleper
R, Ben-Shachar
S. High prevalence of elevated blood pressure among children with neurofibromatosis type 1. Pediatr Nephrol Berl Ger. 2016;31(1):131–136.
+
Ferner
RE, Huson
SM, Thomas
N,
et al. Guidelines for the diagnosis and management of individuals with neurofibromatosis.
J Med Genet. 2007;44(2): 81–8.
[PubMed: 17105749]
+
Friedman
J, Gutmann
D, MacCollin
M, Riccardi
V. Neurofibromatosis: Phenotype, Natural History, and Pathogenesis. 3rd ed. Baltimore: Johns Hopkins University Press; 1999.
+
Gottfried
ON, Viskochil
DH, Couldwell
WT. Neurofibromatosis type 1 and tumorigenesis: molecular mechanisms and therapeutic implications.
Neurosurg Focus. 2010;28(1):E8.
[PubMed: 20043723]
+
Gonzalvo
A, Fowler
A, Cook
RJ,
et al. Schwannomatosis, sporadic schwannomatosis, and familial schwannomatosis: a surgical series with long-term follow-up: clinical article.
J Neurosurg. 2011;114(3):756– 762.
[PubMed: 20932094]
+
Goutagny
S, Kalamarides
M. Medical treatment in neurofibromatosis type 2. Review of the literature and presentation of clinical reports.
Neurochirurgie [online]. February 3, 2017.
https://doi.org/10.1016/j.neuchi.2016.09.004. Accessed October 24, 2017.
+
Gumbiene
L, Petrulioniene
Z, Rucinskas
K,
et al. Pulmonary hypertension: a fatal complication of neurofibromatosis type 1.
Respir Care. 2011;56(11):1844–1848.
[PubMed: 21605478]
+
Heervä
E, Leinonen
P, Kuorilehto
T,
et al. Neurofibromatosis 1-related osteopenia often progresses to osteoporosis in 12 years.
Calcif Tissue Int. 2012;92(1):23–27.
[PubMed: 23108390]
+
Herdman
S, Clendaniel
R, Mattox
D, Holliday
M, Niparko
J. Vestibular adaptation exercises and recovery: acute stage after acoustic neuroma resection.
Arch Otolaryngol Head Neck Surg[Archives of Otolaryngology Full Text]. 1995;113:77–87.
+
Hoa
M, Slattery
W. Neurofibromatosis 2. Otolaryngol Clin N Amer. 2012;45(2):315–332.
+
Hsieh
H-Y, Fung
H-C, Wang
C-J, Chin
S-C, Wu
T. Epileptic seizures in neurofibromatosis type 1 are related to intracranial tumors but not to neurofibromatosis bright objects.
Seizure. 2011;20(8):606–611.
[PubMed: 21621428]
+
Hu
Z, Liu
Z, Qiu
Y, Xu
L, Yan
H, Zhu
Z. Morphological differences in the vertebrae of scoliosis secondary to neurofibromatosis type 1 with and without paraspinal neurofibromas.
Spine. 2016;41(7):598–602.
[PubMed: 26780616]
+
Huang
JH, Simon
SL, Nagpal
S, Nelson
PT, Zager
EL. Management of patients with schwannomatosis: Report of six cases and review of the literature.
Surg Neurol. 2004;62(4):353–361.
[PubMed: 15451291]
+
Hulsebos
TJ, Plomp
AS, Wolterman
RA, Robanus-Maandag
E, Baas
F, Wesseling
P. Germline mutation of INI1/SMARCBI in familial schwannomatosis.
Am J Hum Genet. 2007;80(4):805–10.
[PubMed: 17357086]
+
Johnson
B, Salzberg
C, Stevenson
D. Effects of plyometric training program for 3 children with neurofibromatosis type 1.
Pediatr Phys Ther. 2012;24(2):199–208.
[PubMed: 22466394]
+
Jouhilahti
E-M, Peltonen
S, Heape
AM, Peltonen
J. The pathoetiology of neurofibromatosis 1. Amer J Pathol. 2011;178(5):1932–1939.
+
Kahn
J, Gillespie
A, Tsokos
M,
et al. Radiation therapy in management of sporadic and neurofibromatosis type 1-associated malignant peripheral nerve sheath tumors.
Front Oncol. 2014;4:324.
[PubMed: 25452937]
+
Kida
Y, Kobayashi
T, Tanaka
T, Mori
Y. Radiosurgery for bilateral neurinomas associated with neurofibromatosis type 2.
Surg Neurol. 2000;53:383–390.
[PubMed: 10825525]
+
Koontz
NA, Wiens
AL, Agarwal
A, Hingtgen
CM, Emerson
RE, Mosier
KM. Schwannomatosis: the overlooked neurofibromatosis? Amer J Roentgenology. 2013;200(6):W646–W653.
+
Kulku
DG, Vavulzer
G. Usual presentation of neurofibromatosis type 2 with hemiparesis and ataxia. Turkish J Phys Med Rehab. 2007;53(1):38–40.
+
Lee
V, Ragge
N, Collin
J. The surgical management of childhood orbito-temporal neurofibromatosis.
Br J Plast Surg. 2003;56: 380–387.
[PubMed: 12873467]
+
Li
P, Zhao
F, Zhang
J,
et al. Clinical features of spinal schwannomas in 65 patients with schwannomatosis compared with 831 with solitary schwannomas and 102 with neurofibromatosis type 2: a retrospective study at a single institution.
J Neurosurg Spine. 2016;24(1):145–154.
[PubMed: 26407091]
+
Maini
S, Cohen
MA, Hollow
R, Briggs
R. Update on long -term results with auditory brainstem implants in NFR2 patients. Cochlear Implants Int: An Interdisc J. 2009;10: Suppl 1:33–7.
+
Martins
AS, Jansen
AK, Rodrigues
LOC,
et al. Lower fasting blood glucose in neurofibromatosis type 1.
Endocr Connect. 2016;5(1):28–33.
[PubMed: 26631381]
+
Mathieu
D, Kondziolka
D, Flickinger
JC,
et al. Stereotactic radiosurgery for vestibular schwannomas in patients with neurofibromatosis type 2: an analysis of tumor control, complications, and hearing preservation rates. Neurosurg. 2007;60(3):460–8.
+
MacCollin
M, Chiocca
E, Evans
D,
et al. Diagnostic criteria for schwannomatosis.
Neurology. 2005;64(11):1838–1845.
[PubMed: 15955931]
+
Merker
VL, Esparza
S, Smith
MJ, Stemmer-Rachamimov
A, Plotkin
SR. Clinical features of schwannomatosis: a retrospective analysis of 87 patients.
Oncologist. 2012;17(10):1317–1322.
[PubMed: 22927469]
+
Neff
BA, Welling
DB. Current concepts in the evaluation and treatment of neurofibromatosis type II.
Otolaryngol Clin North Am. 2005;38(4):671–684.
[PubMed: 16005725]
+
Nguyen
R, Ibrahim
C, Friedrich
RE, Westphal
M, Schuhmann
M, Mautner
V-F. Growth behavior of plexiform neurofibromas after surgery.
Genet Med. 2013;15:691–697.
[PubMed: 23598713]
+
North
K, Hyman
S, Barton
B. Review Article : Cognitive deficits in neurofibromatosis 1.
J Child Neurol. 2002;17(8):605–612.
[PubMed: 12403559]
+
Nowak
A, Dziedzic
T, Czernicki
T, Kunert
P, Marchel
A. Management of spinal tumors in neurofibromatosis type 2 patients.
Neurologia i Neurochirurgia Polska. 2016;50(1):31–35.
[PubMed: 26851687]
+
Nowak
A, Dziedzic
T, Czernicki
T,
et al. Strategy for the surgical treatment of vestibular schwannomas in patients with neurofibromatosis type 2.
Neurologia i Neurochirurgia Polska. 2015;49(5):295–301.
[PubMed: 26377980]
+
Otto
S, Brackmann
D, Hitselberger
W, Shannon
R. Brainstem electronic implants for bilateral anacusis following surgical removal of cerebello pontine angle lesions.
Otolaryngol Clin North Am. 2001;34(2):485–499.
[PubMed: 11419419]
+
Parsons
CM, Canter
RJ, Khatri
VP. Surgical management of neurofibromatosis. Surg Oncol Clin North Am. 2009;18(1):175–96.
+
Plotkin
SR, Blakeley
JO, Evans
DG,
et al. Update from the 2011 International Schwannomatosis Workshop: from genetics to diagnostic criteria. Amer J Med Genetics Part A. 2013;161(3):405–416.
+
Pride
N, Payne
JM, Webster
R, Shores
EA, Rae
C, North
KN. Corpus callosum morphology and its relationship to cognitive function in neurofibromatosis type 1.
J Child Neurol. 2010;25(7):834–841.
[PubMed: 20142468]
+
Rea
D, Brandsema
JF, Armstrong
D,
et al. Cerebral arteriopathy in children with neurofibromatosis type 1.
Pediatrics. 2009;124(3):e476–e483.
[PubMed: 19706560]
+
Shnabel
C, Jett
K, Friedman
J, Frieling
I, Kruse
H, Mautner
V. Effect of vitamin D3 treatment on bone density in neurofibromatosis type 1 patients: a retrospective study.
Joint Bone Spine. 2013;80(3):315–319.
[PubMed: 23021159]
+
Shofty
B, Constantini
S, Ben-Shachar
S. Advances in molecular diagnosis of neurofibromatosis type 1.
Semin Pediatr Neurol. 2015;22(4): 234–239.
[PubMed: 26706011]
+
Slattery
WH, Fisher
LM, Hitselberger
W, Friedman
RA, Brackmann
DE. Hearing preservation surgery for neurofibromatosis type 2- related vestibular schwannoma in pediatric patients.
J Neurosurg. 2007;106:255–260.
[PubMed: 17465357]
+
Steen
RG, Taylor
JS, Langston
JW,
et al. Prospective evaluation of the brain in asymptomatic children with neurofibromatosis type 1: relationship of macrocephaly to T1 relaxation changes and structural brain abnormalities. Am J Neuroradiol. 2001;22(5):810–817.
+
Tonsgard
JH. Clinical manifestations and management of neurofibromatosis type 1.
Semin Pediatr Neurol. 2006;13:2–7.
[PubMed: 16818170]
+
Ward
BA, Gutmann
DH. Neurofibromatosis 1: from lab bench to clinic.
Pediatr Neurol. 2005;32:221–228.
[PubMed: 15797177]
+
Widemann
BC, Acosta
MT, Ammoun
S,
et al. CTF meeting 2012: Translation of the basic understanding of the biology and genetics of NF1, NF2, and schwannomatosis toward the development of effective therapies. Am J Med Genet A. 2014;164(3):563–78
+
Williams
V, Lucas
J, Babcock
M, Gutmann
D, Korf
B, Maria
B. Neurofibromatosis type 1, revisited.
Pediatrics. 2009;123(1):124–33.
[PubMed: 19117870]
+
Wong
W, Hirose
R, Scheithauer
B, Schild
S, Gunderson
L. Malignant peripheral nerve sheath tumor: analysis of treatment outcome. Int J Radia Oncol Biol Phys. 1998;42:351–360.
+++
Normal Pressure Hydrocephalus
++
Normal Pressure Hydrocephalus (NPH) occurs when ventricles slowly enlarge due to poor drainage of cerebrospinal fluid (CSF) from the ventricles (Dalvi, 2012)
Due to decreased CSF absorption, not excess formation (Verrees, 2004)
The arachnoid granulations are unable to sustain their baseline removal of CSF; this is often secondary to fibrosis and scarring that can obscure absorptive interfaces (Verrees, 2004; LaFlamme, 2005)
There are two main types of NPH (Tuppeny, 2014)
Idiopathic NPH
Secondary NPH
- Usually results from hemorrhage, infection, inflammatory conditions, or Paget's disease of the skull (Tuppeny, 2014)
Usually the cause is unknown (Dalvi, 2012), but hypertension and diabetes are both risk factors for developing NPH (Fraser, 2007).
The name is actually misleading since the CSF pressure is elevated above its normal values.
The mass from extra CSF volume in the ventricles causes impingement on other areas of the brain (Klassen, 2011).
Decreased blood supply and ventricular enlargement are the two major anatomic abnormalities present in NPH (Bradley, 2000)
The process usually develops slowly and almost always in older adults (average age over 60) (Klassen, 2011).
++
++
If left untreated, patients with NPH will slowly get worse, eventually resulting in death (Ettinger, 2011).
Shunt placement has been shown to give less improvement on dementia and urinary problems while the greatest improvement is usually in gait (Klassen, 2011).
Studies have shown that shunt placement surgery success is related to classical triad features present (gait disturbance, dementia, and urinary incontinence), "positive result on lumbar drain test, and the presence of pressure signs on CT or MRI such as periventricular transependymal effusion (Black, 2007)."
Studies have shown no correlation of surgery success with the person's age and degree of dementia (Black, 2007).
Shunt placement has shown to have improvement in only 1/3 of the patients after three years (Klassen, 2011).
Early diagnosis and treatment improves the chance of a good recovery (Chen, 2010).
Possible complications associated with NPH include:
Surgical complications (bleeding, infection), worsening dementia, injury from falls, and shortened life span.
The decision to conduct surgery requires considerations of the probability of improvement (Oliveira, 2014).
Favorable indicators of post-operative improvement include early onset of gait disorders and onset of symptoms for less than six months
Adverse indicators include absence of changes in gait or its emergence after the onset of the disease, early onset dementia, mild to severe dementia, dementia for more than two years, atrophy, and severe impairment of the white matter
++
Ventriculoperitoneal (VP) shunt placement
Most common treatment is a permanent VP shunt placed surgically into the ventricle running down to the peritoneal cavity that can regulate the pressure of the CSF (Black, 2007; Tuppeny, 2014)
Current gold-standard treatment for idiopathic NPH (Oliveira, 2014; Tuppeny, 2014).
Guidelines for determining whether shunting should be performed on a patient with NPH (Shprecher, 2008):
Normalized power variance (NPV) of non-invasive electroencephalography (EEG) waves in the beta frequency band, before surgery, can distinguish responders and non-responders to VP shunt placement (Aoki, 2015).
Surgical CSF diversion is used to determine if patients are good candidates for shunting and can be performed several ways: (Tuppeny, 2014; Williams, 2006)
- Continuous CSF Drainage
A spinal catheter is temporarily inserted over a 3-day period to remove a pre-determined amount of CSF every 2 hours at a rate of 10 mL/hr (Tuppeny, 2014; Williams, 2006). This procedure requires an overnight stay in the hospital (Tuppeny, 2014).
- High Volume Lumbar Puncture
This process removes 30-50 mL of CSF using an 18 or 20 gauge spinal needle (Tuppeny, 2014; Williams, 2006).
Gait is assessed over the next 4-6 hours to determine if CSF diversion is effective (Williams, 2006).
- Prolonged drainage of 300 mL or more is associated with a better response than surgical CSF diversion with a permanent shunt (Tuppeny, 2014).
Patients who underwent ventriculoperitoneal shunt (VS) placement showed significant improvement compared to the control at both 1-month and 6-month post-op on the Barthel Index and Mini-Mental State Examination (Chen, 2010).
Studies have found that as much as 80% of patients will see some benefit from VS in particular concerning gait and urinary problems (Black, 2007).
Kazui found that those with mild symptoms including a low idiopathic normal pressure hydrocephalus grading scale (iNPHGS) gait score, a low TUG score, and a high MMSE total score were significant predictors that all three triad symptoms would reverse post shunt surgery (Kazui, 2013).
- Young age, which was not defined by the study, was also a predictor of the disappearance of gait disturbances; and a successful visuconstruction subtest as well as an absence of hypertension were predictors of disappearance of cognitive impairment for patients receiving shunt (Kazui, 2013).
Endoscopic third ventriculostomy
Sometimes a hole is made in the bottom of the third ventricle giving another exit for CSF (Dalvi, 2012).
This technique has been successful, but pressures commonly found in NPH may not allow the ventriculostomy to remain open (Tuppeny, 2014).
Regular lumbar punctures can be used to relieve symptoms.
No medication is significantly beneficial (Ninds, 2011)
Surgical excision of tumor if it is the cause of NPH
+++
V. Implications for Therapeutic Management (LaFlamme, 2005)
++
Physical Therapy Preferred Practice Pattern (APTA, 2017)
Practice Pattern 5E: Impaired Motor Function and Sensory Integrity Associated With Progressive Disorders of the Central Nervous System
Practice Pattern 5A: Primary Prevention/Risk Reduction for Loss of Balance and Falling
Rehabilitation strategies should always be included in the treatment of NPH as a supplementary treatment option to focus on recovery of function (Oliveira, 2014).
Physical exercise to preserve cognitive function: moderate exercise (including 5 minutes on a UBE and then 3 sets of 10 for bicep curls, shoulder and arm presses with 1-3 lb weights, and shoulder external rotation exercises) increases levels of vascular endothelial growth factor (VEGF) in patients with chronic NHP. This plays an important role in preserving cognitive function (Yang, 2015).
Intervention for impaired ROM and muscle performance
Muscle strengthening and re-education, including moderate upper-body resistance exercise (upper body bike, bicep curls, shoulder and arm presses) (Yang, 2015)
Increase ROM (in particular trunk rotation and hip extension) (LaFlamme, 2005)
Intervention to improve function and ADLs
Gait, balance, bed mobility, assistive device training, and transfer training
Patient Education
The role of interdisciplinary teams
+++
VI. Consumer and Professional Resources
++
Website for patient information is http://www.lifenph.com/index.asp.
The site has a self-assessment for NPH.
A free NPH information kit based on the patient's current symptoms.
Personal stories submitted by people with NPH can be found on the site.
It also has a variety of information from articles, signs, and local resources for help (Lifenph, 2012).
Website for helping a person with hydrocephalus is http://www.hydroassoc.org/.
It has information about NPH and hydrocephalus
It has events, support groups, a hydrocephalus library, and a frequently asked questions page. (Hydrocephalus Association, 2012)
National Hydrocephalus Foundation - http://nhfonline.org
Includes the following: 24/7 telephone hotline, parent-to-parent and adult-to-adult contacts, support groups, help guides, access to NHF Reference Library, Physician referrals, and a newsletter.
The NINDS has an information page on NPH and can be found at: http://www.ninds.nih.gov/disorders/normal_pressure_hydrocephalus/normal_pressure_hydro cephalus.htm.
+
Andrén
K, Wikkelsø
C, Tisell
M, Hellström
P. Natural course of idiopathic normal pressure hydrocephalus.
J Neurol Neurosurg Psychiatry. 2014;85(7):806–810.
[PubMed: 24292998]
+
Aoki
Y, Kazui
H, Tanaka
T, Ishii
R, Wada
T, Ikeda
S, Hata
M, Canuet
L, Katsimichas
T, Musha
T, Matsuzaki
H, Imajo
K, Kanemoto
H, Yoshida
T, Nomura
K, Yashiyama
K, Iwase
M, Takeda
M. Noninvasive prediction of shunt operation outcome in idiopathic normal pressure hydrocephalus.
Sci Rep. 2015;5:7775. doi:10.1038/srep07775.
[PubMed: 25585705]
+
Black
P, Ingraham
FD. Normal pressure hydrocephalus. In: Rajshekhar
V, Bhattacharyya
KB. Progress in Clinical Neurosciences. 22nd ed. Delhi, India: Byword Books Private Limited; 2007: 289–294.
+
Bloomsterwall
E, Bilting
M, Stephensen
H, Wikkelso
C; Gait Abnormality is not the Only Motor Disturbance in Normal Pressure Hydrocephalus. Scand J Rehabil Med. 1995 Dec; 27 (4): 205–9.
+
Bradley
WG. Normal pressure hydrocephalus: new concepts on etiology and diagnosis. Am J Neuroradiol. 2000;21:1586–1590.
+
Chen
Z, Song
W, Du
J, Li
G, Yang
Y, Ling
F. Rehabilitation of patients with chronic normal-pressure hydrocephalus after aneurysmal subarachnoid hemorrhage benefits from ventriculoperitoneal shunt. Topics in Stroke Rehabilitation, 2009 Sep-Oct; 16 (5): 330–8.
+
Feick
D, Sickmond
J, Liu
L, Metellus
P, Williams
M, Rigamonti
D, Hill-Briggs
F. Sensitivity and predictive value of occupational and physical therapy assessments in the functional evaluation of patients with suspected normal pressure hydrocephalus.
J Rehabil Med. 2008;40(9):715–720.
[PubMed: 18843422]
+
Fraser,
J, Fraser,
C. Gait Disorder Is the Cardinal Sign of Normal Pressure Hydrocephalus: A Case Study. J Neurosci Nursing. 2007; 39(3):132–134, 192.
+
Gor-Garcia-Fogeda
M, Cuerda
R, Tejada
M, Alguacil-Diego
I, Molina-Rueda
F. Observational Gait Assessments in People With Neurological Disorders: A Systematic Review. Amer Congress Rehabil Med. 2016; 97:131–140.
+
Ghosh
S, Lippa
C. Diagnosis and prognosis in idiopathic normal pressure hydrocephalus.
Am J Alzheimers Dis Other Demen. 2014;29(7):583–589.
[PubMed: 24550545]
+
Irie
R, Tsuruta
K, Hori
M, Suzuki
M, Kamagata
K, Nakanishi
A,
et al. Neurite Orientation dispersion and density imaging for evaluation of corticospinal tract in idiopathic normal pressure hydrocephalus.
Jpn J Radiol. 2017;35:25–30.
[PubMed: 27787666]
+
Kazui
H, Mori
E, Ohkawa
S, Okada
T, Kondo
T, Sakakibara
R, Ueki
O, Nishio
Y, Ishii
K, Kawaguchi
T, Ishikawa
M, Takeda
M. Predictors of the disappearance of triad symptoms in patients with idiopathic normal pressure hydrocephalus after shunt surgery.
J Neurol Sci. 2013;328(1–2):64–69.
[PubMed: 23510566]
+
Klasseb
BT, Alskhog
JE. Normal Pressure Hydrocephalus: How Often Does the Diagnosis Hold Water. Neurology 2011;77;1119.
[PubMed: 21849644]
+
Oliveira
MF de, Reis
RC, Trindade
EM, Pinto
FCG. Evidences in the treatment of idiopathic normal pressure hydrocephalus.
Rev Assoc Med Bras. 2015;61(3):258–262.
[PubMed: 26248249]
+
Shprecher
D, Schwalb
J, Kurlan
R. Normal Pressure Hydrocephalus: Diagnosis and Treatment.
Curr Neurol Neurosci Rep. 2008;8:371–376.
[PubMed: 18713572]
+
Tawfik
A, Elsorogy
L, Abdelghaffar
R, Abdel
Naby A, Elmenshawi
I. Phase-Contrast MRI CSF Flow Measurements for the Diagnosis of Normal-Pressure Hydrocephalus: Observer Agreement of Velocity Versus Volume Parameters. Am J Roentgenology. 2016;208:1–6.
+
Tuppeny
M. Normal-pressure hydrocephalus: The "wet, wobbly, and wacky" triad. Am Nurse Today. 2014;9(5):50–63.
+
Verrees
M, Selman
WR. Management of normal pressure hydrocephalus.
Am Fam Physician. 2004;70:1071–1090.
[PubMed: 15456116]
+
Williams
MA, Rigamonti
D. Normal pressure hydrocephalus. In: Johnson
RT, Griffin
JW, McArthur
JC. Current Therapy in Neurologic Disease. 7th ed. Philadelphia, PA: Mosby; 2006: 336.
+
Yang
J, Shanahan
KJ, Shriver
LP, Luciano
MG. Exercise-induced changes of cerebrospinal fluid vascular endothelial growth factor in adult chronic hydrocephalus patients.
J Clin Neurosci. 2016;24:52–56.
[PubMed: 26498093]
+++
Osteogenesis Imperfecta
++
Osteogeneis imperfecta (OI), also known as brittle bone disease, is an inherited disorder of connective tissue characterized by diffuse osteoporosis, bone fragility and fractures that may be accompanied by boney deformity. Severity and clinical manifestations range widely (van Dijk, 2010; Monti, 2010; Donohoe, 2017).
Genetic expression is variable but about 90% of individuals with OI have autosomal dominant mutations in the type I collagen genes COL1A1 and COL1A2; mutations cause either less collagen than normal, or poorer quality collagen to be produced during embryonic development (Ben Amor, 2013; Monti, 2010; Rauch, 2004).
Incidence of OI in the United States is between 1/15,000 and 1/60,000 births (Greeley, 2013).
Pathophysiology (Donohoe, 2017; Rauch, 2004; Culav, 1999; Glorieux, 2008; Eyre, 2013):
Increased osteoblast activity, but failure to produce and properly organize collagen, reduced rate of osteogenesis, and increased bone resorption; collagen fibers do not mature beyond the reticular fiber stage
Abnormal formation of both chondral and intramembranous bone
Slow periosteal bone formation and abnormally thin trabeculae
Classification:
Sillence (1979) identified four types of OI; not ordered by severity; most autosomal dominant inheritance; defect in type I collagen structure (mutation in genes COLIAI and COLIA2 located on chromosomes 7 and 17)
Type I: 50% of OI population; mildest type; normal collagen, but decreased production; near normal stature; bone deformity variable; lax joints; 10% have fractures at birth; blue sclerae; pre-senile conductive hearing loss; variable dentinogenesis imperfecta; average life expectancy (Monti, 2010; Donohoe, 2017)
Type II: most severe form; perinatal type; infants can be stillborn or die shortly after birth; severe bone deformities; severely delayed ossification of skull and facial bones; underdeveloped lungs; surviving patients have chronic respiratory problems and require supplemental oxygen (Monti, 2010; Donohoe, 2017).
Type III: usually autosomal dominant but sometimes recessive; more severe than type I and the most severe type in children who survive the neonatal period; progressive rib cage, spine and long bone deformities (often severe); skull fragility; very short stature; dozens to several hundreds of fractures in lifetime; respiratory problems can be fatal; near average life expectancy; variable blue sclerae that tends to lessen with age; dentinogenesis imperfecta in 45%; hearing loss common; lax joints and poor muscle development in arms and legs; kyphoscoliosis predisposes to life-threatening respiratory complications and possible death in childhood; (Monti, 2010; Donohoe, 2017).
Type IV: severity in between Types I and III; frequent fractures; mild to moderate bone deformities; average life expectancy; barrel shaped rib-cage; dentinogenesis imperfecta common; hearing loss variable (Monti, 2010; Fahiminiya, 2013; Donohoe, 2017).
Additional types (7) identified based primarily on genetic, histologic, biochemical and radiologic findings; do not have type I collagen defect but are clinically similar (van Dijk, 2010; Christiansen, 2010; Donohoe, 2017)
++
Clinical manifestations vary greatly among individuals, even those diagnosed with the same type of OI (Venturi, 2006).
Signs and symptoms: variable in severity and type (Donohoe, 2012; Engelbert, 2000; Monti, 2010; Rauch, 2004; Nicolaou, 2012).
Brittle/fragile bones resulting in frequent fractures (with no significant trauma) that usually decrease during or after puberty but may produce significant boney deformity
Varying degrees of short stature; usually of normal height at birth; cause may be combination of boney deformity and impairments in epiphyseal plate
Measurable hearing loss is evident in 50% of adults with OI, typically starting in the 2nd to 3rd decade; cause may be conductive or sensorineural; may be structural abnormalities of bones in inner ear; may have tinnitus and vertigo; treatment includes hearing aids and/or stapedectomy or cochlear implant surgery (Osteogenesis Imperfecta Foundation, 2015b)
Blue sclera (typically with no related visual deficit)
Dentinogenesis imperfecta in about 50%; abnormal growth of jaw and teeth; impacted teeth; malocclusion (Osteogenesis Imperfecta Foundation, 2015b)
Increased risk of neural compromise (thin skull) and cardiopulmonary compromise (osteoporotic ribs).
Joint hypomobility, excessive coxa vara (Brizola, 2014)
Spinal curvature – scoliosis and kyphoscoliosis
Muscle weakness; poor muscle development
Social-emotional-behavioral consequences for patient and family, including fear, anxiety, social isolation, decreased infant/parent bonding
Diagnostic testing (Donohoe, 2017; Glorieux, 2008; Swinnen, 2012; Byers, 2006; Rauch, 2010)
Prenatal
Chorionic villous biopsy or amniocentesis as early as 10 weeks gestation (if family history places fetus at risk and genetic defect is known)
Ultrasound during 2nd trimester may identify in utero fractures, deformities and decreased bone mineral density (BMD)
Postnatal
Bone fractures occurring with little or no trauma
Skin biopsy for collagen and biochemical studies
DNA sequencing from blood sample
Bone biopsy
Dual energy X-ray absorptiometry (DEXA) scan to examine bone mineral density (Donohoe, 2012; Swinnen,2012).
Other clinical manifestations common to the disorder
Differential diagnosis between non-accidental injuries (i.e., child abuse) and OI fractures warrants evaluation/sensitivity; fracture of epiphysis rare in OI (Byers, 2006; D'Eufemia, 2012; Donohoe, 2017).
++
Some types are progressive; dependent upon type and severity
Assessment of clinical severity – scoring system (Aglan, 2012)
Expected Sequelae:
Age of gait acquisition associated with overall joint range of motion and inverse association with overall muscle strength; variable with severity (type) of OI; almost all children with type I OI acquired unsupported gait by 18 months of age; delayed gait in 45% of children with type IV, 100% of children with type II and 16.1% with type 1 (Brizola, 2014; Blow, 2014).
Malunion, bowing, and angulation of fractures due to abnormal callus formation
In a study of 86 patients with type I OI (36 male, 50 female; mean age 13.3 years; range, 0.6 to 54 years) with COL1A1 haploinsufficiency mutations (Ben Amor, 2013):
Birth history of long bone fracture or deformity in 12%
Average rate of long-bone fracture was 0.62 per year (50% affecting tibia/fibula); negatively associated with age and lumbar spine areal bone mineral density; vertebral compression fractures in 71% of the 58 patients age 21 years or less; median number of vertebral fractures higher for females than males; scoliosis present in about 30% of pediatric patients (all with Cobb angle <30 degrees); shortened stature (mean 8 to 10 cm below general population
Progressive spinal deformities secondary to vertebral compression fractures, osteoporosis, ligamentous laxity and boney deformity; incidence of scoliosis or kyphosis in adolescents and adult with severe OI 80-90% (Engelbert, 2003).
Development of thoracic insufficiency syndrome (TIS), resulting in chest collapsing- Type III (Kaplan, 2013)
Rib fractures contribute to pulmonary dysfunction; skull fractures may compromise neural function
Significant reduction of muscle strength and exercise tolerance, fatigue, motor delay (Takken, 2004)
Difficulty ambulating due to frequent fractures or bone deformities
Respiratory compromise most common cause of early death (Monti, 2010).
+++
IV. Medical/Surgical Management
++
+++
V. Therapeutic Management
++
Multidisciplinary approach essential – increase function, promote independence, prevent long term problems as much as possible, move into community-based physical activity and social involvement
Physical Therapy Management (Donohoe, 2017; Monti, 2010):
Evaluation:
Brief Assessment of Motor Function – gross motor, fine motor and oral motor function; can help predict ambulatory potential (Cintas, 2003)
Joint range of motion (active not passive), strength, motor function
10-meter Shuttle Ride Test (SRiT) a feasible, safe and reproducible measure of cardiovascular fitness for children with OI who self-propel a wheelchair (Bongers, 2016)
Pediatric Evaluation of Disability Inventory useful in contributing to a functional disability profile in children with OI (Engelbert, 1997)
School Function Assessment; Peabody Developmental Motor Scales – 2; Bayley Scales of Infant Toddler Development III
FLACC Behavioral Pain Assessment Scale (Merkel, 1997) for quantitative assessment of pain in preverbal children ages 2 months to 7 years; scored 0-2 for each of 5 categories: face, legs, activity, cry, consolability; a copy of scale: http://wps.prenhall.com/wps/media/objects/3103/3178396/tools/flacc.pdf
Intervention:
Caregiver education including safe handling and positioning techniques
In general, want to distribute pressure across as large an area as possible (e.g., may support infant on a pillow) and avoid placing extremities where they are vulnerable to accidental bump or pressure
Provide safe options for supine, prone, sidelying, and supported sitting positions using pillows, wedges, foam, rolls, etc.
Use caution when changing diaper (roll onto diaper… not lift by legs); when removing from car seat (may need to use car bed); when pulling to sit (support shoulders – not pull up by hands/arms); when dressing (loose clothing, velcro, snaps)
Instruct in safe motor control strategies for transitioning from one position to another (e.g., supine to sit)
Anticipate potential dangers in environment; do not use walkers, jumpers or other similar devices
Prevent osteoporosis: early weight bearing and increased mobility- aquatics or land
Intervention for developmental delay: comfortable play positions (rolling, supported sitting), facilitation of motor control strategies, and appropriate toys or assistive technology.
Intervention for prevention/management of fractures and contractures, promotion of functional mobility, strengthening, balance: splints, elastic wraps, orthoses, assistive technology; and compensatory strategies
Post-operative intervention: aquatic therapy recommended; strengthening of limbs not involved in surgery (Osteogenesis Imperfecta Foundation, 2015)
Intervention for scoliosis/kyphosis: fitting for appropriate wheelchairs, seating and mobility devices; referral for medical/surgical management
Assistive technology evaluation, recommendation, procurement and management; positioning, assistive gait devices, mobility, seating systems, etc.
Environmental adaptation
Intervention for decreased strength and endurance: progressive resistance program; functional strengthening; aquatic exercise
Whole body vibration (WBV) - a systematic review and meta-analysis revealed some evidence to support small but significant increases in bone mineral density in some joints of postmenopausal women, children and adolescents (Slatkovska, 2010) but disappointing outcomes in a pilot study using WBV in children with OI (increase in total lean mass without changes in muscle function or bone mass) suggesting a different biomechanical responsiveness of the muscle-bone unit in children with OI (Hogler, 2017)
Pain management support/monitoring
Health promotion including weight management, nutrition, physical activity, peer engagement
Resource from Osteogenesis Imperfecta Foundation (2017) with contributions from multiple therapists, Physical and Occupational Therapists' Guide to Treating Osteogenesis Imperfecta, offers practical guidance on a broad array of topics: http://www.oif.org/site/DocServer/PT_guide_final.pdf?docID=201
+
Aglan
MS, Hosny
L, El-Houssini
R,
et al. A scoring system for the assessment of clinical severity in osteogenesis imperfecta.
J Child Orthop. 2012;6(1):29–35.
[PubMed: 23449141]
+
Ben Amor
IM, Roughley
P, Glorieux
FH, Rauch
F. Skeletal clinical characteristics of osteogenesis imperfecta caused by haploinsufficiency mutations in COL1A1.
J Bone Miner Res. 2013;28(9):2001–2007.
[PubMed: 23529829]
+
Besio
R, Forlino
A. New frontiers for dominant osteogenesis imperfecta treatment: gene/cellular therapy approaches.
Advances Regen Bio. 2015;2(1):27964.
http://dx.doi.org/10.3402/arb.v2.27964. Accessed October 26, 2017.
+
Bishop
N, Adami
S, Ahmed
SF,
et al. Risedronate in children with osteogenesis imperfecta: a randomised, double-blind, placebo-controlled trial.
Lancet. 2013;382(9902):1424–1432.
[PubMed: 23927913]
+
Blow
CA, McElroy
JJ. Commentary on "Muscle strength, joint range of motion, and gait in children and adolescents with osteogenesis imperfecta."
Pediatr Phys Ther. 2014;26:252.
[PubMed: 24675131]
+
Bongers
B, Rijks
E, Harsevoort
A, Takken
T. 10-m shuttle ride test in youth with osteogeneis imperfecta who use wheelchairs: feasibility, reproducibility, and physiological response.
Phys Ther. 2016;96(5):679–686.
[PubMed: 26494769]
+
Brizola
E, Staub
ALP, Felix
TM. Muscle strength, joint range of motion, and gait in children and adolescents with osteogenesis imperfecta.
Pediatr Phys Ther. 2014;26:245–252.
[PubMed: 24675130]
+
Byers
PH, Krakow
D, Nunes
ME, Pepin
M. Genetic evaluation of suspected osteogenesis imperfecta (OI).
Genet Med. 2006;8(6):383–388.
[PubMed: 16778601]
+
Caudill
A, Flanagan
A, Hassani
S,
et al. Ankle strength and functional limitations in children and adolescents with type I osteogenesis imperfecta.
Pediatr Phys Ther. 2010;22(3):288–295.
[PubMed: 20699778]
+
Chagas
CE, Roque
JP, Santaros
EPB, Lazaretti-Castro
M, Martini
LA. Do patients with osteogenesis imperfecta need individualized nutritional support?.
Nutrition. 2012;28(2):138–142.
[PubMed: 21795023]
+
Christiansen
HE, Schwarze
U, Pyott
SM,
et al. Homozygosity for a missense mutation in SERPINH1, which encodes the collagen chaperone protein HSP47, results in severe recessive osteogenesis imperfecta.
Am J Hum Genet. 2010;86(3):389–398.
[PubMed: 20188343]
+
Cintas
HL, Siegel
KL, Furst
GP, Gerber
LH. Brief assessment of motor function: reliability and concurrent validity of the Gross Motor Scale.
Am J Phys Med Rehabil. 2003;82:33–41.
[PubMed: 12510183]
+
Culav
EM, Clark
CH, Merrilees
MJ. Connective tissues: matrix composition and its relevance to physical therapy.
Phys Ther. 1999;79(3):308–319.
[PubMed: 10078774]
+
D'Eufemia
P, Palombaro
M, Lodato
V, Zambrano
A, Celli
M, Persiani
P, De Bari
MP, Sangiorgi
L. Child abuse and osteogenesis imperfecta: how can they be still misdiagnosed? A case report. Clin Cases Miner Bone Metab. 2012 Sep;9(3):195–7.
+
Donohoe
M. Osteogenesis Imperfecta. In: Palisano
RJ, Orlin
MN, Schreiber
J. Campbell's Physical Therapy for Children. 5th ed. St Louis, MO: Elsevier. 2017: 224–241.
+
Donohoe
M, Cintas
H. Commentary on "Ankle strength and functional limitations in children and adolescents with type I osteogenesis imperfecta."
Pediatr Phys Ther. 2010;22(3):295.
[PubMed: 20699779]
+
Engelbert
RH, Custers
JWH, van der Net
J, van der Graaf
Y, Beemer
FA, Helders
PJM. Functional outcome in osteogenesis imperfecta: disability profiles using the PEDI. Pediatr Phys Ther. 1997;9:18–22.
+
Engelbert
RH, Uiterwaal
C, Gerver
W,
et al. Osteogenesis imperfecta in childhood: impairment and disability. A prospective study with 4-year follow-up.
Arch Phys Med Rehabil. 2004;85:772–778.
[PubMed: 15129402]
+
Engelbert
RH, Uiterwaal
C, Gulmans
V,
et al. Osteogenesis imperfecta in childhood: prognosis for walking.
J Pediatr. 2000;137:397–402.
[PubMed: 10969267]
+
Engelbert
RH, Uiterwaal
CS, van der Hulst
A, Witjes
B, Helders
PI, Pruijs
HE: Scoliosis in children with osteogenesis imperfecta: influence of severity of disease and age of reaching motor milestones.
Eur Spine J. 2003;12:130–134.
[PubMed: 12709850]
+
Eyre
DR, Weis
MA. Bone Collagen: New clues to its mineralization mechanism from recessive osteogenesis imperfecta.
Calcif Tissue Int. 2013;93(4):338–347.
[PubMed: 23508630]
+
Falk
M, Heeger
S, Lynch
K,
et al. Intravenous biphosphonate therapy in children with osteogenesis imperfecta. Pediatr. 2003;111(3):573–578.
+
Fahiminiya
S, Majewski
J, Mort
J, Moffatt
P, Glorieux
FH, Rauch
F. Mutations in WNT1 are a cause of osteogenesis imperfecta.
J Med Genet. 2013;50(5):345–348.
[PubMed: 23434763]
+
Glorieux
FH. Osteogenesis imperfecta.
Best Pract Res Clin Rheumatol. 2008;22(1):85–100.
[PubMed: 18328983]
+
Greeley
CS, Donaruma-Kwoh
M, Vettimattam
M, Lobo
C, Williard
C, Mazur
L. Fractures at diagnosis in infants and children with osteogenesis imperfecta. J Pediatr Orthop. 2013 Jan;33(1):32–6.
+
Hogler
W, Scott
J, Bishop
N,
et al. The effect of whole body vibration training on bone and muscle function in children with osteogenesis.
J Clin Endocrinol Metab. 2017;102(8):2734–2743.
[PubMed: 28472303]
+
Kaplan
L, Barzilay
Y, Hashroni
A, Itshayek
E, Schroeder
J. Thoracic elongation in Type III Osteogenesis Imperfecta patients with thoracic insufficiency syndrome.
Spine. 2013; 38(2):94–100.
[PubMed: 23242110]
+
Liu
Y, Wu
J, Zhu
Y, Han
J. Therapeutic application of mesenchymal stem cells in bone and joint disease.
Clin Exp Med. 2012; 14(1):13–24.
[PubMed: 23124706]
+
Merkel
SI, Voepel-Lewis
T, Shayevitz
JR, Malviya
S. The FLACC: A behavioral scale for scoring postoperative pain in young children.
Pediatr Nurs. 1997;23(3):293–297.
[PubMed: 9220806]
+
Monti
E, Mottes
M, Fraschini
P,
et al. Current and emerging treatments for the management of osteogenesis imperfecta. Ther Clin Risk Manage. 2010;6:367–381.
+
Nicolaou
N, Agrawal
Y, Padman
M, Fernandes
JA, Bell
MJ. Changing pattern of femoral fractures in osteogenesis imperfect with prolonged use of bisphosphonates.
J Child Orthop. 2012;6(1):21–7.
[PubMed: 23450103]
+
Plante
L, Veilleux
LN, Glorieux
FH, Weiler
H, Rauch
F. Effect of high-dose vitamin D supplementation on bone density in youth with osteogenesis imperfecta: a randomized controlled trial.
Bone. 2016;86:36–42.
[PubMed: 26924265]
+
Rauch
F, Glorieux
F. Osteogenesis imperfecta.
Lancet. 2004;363:1377–1385.
[PubMed: 15110498]
+
Rauch
F, Lalic
L, Roughley
P, Glorieux
FH. Relationship between genotype and skeletal phenotype in children and adolescents with osteogenesis imperfecta.
J Bone Miner Res. 2010;25(6):1367–1374.
[PubMed: 19929435]
+
Russell
RGG. Bisphosphonates: mode of action and pharmacology. Pediatr. 2007;119 Suppl 2:S150–162.
+
Sillence
DO, Senn
A, Danks
DM. Genetic heterogeneity in osteogenesis imperfecta.
J Med Genet. 1979;16:101–116.
[PubMed: 458828]
+
Slatkovska
L, Alibhai
SM, Beyene
J, Cheung
AM. Effect of whole-body vibration on BMD: a systematic review and meta-analysis.
Osteoporos Int. 2010;21(12):1969–1980.
[PubMed: 20407890]
+
Swinnen
FKR, De Leenheer
EMR, Goemaere
S, Cremers
CWRJ, Coucke
PJ, Dhooge
IJM. Association between bone mineral density and hearing loss in osteogenesis imperfecta.
Laryngoscope. 2012;122(2):401–408.
[PubMed: 22252604]
+
Takken
T, Terlingen
H, Helders
P,
et al. Cardiopulmonary fitness and muscle strength in patients with osteogenesis imperfecta type I.
J Pediatr. 2004;145:813–818.
[PubMed: 15580207]
+
van Dijk
FS, Pals
G, Van Rijn
RR, Nikkels
PGJ, Cobben
JM. Classification of osteogenesis imperfecta revisited.
Eur J Med Genet. 2010;53(1):1–5.
[PubMed: 19878741]
+
Venturi
G, Tedeschi
E, Mottes
M,
et al. Osteogenesis imperfecta: clinical, biochemical and molecular findings.
Clin Genet. 2006;70(2):131–139.
[PubMed: 16879195]
+
Ward
LM, Rauch
F, Whyte
MP,
et al. Alendronate for the treatment of pediatric osteogenesis imperfecta: a randomized placebo-controlled study. J Clin Endocrinol Metab. 2011 Feb;96(2):355–64.
++
Incidence and Prevalence
Most common movement disorder and second most common neurodegenerative disorder of aging (Mhyre, 2012)
60,000 Americans diagnosed per year; > 1 million Americans have Parkinson's Disease (PD) which is more than the combined number of people with MS, Muscular Dystrophy, and ALS (Parkinsons Disease Foundation, 2017).
Men are 1.5 times more likely to develop PD than women (Taylor, 2007).
PD is more common in the elderly population; diagnosis peaks at ages 60-69 and is less prevalent in ages under 50
There is a steady increase in prevalence of PD with age: 41 per 100,00 in individuals 40 to 49 years versus 1,087 per 100,000 in individuals 70 to 79 years (Pringsheim, 2014)
Risk Factors
Evidence for exposure to pesticides and the environmental endotoxin, lipopolysaccharide produced Salmonella minnesota (Allam, 2005). People with Parkinson disease are significantly more likely to report direct pesticide application and higher cumulative exposure (Hancock, 2008). The intensity of exposure to pesticides is more dangerous than duration of pesticide exposure. (Moisan, 2015)
Other risk factors include a history of being knocked unconscious (exposure-response relationship), use of hyponotic, anxiolytic, or antidepressant for > 1 year, and family history of Parkinson disease (Dick, 2007)
Traumatic Brain Injury: Among patients aged > 55 presenting to an emergency department or inpatient setting, a TBI results in a 44% risk of PD compared to a trauma of the rest of the body over a follow-up of just 5 to 7 years (Gardner, 2015). Head injuries have been linked the diagnosis of PD. The earlier in life a head injury occurs the higher the risk of being diagnosed with PD. (Taylor, 2016).
Pathophysiology:
~90% of cases of Parkinson's disease are sporadic, arising from unknown causes
The remaining ~10% of PD cases are caused by genetic factors:
Mutations in the alpha-synuclein, Parkin, PINK, LRRK2, and other genes. (Agim, 2015)
Significant evidence for linkage -- chromosome 18q11 (maximum lod score [MLOD] = 4.1) and suggestive evidence --chromosome 3q25 (MLOD = 2.5) (Gao, 2009).
Only 5-10% of cases are caused by the mutation of several specific genes, which include α-synuclein (SNCA), parkin (PRKN), leucin-rich repeat kinase 2 (LRRK2), PTEN-induced putative kinase 1 (PINK1), DJ-1 and ATP13A2 (Chen, 2015).
Damage to nigrostriatal pathway
Degeneration of dopamine neurons within the pars compacta of the substantia nigra and attendant projectios to the putamen leading to dopamine depletion in striatum and characteristic motor symptoms (Dickson, 2012, Benazzouz, 2014)
Characterized by accumulation of intracellular protein aggregates composed of protein α-synuclein (called Lewy bodies and neurites) (Dickson, 2012)
Parkinsonism:
Parkinson's Disease-like symptoms, usually termed parkinsonism
Can also be components of syndromes that share some or most of the signs of PD plus other signs or symptoms that are not characteristic of PD
+++
II. Diagnostic procedures
++
No single definitive test. Usually diagnosed by history and clinical exam.
The clinical criteria for diagnosing PD are Tremor, Rigidity, Akinesia, Postural Instability (TRAP) (Frank, 2006)
A clinical diagnosis of PD requires the following: distal resting tremor of 3-6 Hz, rigidity, bradykinesia, and asymmetrical onset. Also, "patients diagnosed with PD must respond to an adequate therapeutic levodopa or a dopamine agonist challenge. The clinical diagnosis is further supported by progressive functional and motor impairment" (Nolden, 2014)
Diagnosis requires 2 of 4 cardinal features. (Turnbull,1992)
-
Tremor (4-5 Hz): typically, unilateral and at rest; in forearm, hand, fingers, or foot (classic "pill-rolling") (Mhyre, 2012)
Rigidity: increased muscle tone with resistance to passive stretch (Mhyre, 2012)
Akinesia (hypokinesia and bradykinesia):
Hypokinesia: decreased bodily movements; including decreased blinking and facial expression rate, reduced arm swing while walking, and absence of associated movements when rising from chair or bed (Mhyre, 2012)
Bradykinesia: slowed bodily movements; including slowed alternating movements such as finger tapping, making a fist, hand dexterity, and gait (Mhyre, 2012)
Postural instability/impaired balance or coordination
Other Signs and Symptoms (Parkinsons Disease Foundation, 2017; Turnbull, 1992)
Characteristic flexed trunk and knees, patient appears "stuck," difficulty initiating movement
Loss of facial expression or "masking"
Dementia or confusion, memory difficulties, slowed thinking, compulsive behavior
Speech changes, difficulty swallowing, , drooling
Dysphagia usually occurs later in the disease process, while dysarthria may be an early symptom (Goroll, 2009).
Swallowing dysfunction emerges in early PD (Ciucci, 2013) and can increase the risk of aspiration, pneumonia, and mortality (Cereda, 2014).
Others:
Pain, aching, constipation, sexual dysfunction, urinary problems, fatigue and loss of energy, micrographia (small, cramped handwriting)
Olfactory disturbances including hyposmia and decreased odor detection, identification, and discrimination may also be present (Mhyre, 2012)
Olfactory deficit, constipation, rapid-eye movement sleep behavior disorder, and depression, can precede the appearance of motor symptoms by many years (Valkovic, 2014)
Gait varies with stage of disease (Turnbull, 1992)
Early:
Later:
"Typical" pattern of shuffling gait with little or no UE or trunk associated movements- diminished arm swing, overall flexion when walking, head projects abnormally anteriorly, kyphotic thoracic spine, arms and legs assume a flexed and adducted posture, increasingly shorter steps, slow walking speed, reduction in spontaneous movement.
Balance
Twice as likely to fall as other older persons (Canning, 2009). Involved upper limb is slightly flexed at elbow, wrist, and fingers as flexor tone predominates (Mhyre, 2012).
Nonmotor symptoms in PD can be divided into four categories (Stacy, 2011)
1) Cortical manifestations, such as dementia, mild cognitive impairment, and psychosis
2) Basal ganglia symptoms, such as apathy, restlessness, and impulse control disorders
3) Brainstem linked symptoms, such as depression, anxiety, and sleep disorders
4) Peripheral nervous system disturbances, such as orthostatic hypotension, constipation, pain, and sensory disturbance
Depression, apathy, sleep disturbance, and anxiety are most common early neuropsychiatric symptoms and probably worsened throughout the disease due to the physiological changes in the brain and secondary stress. (Aarsland, 2009).
Diagnostic testing
Early diagnosis lab tests and neuroimaging usually normal
Genetic testing, olfactory testing, transcranial sonography, magnetic resonance imaging, computed tomography scanning, and DaTscan SPECT (Berardelli, 2012)
Example: UK Parkinson's Disease Society Brain Bank Clinical Diagnostic Criteria (Hughes, 1992)
Genetic testing, olfactory testing, transcranial sonography, magnetic resonance imaging, computed tomography scanning, and DaTscan SPECT (Berardelli, 2012)
Olfactory screening: "Odor identification testing provides excellent diagnostic accuracy in the distinction of PD patients from controls and diagnostic mimic. A reduced set of eight odors could be used as a quick tool in the workup of patients presenting with parkinsonism and for PD risk indication". (Mahlknecht, 2016).
++
Progressive disease: The progression of PD greatly impacts prognosis, specifically when dealing with an increased inability to perform daily activities, loss of independence, and a decreased quality of life (Carvalho, 2015).
Scales used to document progression:
Hoehn-Yahr Classification of Disability Scale: categorized by character of disability; (Hoehn and Yahr, 1967)
Stage 1 – minimal or absent signs; unilateral if present with minimal or no functional disability
Stage 2- minimal bilateral or midline involvement, balance not impaired
Stage 3 – impaired righting reflexes, unsteadiness when turning or rising from chair, some activities restricted, but can live independently and continue some forms of employment,
Stage 4 - all symptoms present and severe, standing and walking possible only with assistance
Stage 5 - confined to bed or WC
Schwab and England scale, Unified Parkinson's Disease Rating Scale (Sage, 2009), Scales for Outcomes in Parkinson's Disease-Motor Scale, Clinical Global Impression of Severity (CGIS), Clinical Impression of Severity Index for Parkinson's Disease (CISI-PD)
Based on UPDRS scores, people may be classified as having PD that is Postural Instability and Gait Disorder predominant (PIGD), tremor predominant, or mixed.
Compared with patients who have the PIGD subtype, patients with the tremor predominant subtype experience: slower disease progression, longer survival, and less cognitive impairment (Helmich, 2012)
Higher scores on activity of daily living and motor complications in the UPDRS were significantly and independently associated with falling (Hiorth, 2013)
Greater baseline impairment, early cognitive disturbance, older age, and lack of tremor at onset appear to be adverse prognostic factors (Marras, 2002)
Saccadic latency reflects central decision-making operation and is used to quantify general cerebral performance (Temel, 2009).
Patients who have an earlier onset of symptoms (age 50 or before) have a more favorable prognosis and show a longer duration period before reaching stage 3, 4, and 5 Hoehn-Yahr (Sato, 2006).
Patients who are tremor-dominant show a significantly longer duration period to reach stage 3 Hoehn-Yahr as compared to patients with gait-disturbance onsets (Sato, 2006).
+++
IV. Medical and surgical management
++
Medical
Monamine oxidase-B inhibitor (MAOB): Selegiline; used early, delays primary endpoint for taking levadopa by about 9 months; may slow normal progression of disease (Rasagiline (Singer, 2012). Rasagiline doses of 1 mg per day decreased levels of fatigue, which affects 40%-50% of all PD patients. (Lim, 2015).
Dopamine replacement therapy: Levodopa; "gold standard" as treatment for symptomatic treatment
Side effects: on/off effect, dystonia, abnormal, involuntary movement, nausea, vomiting, confusion, hallucinations (Clarke, 2006; Widnell, 2005)
Drug holidays (weaning off medications) not recommended but may be used for l-dopa induced psychiatric toxicity (Parkinsons Disease Foundation, 2006).
"People in assisted living that have been diagnosed with PD are more likely to be taking significantly lower doses of levodopa and had more advanced stages of PD most likely due to the requirement to balance symptom management with drug side-effects" (Hand, 2016)
COMT inhibitors: Tolcapone, Entacapone; adjunct to levodopa; improves wearing off, number of doses and dosage reduction of l-dopa, reduces off time with a corresponding increase in on time (Widnell, 2005)
Dopamine agonists: adjunct, dosage reducer and enhancer to l-dopa, reduces motor fluctuations; apomorphine (Apokyn)-used as a rescue drug for sudden "off time," bromocriptine (Parlodel), cabergoline (Dostinex), pergolide (Permax), pramipexole (Mirapex), rotigotine (Nuepro), ropinirole (Requip) (Widnell, 2005; Shin, 2013). Pardoprunox (partial dopamine agonist) safety and tolerability profile improves with a gradual titration schedule with intermediate dose steps. Doses up to 18 mg/day well tolerated(Hauser,2009).
Botulinum toxin type-B (BTX-B) injections into the parotid glands to reduce drooling (Lagalla, 2009).
Amantadine: may reduce dyskinesias in later stages; may cause greater amounts of dopamine to be released in the brain as well as blocking acetylcholine receptors in the brain which may help to control movement (Singer, 2012); side effects: nausea, constipation, dizziness, insomnia, depression, anxiety, somnolence, hallucinations, and symptomatic orthostatic hypotension (Shin, 2013).
Anticholinergics: Biperiden, Orphenadrine, Diphenhydramine, Procyclidine, Trihexyphenidyl, Benzotropine; oral medications which are competitive antagonists of muscarinic receptors; may reduce imbalance between dopaminergic and cholinergic neurological pathways, reducing involuntary resting tremor (Brocks, 1999)
Irisflorentin is an herb that is derived from the roots of Belamcanda chinensis. It has shown recent potential to become an anti-Parkinsonian drug candidate by preventing α-synuclein accumulation and improving dopaminergic neuron degeneration, food-sensing behavior, and life span in a 6-hydroxydopamine-induced Caenorhabditis elegans model (Chen, 2015).
American Geriatrics Society (AGS) Beers Criteria recommends avoiding all antipsychotics (except quetiapine and clozapine) and some antiemetics (metoclopramide, prochlorperazine, and promethazine) to avoid worsening of parkinsonian symptoms (AGS, 2015)
Surgical
Pallidotomy: lesion in basal ganglia to reduce excessive inhibitory activity and results in hypoactivity; reduces tremor, dyskinesias, motor fluctuations; improves bradykinesia and functional performance (Follett, 2000); probing and destruction of the globus pallidus (Singh, 2007)
Thalamotomy: lesion made to destroy thalamus to reduce tremor, rigidity and dyskinesias (Follett, 2005); but now rarely performed (Singh, 2007)
Deep brain stimulation (DBS): deeply implanted electrodes the most commonly, subthalamic nucleus (Temel, 2009) and the pallidum; produces reversible functional lesion adjustable to patient needs (Follett, 2000).
Thalamic stimulation: involves inserting electrode into thalamus to produce benefits of thalamotomy without incising the skin while treating tremor (Singh, 2007)
Transplantation of fetal substantia nigra tissue into the striatum has been receiving attention as a potential option. (Politis, 2012).
DUOPA is an enteral suspension of carbidopa/levodopa delivered the small intestine by way of a tube in the stomach. It is designed to improve absorption and reduce off-times by delivering the drug directly to the small intestine (Parkinson's Disease Foundation, 2017)
Non-pharmacological treatments: still under investigation: include PD Vaccine and cell transplantation (Singh, 2007)
+++
V. Preview of therapeutic management
++
Preferred Practice Pattern
Physical rehabilitation can be a major factor in helping maintain and improve mobility, posture, and balance. Interventions such as physiotherapy, walking, running, strength training, functional exercises, and whole body vibration have been shown to significantly reduce the risk of falls and improve motor performance, balance, gait, and executive functions (Carvalho, 2015).
Goals of therapy: delay progression of disease, relieve symptoms, and maintain functional capabilities (Goroll, 2009).
Early intervention critical to prevent musculoskeletal impairments and individually based on impairments and functional limitations:
Relaxation exercises, flexibility exercises, mobility exercises, balance activities, motor learning strategies, functional adaptations, respiratory exercises, aerobic conditioning, strength training, group and home exercises, patient and family education. Dance intervention: Tango and walt/foxtrot targets may benefit balance and locomotion (Hackney, 2009).
General Physical Therapy Options include treatment for: transfer training, postural correction, reaching and grasping, balance and fall prevention, and gait training (Keus, 2006).
Correction of postural deficits and ROM limitations
Balance training
Addressing impairments contributing to falling should also be implemented due to the increased risk of falls seen with this population:
Tai chi training is associated with significant improvement in balance; Tai chi also shows significant improvement in gait, strength, and motor scores on Unified Parkinson Disease Rating Scale (Li, 2012)
Aerobic considerations
Gait Training
Dual task gait training: Practice prioritizing focus on certain aspects of the gait cycle have produced improved walking stability and enhanced abilities in completing activities of daily living.
Treadmill training, acting as an external cue, with auditory and visual cues improves results over conventional treatments (Frazzitta, 2009).
Gait cues that focus attention on temporal and spatial parameters significantly improve single and dual-task walking speed and stride amplitude in mild PD (Rochester, 2009).
Others
Forced-rate lower extremity exercise: Researchers have found that people with Parkinson's benefit from exercise programs on stationary bicycles, with the greatest effect for those who pedal faster. Functional MRI data showed that faster pedaling led to increases in task-related connectivity between the primary motor cortex and the posterior region of the brain's thalamus - two regions of the brain associated with motor ability (Rosenfedlt, 2015)
Box training: Box training incorporates whole-body movements with upper-extremity punching motions and lower-extremity footwork in multiple directions. Other motions facilitate trunk rotation and anticipatory postural adjustments; patients of differing progressions of the disease can derive short-term and long-term improvements in balance, gait, ADLs, and quality of life with a box training program. (Combs, 2011)
Sensory-based exercise (PD SAFEx) -- improvement in symptoms and functional movement control (Sage, 2009).
The use of music combined with rhythmic movements and the use of Wii-Nintendo patterned movement games have also been successful training tools in dual task activities for individuals with Parkinson's disease (Ford, 2015)
Whole body vibration intervention--research at present shows placebo effect only (Arias, 2009).
Deep Brain Stimulation (DBS) is well established and has shown significant benefits for the treatment of PD when delivered to the subthalamic nucleus and globus pallidus internus. Neurosurgeons, movement disorder neurologists, nurses, occupational therapists, and physical therapists can all perform DBS programming. It has been shown to have a therapeutic effect on patients with PD, which includes decreasing the total kinesia motor score and improving bradykinesia and tremors (Pulliam, 2015).
Low frequency repetitive transcranial magnetic stimulation (rTMS): a noninvasive procedure that delivers repeated magnetic pulses to a specific brain area within a short time affecting the excitability of the stimulation site and the immediate surrounding areas; rTMS has the ability to produce changes in neural activity and behavior that last well after stimulation, this technique has good potential to be an intervention for the treatment of PD (Reithler, 2011; Chou, 2015)
Resistance training generally effective in increasing strength, and in some cases mobility, but has thus far been conservatively approached.
Recent reports suggest that strength, endurance, and functional gains are greater when volume is increased in older adults. No reports exist suggesting that resistive exercise may exacerbate symptoms of PD, considerable attention must be paid to the development and management of fatigue (Falyo, 2008)
Palliative care should be discussed as an option for patients and their family, as it can be given throughout all stages of the disease, including early disease, moderate disease, advanced disease, hospice, and bereavement (Bunting-Perry, 2006).
Contraindications/Precautions include cardiac and pulmonary contraindications for strength training with maximal loading, increased risk of falling with activity, and additional consideration to patients suffering from hallucinations and other cognitive disorders (Borg, 2008)
+++
VI. Consumer and Professional Resources
++
The National Parkinson Foundation website, detailing specific chapters and support groups offering health and wellness programs, free educational material, and a toll-free helpline. http://www.parkinson.org/Improving-Care/Outreach/NPF-Chapters---Support-Groups
The American Parkinson Disease Association website offering new research, rehabilitation resources, and an area to "ask the doctor" about issues dealing with Parkinson's disease. http://www.apdaparkinson.org
The National Institute of Neurological Disorders and Stroke website listing numerous Parkinson's disease organizations all across the United States with mailing addresses, phone numbers, email addresses, and other contact information. http://www.ninds.nih.gov/disorders/parkinsons_disease/org_parkinsons_disease.htm
The Michael J. Fox Foundation for Parkinson's Research website allowing individuals to understand the disease, participate in clinical trials, and donate to future research. https://www.michaeljfox.org
The Parkinson's Disease Foundation, a leading national presence in Parkinson disease research, education, and public advocacy. https://www.pdf.org
+
Aarsland
D, Brønnick
K, Alves
G, Tysnes
OB, Pedersen
KF, Ehrt
U, Larsen
JP. The spectrum of neuropsychiatric patients symptoms in patients with early untreated Parkinsons. J Neurol Neurosurg Psychiatry. 2009 Aug;80(8):928–30.
+
Agim
ZS, Cannon
JR, Agim
ZS, Cannon
JR. Dietary Factors in the Etiology of Parkinson's Disease, Dietary Factors in the Etiology of Parkinson's Disease. BioMed Res Int BioMed Res Int. 2015;2015, 2015:e672838. doi:10.1155/2015/672838, 10.1155/2015/672838.
+
Allam
M, Del Castillo
A, Navajas
R. Parkinson's disease risk factors: Genetic, environmental, or both? Neurolo Res. 2005;27(2):206–208.
+
American Geriatrics Society. 2015 Beers Criteria Update Expert Panel. American Geriatrics Society 2015 Updated Beers Criteria for Potentially Inappropriate Medication Use in Older Adults.
J Am Geriatr Soc. 2015;63(11):2227–2246.
[PubMed: 26446832]
+
Arias
P, Chouza
M, Vivas
J, Cudeiro
J. Effect of whole body vibration in Parkinson's disease: a controlled study. Mov Disord. 2009 Apr 30;24(6):891–8.
+
Benazzouz
A, Mamad
O, Abedi
P, Bouali-Benazzouz
R, Chetrit
J. Involvement of dopamine loss in extrastriatal basal ganglia nuclei in the pathophysiology of Parkinson's disease.
Front Aging Neurosci. 2014;6:87. doi:10.3389/fnagi.2014.00087.
[PubMed: 24860498]
+
Beraradelli
A, Wenning
G, Antonini
A, Berg
D,
et al. EFNS/MDS-ES recommendations for the diagnosis of Parkinson's disease.
European Journal of Neurology. 2013;20:16–34.
[PubMed: 23279440]
+
Birchall
E, Walker
H, Cutter
G
et al.. The effect of unilateral subthalamic nucleus deep brain stimulation on depression in Parkinson's disease. Brain Stimul 2016. doi:10.1016/j.brs.2016.12.014.
+
Borg
K, Bekkelund
S, Henrisksson
M. Parkinson's Disease. Physical activity in the prevention and treatment of disease. FYSS. 2008:541–546.
+
Brocks
D. Anticholinergic drugs used in Parkinson's disease: an overlooked class of drugs from a pharmacokinetic perspective. J Pharm Pharmaceut Sci. 1999 ;2(2):39–46.
+
Braak
H
et al.. Cognitive status correlates with neuropathic stage in Parkinson disease.
Neurology. 2005; 64(8):1404–10.
[PubMed: 15851731]
+
Bunting-Perry
L. Palliative care in Parkinson's disease: Implications for neuroscience nursing.
Journal of Neuroscience Nursing. 2006;38(2):106–113.
[PubMed: 16681291]
+
Canning
CG, Sherrington
C, Lord
SR, Fung
VS, Close
JC, Latt
MD, Howard
K, Allen
NE, O'Rourke
SD, Murray
SM. Exercise therapy for prevention of falls in people with Parkinson's disease: a protocol for a randomised controlled trial and economic evaluation. BMC Neurol. 2009 Jan 22;9:4.
+
Carvalho
A, Barbirato
D, Araujo
N, Martins
JV, Cavalcanti
JLS, Santos
TM, Coutinho
ES, Laks
J, Deslandes
AC. Comparison of strength training, aerobic training, and additional physical therapy as supplementary treatments for Parkinson's disease: pilot study.
Clin Interv Aging. 2015;10:183–91.
[PubMed: 25609935]
+
Chen
YM, Liu
SP, Lin
HL, Chan
MC, Chen
YC, Huang
YL, Tsai
MC, Fu
RH. Irisflorentin improves α-synuclein accumulation and attenuates 6-OHDA-induced dopaminergic neuron degeneration, implication for Parkinson's disease therapy.
Biomedicine (Taipei). 2015;5(1):4. doi: 10.7603/s40681-015-0004-y.
[PubMed: 25705584]
+
Chou
Y, Hickey
PT, Sundman
M, Song
AW, Chen
N. Effects of Repetitive Transcranial Magnetic Stimulation on Motor Symptoms in Parkinson Disease.
JAMA Neurol. 2015;72(4):432–440.
[PubMed: 25686212]
+
Clarke
CE. Medical management of parkinson's disease. Journal of Neurology, Neurosurgery, and Psychiatry. 2002;72:122–27.
+
Combs
SA, Diehl
MD, Staples
WH, Conn
L, Davis
K, Lewis
N, Schaneman,
K. Boxing Training for Patients With Parkinson Disease: A Case Series.
Phys Ther. 2011;91(1):132–142.
[PubMed: 21088118]
+
Dick
FD, De Palma
G, Ahmadi
A, Scott
NW, Prescott
GJ, Bennet
J, Semple
S, Dick
S, Counsell
C, Mozzoni
P, Haites
N, Wettinger
S, Bezzina, Mutti
A, Otelea
M, Seaton
A, Soderkvist
P, Felice
A. Environmental risk factors for Parkinson's disease and parkinsonism: the Geoparkinson study.
Occup Environ Med. 2007;64(10):666–672.
[PubMed: 17332139]
+
Dickson
D. Parkinson's disease and parkinsonism: Neuropathology. Cold Spring Harb Perspect Med. 2012;2.
+
Frank
C, Pari
G, Rossiter
JP. Approach to diagnosis of Parkinson disease.
Can Fam Physician. 2006;52(7):862–868.
[PubMed: 16893149]
+
Frazzitta
G, Maestri
R, Uccellini
D, Bertotti
G, Abelli
P. Rehabilitation treatment of gait in patients with Parkinson's disease with freezing: a comparison between two physical therapy protocols using visual and auditory cues with or without treadmill training. Mov Disord. 2009 Jun 15;24(8):1139–43.
+
Follet
KA. The surgical treatment of parkinson's disease.
Annual Review of Medicine. 2000;51:135–147.
[PubMed: 10774457]
+
Ford
MM, Howell
JB, Moore
BC, St. Aimie
ST, Cook
LC, Weaver
KR, Dolbow
DR. The Effect of Dual Task Activities on the Walking Gait of Individuals with Parkinson's Disease. Clin Kinesiol Online Ed. 2015;69(1):1–4.
+
Gao
X, Martin
ER, Liu
Y, Mayhew
G, Vance
JM, Scott
WK. Genome-wide linkage screen in familial Parkinson disease identifies loci on chromosomes 3 and 18. Am J Hum Genet. 2009 Apr;84(4):499–504.
+
Gardner
RC, Burke
JF, Nettiksimmons
J, Goldman
S, Tanner
CM, Yaffe
K. Traumatic brain injury in later life increases risk for Parkinson disease.
Ann Neurol. 2015;77(6):987–995.
[PubMed: 25726936]
+
Goroll
A, Mulley Jr.
A. Primary care medicine: Office evaluation and management of the adult patient. 6th ed. Philadelphia: Lippincott Williams & Wilkins. 2009; 1223–1225.
+
Hackney
ME, Earhart
GM. Effects of dance on movement control in Parkinson's disease: a comparison of Argentine tango and American ballroom. J Rehabil Med. 2009 May; 41(6): 475–81.
+
Hancock
DB, Martin
ER, Mayhew
GM, Stajich,
JM, Jewitt
R, Stacy
MA, Scott
BL, Vance
JM, Scott
WK. Pesticide exposure and risk of Parkinson's disease: A family-based case-control study.
BMC Neurol. 2008;8:1–12.
[PubMed: 18211709]
+
Hand
A, Gray
W, Walker
R,
et al. Medication use in people with late stage Parkinson's disease and parkinsonism living at home and in institutional care in north-east England: A balance of symptoms and side-effects?. PRD November 2016;32:120–123.
+
Hauser
RA
et al.. Safety and tolerability of pardoprunox, a new partial dopamine agonist, in a randomized, controlled study of patients with advanced Parkinson's disease.
Eur Neurol. 2009;62(1):40–8.
[PubMed: 19407454]
+
Helmich
RC, Hallett
M, Deuschl
G, Toni
I, Bloem
BR. Cerebral causes and consequences of parkinsonian resting tremor: a tale of two circuits?
Brain. 2012;135(11):3206–3226.
[PubMed: 22382359]
+
Hiorth
YH, Lode
K, Larsen
JP. Frequencies of falls and associated features at different stages of Parkinson's disease.
Eur J Neurol. 2013;20(1):160–166.
[PubMed: 22816560]
+
Hoehn
M, Yahr
M. Parkinsonism: onset, progression, and mortality.
Neurology. 1967;17:427–42.
[PubMed: 6067254]
+
Hughes
AJ, Daniel
SE, Kilford
L, Lees
AJ. Accuracy of clinical diagnosis of idiopathic Parkinson's disease: a clinico-pathological study of 100 cases.
J Neurol Neurosurg Psychiatry. 1992;55(3):181–4.
[PubMed: 1564476]
+
Keus
S, Bloem
B, Hendriks
E, Bredero-Cohen
A,
et al. Evidence-based analysis of physical therapy in Parkinson's disease with recommendations for practice and research. Movement Disorders. 2007;22.4:451–460.
+
Lagalla
G, Millevolte
M, Capecci
M, Provinciali
L, Ceravolo
MG. Long-lasting benefits of botulinum toxin type B in Parkinson's disease-related drooling. J Neurol. 2009 Apr;256(4):563–7.
+
Li
F, Harmer
P, Fitzgerald
K, Eckstrom
E, Stock
R, Galver
J, Maddalozzo
G, Batya
S. Tai chi and postural stability in patients with Parkinson's disease. Engl J Med. 2012;366:511–519.
+
Lim
TT, Kluger
BM, Rodriguez
RL, Malaty
IA, Palacio
R, Ojo
OO, Patel
S, Gujrati
Y, Nutter
B, Swartz
C, Hennessy
C. Rasagiline for the symptomatic treatment of fatigue in Parkinson's disease. MovDisord. November 2015;30(13):1825–1830.
+
Mahlknecht
P, Pechlaner
R, Boesveldt
S,
et al. Optimizing odor identification testing as quick and accurate diagnostic tool for Parkinson's disease. Mov Disord2016;31(9):1408–1413.
+
Marras
C, Rochon
P, Lang
E. Predicting motor decline and disability in Parkinson disease. Archives of Neurology. 2002;59:1724–28.
+
Mhyre
T, Boyd
J, Hamill
R,
et al. Parkinson's Disease: Chapter 16. Protein aggregation and fibrillogenesis. in cerebral and systemic amyloid disease. Subcellular Biochemistry. 2012;65:389–455.
+
Moisan
F, Spinosi
J, Elbaz
A,
et al. Association of Parkinson's Disease and Its Subtypes with Agricultural Pesticide Exposures in Men: A Case-Control Study in France. Environ Health Perspect November 2015;123(11):1123–1129.
+
Nolden
L, Tartavoulle
T, Porche
D. Parkinson's Disease: Assessment, Diagnosis, and Management. JNP July 2014;10(7):500–506.
+
Parkinson's Disease Foundation. Available at
http://www.pdf.org. Accessed April 4
th, 2006.
+
Patel
K. Optimising medication for Parkinson's disease patients with dysphagia. Br J Community Nurs July 2015;20(7):322–326.
+
Pavon
J, Whitson
H, Okun
M. Parkinson's disease in women: a call for improved clinical studies and for comparative effectiveness research. Maturitas, 2010;65(4):352–358.
+
Politis
M, Lindvall
O. Clinical application of stem cell therapy in Parkinson's disease. BMC Medicine, 2012;10(1):1.
+
Pringsheim
T, Jette
N, Frolkis
A, Steeves
TDL. The prevalence of Parkinson's disease: A systematic review and meta-analysis. Mov Disord. 2014;29(13):1583–1590.
+
Pulliam
CL, Heldman
DA, Orcutt
TH, Mera
TO, Giuffrida
JP, Vitek
JL. Motion sensor strategies for automated optimization of deep brain stimulation in Parkinson's disease. Parkinsonism Relat Disord. 2015; In Press. doi:10.1016/j.parkreldis.2015.01.018.
+
Reithler
J, Peters
JC, Sack
AT. Multimodal transcranial magnetic stimulation: Using concurrent neuroimaging to reveal the neural network dynamics of noninvasive brain stimulation. Prog Neurobiol. 2011;94(2):149–165.
+
Rochester
L, Burn
DJ, Woods
G, Godwin
J, Nieuwboer
A. Does auditory rhythmical cueing improve gait in people with Parkinson's disease and cognitive impairment? A feasibility study. Mov Disord. 2009 Apr 30;24(6):839–45.
+
Rosenfeldt
AB, Rasanow
M, Penko
AL, Beall
EB, Alberts
JL. The cyclical lower extremity exercise for Parkinson's trial (CYCLE): methodology for a randomized controlled trial. BMC Neurol. 2015;15. doi:10.1186/s12883-015-0313-5.
+
Sage
MD, Almeida
QJ. Symptom and gait changes after sensory attention focused exercise vs aerobic training in Parkinson's disease. Mov Disord. 2009 Jun 15;24(8):1132–8
+
Sato
K, Hatano
T, Yamashiro
K, Kagohashi
M, Nishioka
K, Izawa
N, Mochizuki
H, Hattori
N, Mori
H, Mizuno
Y, Juntendo Parkinson Study Group. Prognosis of Parkinson's disease: time to stage III, IV, V, and to motor fluctuations. Mov Disord. 2006;21(9):1384–95.
+
Shin
J, Hendrix
C. Management of patients with Parkinson disease. Nurse Practitioner, 2013;38(10):34–44.
+
Singer
C. Managing the patient with newly diagnosed Parkinson disease. Cleveland Clinic Journal of Medicine. 2012;79:2.
+
Singh
N, Pillay
V, Choonara
Y. Advances in the treatment of Parkinson's disease. Progress in Neurobiology. 2007;29–44
+
Stacy
M. Nonmotor symptoms in Parkinson's disease. Int J Neeurosic. 2011;121(S2):9–17.
+
Taylor
K, Cook
J, Counsell
C. Heterogeneity in male to female risk for Parkinson's disease. J Neurol Neurosurg Psychiatry. 2007;78:905–906.
+
Taylor
K, Saint-Hilaire
M, Weisskopf
M,
et al. Head injury at early ages is associated with risk of Parkinson's disease. Parkinsonism Relat Disord. February 2016;23:57–61.
+
Temel
Y, Visser-Vandewalle
V, Carpenter
RH. Saccadometry: a novel clinical tool for quantification of the motor effects of subthalamic nucleus stimulation in Parkinson's disease. Exp Neurol. 2009 Apr;216(2):481–9.
+
Turnbull
GI. Physical Therapy Management of Parkinson's Disease. New York, NY: Churchill Livingstone; 1992.
+
Valkovic
P, Harsany
J, Hanakova
M, Martinkova
J, Benetin
J. Nonmotor Symptoms in Early- and Advanced-Stage Parkinson's Disease Patients on Dopaminergic Therapy: How Do They Correlate with Quality of Life?. ISRN NeurologyJanuary 2014;:1–4.
+
Widnell
KL, Comella
C. Role of COMT inhibitors and dopamine agonists in the treatment of motor fluctuations. Movement Disorders. 2005;20:S30–S37.
+
Wirdefeldt
K, Adami
H, Cole
P, Trichopoulos
D, Mandel
J. Epidemiology and etiology of Parkinson's Disease: a review of the evidence. Eur J Epidemiol. 2011;26:S1–S58.
+++
Pediatric Infectious Diseases
++
Cytomegalovirus (CMV)
Etiology
CMV contains a genome of double-stranded linear DNA and a viral envelope protein that includes immunogenic glycoprotein B. The biologic characteristic of all herpes viruses is the ability to establish latency following primary infection, which allows for periodic viral reactivations during the life of the human host. (Razonable, 2002)
Herpes virus that is spread through close contact with urine, saliva, blood, and genital secretions
Most common disabling perinatal infectious disease with 0.3-1.2% birth prevalence (Grosse, 2007)
Risk of vertical transmission is very high after primary maternal CMV infection
Pathogenesis
CMV infects a variety of cells, including mononuclear leukocytes and endothelial cells and has a replication cycle of approximately 1 day. During infection, CMV antigens trigger the innate immune system to secrete various antiviral peptides, including interferon. (Compton, 2003)
CMV infection rate increases with each stage of pregnancy (Feldman, 2011)
40% of women with primary infection during pregnancy will transmit CMV to the fetus, but women who are infected before pregnancy will transmit to the fetus only 0.2-2% of the time (McCarthy, 2011)
The virus will spread to the fetus through the placenta by way of infected leukocytes (Chen, 2010)
The two most common ways pregnant women are infected is through sexual contact or through contact with the urine of a young child. (Pass, 2014)
Herpes
Etiology
Viral infection from the Herpes Simplex Virus (HSV)
Neonatal human herpes simplex virus (NHSV) is caused by exposure in the utero. (Upton, 2014) Congenital herpes simplex is caused by exposure peripartum, or postpartum (Kimberlin 2013, Koren, 2013)
Babies normally contract the virus peripartum or in the birth canal during delivery, in utero and postpartum infection are rare. (Marquez, 2011)
Women with primary genital HSV disease are at highest risk of transmitting the virus to baby. (Koren, 2013)
Babies born to mothers who have a first episode of genital HSV near full term have a much greater chance of developing neonatal herpes than babies whose mother has recurrent genital herpes. (AAP, 2012)
Pathogenesis
For transmission to occur, mother must be "shedding virus" which happens 1 out of every 3 days which increases the chances of the baby contracting the virus (Kimberlin, 2004)
The baby has a chance of contracting the virus at 3 distinct times: in utero: 5%; peripartum: 85% (Koren, 2013); postpartum: 10% (Kimberlin,2013)
Peripartum and postpartum can be further classified into sub categories: Disease localized to the skin, eyes, and/or mouth (SEM disease): 45%; Encephalitis with or without SEM involvement (CNS disease): 30%; Disseminated infection involving multiple organs (Disseminated disease: 25% CNS, lungs, liver, adrenal glands, skin, eyes, and/or mouth) (Kimberlin, 2004)
HIV/AIDS
Etiology
Caused by the transferring of the Human Immunodeficiency Virus (HIV) from the mother to the baby either in utero, peripartum, or postpartum through breastfeeding in 1/3 of cases. (Scarlatti, 2004)
Newly acquired pediatric HIV infections have virtually been eliminated in the developed world, with mother-to-child transmission (MTCT) rates reduced to approximately 1–2% since the advent of highly active antiretroviral (HAART) therapy. (Tiemessen, 2006)
More common in developing countries (Sarnquist, 2013)
Perinatal HIV transmission is the most common route from mother to child (Scarlatti, 2004)
By 2016, 2,100,000 children ages 0-14 were infected with HIV around the world. (The WBG, 2017)
Before 1995, children with perinatal HIV had 50% chance of developing AIDS by age 3 and a 90% chance by age 10. (DHHS, 2017)
Death is usually by opportunistic infections secondary to the virus rather than the virus itself (DHHS, 2017)
Meningitis
Etiology
pathogen infection leading to inflammation within the subarachnoid space, affecting both the pia and arachnoid meninges.
Five types: Bacterial meningitis, viral meningitis, parasitic meningitis, fungal meningitis, noninfectious meningitis (develops secondary to cancer, lupus, head injury or brain trauma) (CDC)
Many types of bacteria can cause bacterial meningitis, however, Neisseria meningitidis is the most common cause of meningitis and fatal sepsis (Pace, 2011, Johri, 2005).
Pathogenesis
Typically affects younger children under the age of 3 years and young adults (Johri, 2005)
Transmission of bacterial meningitis occurs through droplets or respiratory secretions. The bacteria then colonize in the mucosal cells in the upper respiratory tract. Infection may spread via the bloodstream to seed in other body locations including the meninges, joints, and pericardium (Johri, 2005). Viral meningitis is spread through fecal contamination (CDC).
Environmental factors: overcrowding in nursery/ school, college campus, military quarters, urban residence, exposure to tobacco smoke, contact with an individual with meningitis or carrier (Johri, 2005).
Rubella
Etiology
Pathogenesis
Maternal infection of rubella results in CRS of approximately 85% of the developing fetuses during the first trimester. The risk of transmission decreases with increased gestational age after the 20th week (White, 2012).
Maternal rubella has very mild signs and symptoms, which can go undetected.
Virus transmission occurs primarily through respiratory secretions (Cooper, 2001). Transmission often occurs in collective groups, either between children, children to parents, or adults to adults (Plotkin, 2006).
Toxoplasmosis
Etiology
Parasitic infection of the brain caused by caused by the protozoan parasite Toxoplasma gondii. People typically become infected by ingesting oocytes excreted by cats, or contaminated soil and water, as well as eating undercooked meat of infected animals with tissue cysts. (CDC)
Pathogenesis
Incubation rate is 4-21 days, 7 days on average.
Risk of transmission from mother to fetus is higher in the third trimester (5% at 12 weeks to 80% at delivery)
Rate of infection varies between countries - 1-10/10,000 in western countries, to 16/1000 in Brazil
Factors associated with congenital infection include: route of transmission, climate, cultural behavior, eating habits and hygienic standards (DiMario, 2013)
++
Cytomegalovirus
Signs and Symptoms
Newborns with congenital CMV infection present with microcephaly, hepatosplenomegaly, neurologic abnormality (poor tone, motor function, and abnormal head lag), petechiae or purpura, and hearing loss. (Michaels, 2003)
Permanent Bilateral or sensorineural hearing loss (in 14.1% of cases) (Grosse, 2007)
Could also have seizures, increased tone and posture abnormalities, hearing loss, ocular abnormalities (Luck, 2008)
Diagnostic testing
Prenatal ultrasound features suggestive of congenital CMV infection include: intrauterine fetal growth restriction, enlargement of the cerebral ventricles, intracranial calcification, microcephaly, and increase or decrease in the volume of amniotic fluid. (Revello, 2004)
A specimen is taken from the infant in first 3 weeks of life and needs 1+ of the following (Michaels, 2003):
Isolation of virus from the urine or other secretions (saliva); IgM in the blood; detection of CMV DNA by polymerase chain reaction in the urine, blood, saliva, or other secretions
Abnormal cranial CT (most sensitive indicator of intellectual and motor deficits). Intracranial calcifications, microcephaly, and cerebral ventricular enlargement may be detected. (Jones, 2009; Michaels, 2003)
Herpes
Signs and Symptoms
Often presents with small, fluid-filled blisters (vesicles) usually on the skin, eyes, or the mouth (SEM disease). These vesicles will burst, crust over and heal, often leaving a small scar.
Does not necessarily have to present with vesicles but the virus can spread to other parts of the body affecting different organs (Disseminated disease) (Kimberlin, 2004). CNS disease (encephalitis): Seizures, coma, lethargy, shock; Other organs: bleeding easily, breathing difficulties (cyanosis, flaring of nostrils, grunting, tachypnea, apneic episodes: periods of not breathing); jaundice, hypothermia, fever, conjunctivitis (Kimberlin, 2004)
Diagnostic testing
Infants with cutaneous lesions
Direct Fluorescent Antibody Test for herpes (scrapings from lesion)
Viral culture of the superficial lesion as well as of the oropharynx, nasopharynx, conjunctiva, and rectum, CSF (Kimberlin, 2013).
Blood and urine test will detect any antibodies toward the herpes virus
Polymerase chain reaction (PCR): a rapid genetic amplification technique, which detects the presence of minute quantities of viral DNA is shown to be highly sensitive to the herpes virus.
Direct immunofluorescent antibody staining: detects the presence of the HSV antigen (Kimberlin, 2013).
For infants presenting with seizures
Not always tested in an Emergency room setting, often overlooked (McGuire, 2012)
HIV/AIDS
Signs and Symptoms
HIV symptoms: vary by age; failure to thrive, failure to gain weight or inability to grow according to the standardized charts; failure to reach developmental milestones (NIAID); CNS involvement: seizures, difficulty learning to walk; frequent childhood illnesses: ear infections, colds, stomach aches, diarrhea
AIDS (opportunistic infections): pneumocystis pneumonia (fungal infection); infection due to the cytomegalovirus (CMV); lymphatic interstitial pneumonitis (lung scarring); thrush; severe candidiasis: diaper rash from a yeast infection (HIV and AIDS Health Center)
Diagnostic testing
Immune complex dissociated HIV p24 antigen assay
Rapid, simple, serologic that is shown to be effective to diagnosis HIV infection in neonates (Miles et al.)
Specific virologic assays that detect HIV must be used in infants younger than 18 months
Preferred Virologic assays: HIV DNA polymerase chain reaction; HIV RNA assays
If there is known exposure to HIV it is recommended to use virologic testing at 14-21 days, 1-2 months, and 4-6 months
Could be done at birth if there is speculation of high risk of HIV transmission form the mother
A positive test should be repeated on a separate specimen to be confirmed
A definitive exclusion can either be 2 negative virologic tests in non-breastfed infants with one obtained at less than 1 month and one at 4 months or 2 negative tests from different specimens at 6 months (DHHS)
Recombinase Polymerase Amplification provides early, rapid diagnosis of the HIV-1 Proviral DNA in infants (Boyle, 2013)
Meningitis
Signs and Symptoms
Meningococcal skin rash assists with diagnosis of bacterial meningitis.
Acute onset: fever, severe headache, nausea and vomiting, photophobia and neck stiffness, seizures, irritability, drowsiness, confusion, and delirium progressing to decreased levels of consciousness due to cerebral involvement. Increased ICP, cerebral edema, hydrocephalus, ischemic necrosis of distal extremities, shock and multiple organ failure may also occur with the infection (Johri, 2005)
Late symptoms: Communicating hydrocephalus, subdural effusions in children, deafness, cranial nerve palsies, and mental retardation (Johri, 2005)
Diagnostic testing
Meningeal irritation: positive nuchal signs, Kernig's and Brudzinski's sign.
Lumbar puncture to obtain CSF sample for identification of pathogen (CDC).
Rubella
Signs and Symptoms
Congenital Rubella Syndrome: Sensorineural hearing loss (deafness), cardiac malformations, cognitive deficits, ophthalmic defects such as cataracts, spleen and liver damage. (Herini, 2017)
Diagnostic testing
Rubella virus in the mother is diagnosed after isolating the rubella virus from a throat culture or nasal specimen. ELISA to isolate rubella specific antibodies is a more time efficient methods for diagnosis (White, 2012).
CRS in fetus/infant diagnosed using a specimen from the cord blood, placental tissue, and/or amniotic fluid (White, 2012) Clinical specimens for the diagnosis of rubella by virus detection utilize throat swabs, oral fluids (OF) or nasopharyngeal secretions, and by antibody detection are usually sera or OF. (Lambert, 2015)
Toxoplasmosis
Signs and Symptoms
Intrauterine death or stillbirth; malformation; cognitive deficits; deafness, blindness; hydrocephalus; microcephaly; cerebral calcifications; seizures; systemic infection signs (fever, rash, enlarged liver and spleen); psychomotor deficits (Kaye, 2011)
Most cases are asymptomatic except for congenital infection and patients who are immunosuppressed.
Diagnostic testing
Serum tests of IgG and IgM for the mother
Amniocentesis test for a fetal infection (detects the parasite or toxoplasma DNA in amniotic fluid)
Cordocentesis includes drawing blood from the umbilical cord to detect the parasite or IgM, IgA in fetal blood.
++
Cytomegalovirus
Herpes
Depends on the classification of the disease (Cherpes, 2012)
Infants with SEM disease will have recurrent lesion and will need to return for treatment throughout life
Infants with disseminated disease are less likely to survive than infants with SEM or CNS disease. (Upton, 2014)
Long-term sequalae, including learning disabilities, cerebral palsy, blindness, and persistent seizures (Malm, 1995)
HIV/AIDS
Good prognosis if mother is treated with antiviral therapy during the pregnancy but adherence if is found to be low (Bardeguez, 2008)
Short courses of Zidovudine (Brocklehurst, 2002)
Single dose Nevirapine (Brocklehurst, 2002)
Depending on if the HIV is HIV-1 or HIV-2
Progression to AIDS is slower for HIV-2 compared to HIV-1 (DHHS)
Perinatal infection accelerates the disease progression for children because of their immature immune systems
25% develop AIDS within their first year of life, 50% by 2 years in resource poor environments (Prendergast, 2007)
Caesarean delivery has the lowest risk of transmission and is done in most cases (Prendergast, 2007; Read, 2005)
Use of formula rather than breastfeeding will decrease the risk of transmission. (Suy et al.)
Meningitis
Non-progressive disease
Bacterial meningitis typically has the most severe symptoms. Mortality is estimated around 5-18% (CDC).
Long-term sequelae: uni/bilateral sensorineural hearing loss, seizures, vision deficits, motor deficits, and behavioral problems (Pace, 2011).
Rubella
Non-progressive disease
Signs and symptoms of congenital rubella syndrome may continue to be present for 10-20 years.
Damage to the child during gestation may remain with the child throughout life, unless it can be corrected by surgery.
Toxoplasmosis
Maternal infection depends on if the infection is acquired during pregnancy or if it was acquired before pregnancy.
Women who had the infection before pregnancy leave the child in a much better condition due to immunity to the parasite. Immunoglobulin confirmation testing is needed.
If fetal infection: treatment through medications goes through the first year of life.
Fetal infections could cause developmental abnormalities which stays with the child for life. (Kaye, 2011; Di Mario, 2013)
+++
IV. Medical/ Surgical Management
++
Cytomegalovirus
Medical/Pharmaceutical
Intravenous ganciclovir or oral valganciclovir is recommended for the treatment of newborns infected with CMV. (Oliver, 2009; Kimberlin, 2015)
There is no treatment available for pregnant women to prevent passing of CMV to the fetus. No currently licensed vaccine for CMV is available, however, several candidate vaccines are in preclinical and early-phase development. Phase II studies have shown promise in reported reduction in CMV infection in pregnant women and transplant recipients. (McCarthy, 2011; Griffiths, 2011)
Repeated eye and ear examinations are needed if infant is infected (Grosse, 2007)
Herpes
Medical/Pharmaceutical
The main treatment is prevention: education to women and men about transferring the virus to others which can be passed to the baby
A cesarean delivery can reduce the risk of contamination of the infant (Kimberlin, 2004, Kimberlin 2013)
Medical treatment cannot undo any damage done to the body, but it can possibly decrease the chances of the damage from getting worse
Treatment only help control the symptoms and does not cure herpes
Medications (Antivirals)
Acyclovir: Drug of choice because of safety and ease of administration
Given by injection to treat the symptoms but will not reverse damage.
38% of infants with SEM disease showed developmental difficulties was reduced to 2% with Acyclovir (Kimberlin, 2004)
There was also a decrease of infant's SEM disease progressing to disseminated or CNS disease (Kimberlin, 2004)
Higher doses led to a decrease in mortality at 12 months from 85% to 29% in disseminated disease and 4% in CNS disease (Kimberlin, 2004)
Increased infants developing normally from 50% to 83% in disseminated disease, did not affect morbidity in CNS disease (Kimberlin, 2004)
Vidarabine
Given in higher doses and not as popular.
Did show improvements but not at the same rate and with the same effectiveness at Acyclovir (Whitley, 1986)
HIV/AIDS
Medical/Pharmaceutical
Antiviral drugs during the pregnancy and 4 hours after birth if the infant is diagnosed with HIV (Cohen, 2013, Management of HIV)
Co-Trimoxazole: Given to HIV positive infants in the first year of life to fight against Pneumocystic Jirovecii pneumonia (Grimwade et al.)
Supplements: Provide oral supplementation when there is a nutritional deficit
Monitor weight and growth (Wang et al. 2008)
Meningitis
Medical/Pharmaceutical
Bacterial meningitis is treated with antibiotics
Prevention: vaccines available for three types of bacteria that can cause meningitis including Neisseria meningitidis (meningococcus), Streptococcus pneumoniae (pneumococcus), and Haemophilus influenzae type b (Hib) (CDC).
No specific treatment available for viral meningitis- most resolve within 7-10 days (CDC).
Anti-fungal medications for treatment of fungal meningitis.
Parasitic meningitis very rare and is nearly always fatal even with treatment of various medications (CDC).
Rubella
Medical/Pharmaceutical
Primary treatment is prevention through vaccination. Most developed countries require immunization of all infants as well as focused prevention in healthcare workers, adult women, and military recruits (Plotkin, 2006).
Developing countries are beginning to give rubella vaccines as well to decrease the prevalence of the disease (Plotkin, 2006).
Vaccination is a part of the MMR series given to infants (12-15 mo.) and followed up with a second booster MMR vaccine years later (4-6 years old) (NCIRD). In the United States, the rubella virus has nearly been eliminated by vaccinations (Plotkin, 2006).
Toxoplasmosis
Medical/Pharmaceutical
Spiramycin regiment should be implemented as soon as possible for infected pregnant women in first and second trimester (CDC)
pyrimethamine/sulfadiazine for the second/third trimester (CDC)
Antitoxoplasma treatment throughout pregnancy, added folinic acid for bone marrow health (haemoatotoxicity monitoring is mandatory) (CDC)
Ultrasounds should be monthly to check for fetal abnormalities (CDC)
Prevention is main factor:
Pregnant women should avoid eating undercooked meat or drinking untreated water
Use gloves while gardening or touching soil/sand
Avoid changing cat litter pans, if necessary use gloves and wash hands right after (Berrebi, 2011)
+++
V. Implications for Therapeutic Management
++
Cytomegalovirus
Education on Prevention: washing hands when dealing with infant's diapers, saliva, or other bodily fluids
Congenital CMV is more likely to cause permanent disability than perinatal but hearing and vision deficiency can develop over time through perinatal infection (CDC)
Focus on specific problems such as lack of coordination and psychomotor deficiencies
Herpes
Focus on symptoms the patient presents with: incoordination, balance, motor control, cognition, breathing difficulties, wound care (Kimberlin, 2004)
Education to the mother about risks during pregnancy and during birth
Always be aware of proper use of PPE. Can be spread from person to person through bodily contact.
Given the potential for significant neurological sequelae in survivors of NHSV infection, affected infants should have a structured follow-up program that allows for neurodevelopmental, ophthalmological, and hearing assessments (Upton, 2014)
HIV/AIDS
Education on the risks of the baby contracting the virus (deRuiter, 2012)
C-section delivery normally done to prevent transmission
Prevention and education on the spread of the virus
Always be aware of proper use of PPE
Symptoms may cause difficulty in therapy if they were seeking treatment of another problem: fatigue, ear infection, colds, stomach aches, diarrhea
Address delayed motor development, failure to thrive, delayed ambulation
Meningitis
Always wash hands to prevent transmission.
Deficits in cognition, arousal, behavior, hearing and vision may be present secondary to the meningitis infection and will require screening to observe impact on therapeutic treatment.
Rubella
Take a thorough history of the patient's disease, secondary complications from birth to their current age.
Expect deficits in hearing, vision, as well as cognitive defects and screen appropriately to help guide interventions.
Importance of health care workers to be vaccinated against Rubella to protect self and prevent spread of disease to others.
Toxoplasmosis
Prevention is the main key for the mother. Education on how the disease is acquired is important.
Wash hands frequently, do not eat undercooked meat, do not drink contaminated water
Check for neuromuscular deficits and systemic infection signs in newborns
Consistent vision and hearing check-ups (Berrebi, 2010)
+
Alford
CA, Jr, Neva
FA, Weller
TH. Virologic and serologic studies on human products of conception after maternal rubella. New Eng J Med. 1964;271:1275–1281.
+
Allen
UD, Robinson
JL. Canadian Paediatric Society, Infectious Diseases and Immunization Committee. Prevention and management of neonatal herpes simplex virus infections. J Paediatr Child Health. 2014;19(4):201–206.
+
American Academy of Pediatrics (AAP). Herpes simplex virus. In: Pickering
LK. Red Book: 2012 Report of the Committee on Infectious Diseases. 28th ed. Elk Grove Village, IL: AAP; 2012:363–73.
+
Bardeguez
AD, Lindsey
JC, Shannon
M. Adherence to antivirals among us women during and after pregnancy. J Acquir Immune Defic Syndr. 2008;48(4):408–17.
+
Berrebi
A, Assouline
C, Bessieres
MH
et al.. Long-term outcome of children with congenital toxoplasmosis. Am J Obstet Gynecol. 2010;203:552.e1–6.
+
Boyle
D, Lehman
D, Lillis
L,
et al. Rapid detection of HIV-1 proviral dna for early infant diagnosis using recombinase polymerase amplification. Am Soc Microbiol. 2013;4(2):e00135–13.
+
Brocklehurst
P, Volmink
J. Antivirals for reducing the risk of mother-to-child transmission of HIV infection. Cochrane Database Syst Rev. 2002(2):CD003510.
+
Chen
CP, Su
YN, Chern
SR,
et al. Detection and comparison of cytomegalovirus DNA levels in amniotic fluid and fetal ascites in a second trimester fetus with massive ascites, hyperechogenic bowel, ventriculomegaly and intrauterine growth restriction. Taiwan J Obstet Gynecol. 2010;49(2):206–210.
+
Cherpes
T, Matthews
D, Maryak
S. Neonatal herpes simplex virus infection. Clin Obstet Gynecol.2012;55(4):938–944.
+
Cohen
J. Early treatment may have cured infant of HIV infection. Science. 2013;339(6124): 1134.
+
Compton
T, Kurt-Jones
EA, Boehme
KW,
et al. Human cytomegalovirus activates inflammatory cytokine responses via CD4 and toll-like receptor 2. J Virol. 2003;77:4588–4596.
+
Cooper
L, Alford
CA. Rubella. In: Remington
JS, Klein
JO Infectious diseases of the fetus and newborn infant. Philadelphia, PA: WB Saunders; 2001:347–88.
+
Department of Health and Human Services (DHHS). AIDS Info. Recommendations for Use of Antiretroviral Drugs in Pregnant HIV-1-Infected Women for Maternal Health and Interventions to Reduce Perinatal HIV Transmission in the United States.
https://aidsinfo.nih.gov/guidelines/html/3/perinatal/0. Accessed Oct. 15, 2017.
+
Di Mario
S, Basevi
V, Gagliotti
C,
et al. Prenatal education for congenital toxoplasmosis. Cochrane Database of Syst Rev. 2013;2.
+
Feldman
B, Yinon
Y, Tepperberg
M, Yoeli
R, Schiff
E, Liptiz
S. Pregestational, periconceptional, and gestational primary maternal cytomegalovirus infection: prenatal diagnosis in 508 pregnancies. Am J Obstet Gynecol. 2011;205(342):1–6.
+
Granerod
J, Davies
NWS, Mukonoweshuro
W,
et al. Neuroimaging in encephalitis: analysis of imaging findings and interobserver agreement. Clin Radiol. 2016;71(10):1050–1058.
+
Griffiths
PD, Stanton
A, McCarrell
E,
et al. Cytomegalovirus glycoprotein-B vaccine with MF59 adjuvant in transplant recipients: a phase 2 randomised placebo-controlled trial. Lancet. 2011;377:1256–1263.
+
Grimwade
K, Swingler
GH. Cotrimoxazole prophylaxis for opportunistic infections in children with HIV infection. Cochrane Database Syst Rev. 2006; Jan 25;(1):CD003508.
+
Grosse
SD, Ross
DS, Dollard
SC. Congenital cytomegalovirus (CMV) infection as a cause of permanent bilateral hearing loss: a quantitative assessment. J Clin Virol. 2007;41(2):57–62.
+
Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection, National Institute of Health, US HAART guidelines for children. July 2008.
+
Herini
ES, Gunadi, Triono
A,
et al. Clinical profile of congenital rubella syndrome in Yogyakarta Indonesia. Pediatr Int. 2017;Oct. 25. doi: 10.1111/ped.13444. [Epub ahead of print]
+
Johri
S, Gorthi
S, Anand
A. Meningococcal meningitis. Med J Armed Forces India. 2005;61(4):369 ––374.
+
Jones
CA, Walker
KS, Henderson-Smart
DJ. Antiviral therapy for symptomatic congenital cytomegalovirus infection in neonates and infants up to 3 months of age. Cochrane Database of Syst Rev. 2009;Issue 3:CD004340.
+
Kaye
A. Toxoplasmosis: diagnosis, treatment, and prevention in congenitally exposed infants. J Ped Healthcare. 2011;25(6):355–364.
+
Kimberlin
D, Baley
j. Guidance on management of asymptomatic neonates born to women with active genital herpes lesions. Am Acad Pediatr 2013;131(2):e635–646.
+
Kimberlin
DW, Jester
PM, Sánchez
PJ,
et al; National Institute of Allergy and Infectious Diseases Collaborative Antiviral Study Group. Valganciclovir for symptomatic congenital cytomegalovirus disease. N Engl J Med. 2015;372:933–943.
+
Kimberlin
D. Neonatal Herpes Simplex Infection. Clin Microbiol Rev. 2004;17(1):1–13.
+
Koren
A, Tasher
D, Stein
M, Yossepowitch
O, Somekh
E. Neonatal herpes simplex virus infections in Israel. Pediatr Infect Dis J. 2013;32(2):120–123.
+
Lambert
N, Strebel
P, Orenstein
W, Icenogle
J, Poland
GA. Rubella. Lancet. 2015;385(9984):2297–2307.
+
Luck
S, Sharland
M. Congenital cytomegalovirus: new progress in an old disease. Pediatr Child Health. 2008;19(4):178–184.
+
Malm
G, Berg
U, Forsgren
M. Neonatal herpes simplex: clinical findings and outcome in relation to type of maternal infection. Acta Paediatr. 1995;84(3):256–60.
+
deRuiter
A, Taylor
GP, Clayden
P,
et al. British HIV Association guidelines for the management of HIV infection in pregnant women 2012. HIV Medicine. 2014;15(Suppl. 4):1–77.
+
Marquez
L, Levy
ML, Munoz
FM, Palazzi
DL. A report of three cases and review of intrauterine herpes simplex virus infection. Pediatr Infect Dis J. 2011;30(2):153–7.
+
McCarthy
FP, Giles
ML, Rowlands
S, Purcell
KJ, Jones
CA. Antenatal interventions for preventing the transmission of cytomegalovirus (CMV) from the mother to fetus during pregnancy and adverse outcomes in the congenitally infected infant. Cochrane Database of Syst Rev. 2011, 3:CD008371.
+
McGuire
J, Zorc
J, Licht
D, Hodinka
R, Shah
S. Herpes simplex testing in neonates in the emergency department. Pediatr Emerg Care. 2012;28(10):949–955.
+
Michaels
MG, Greenberg
DP, Sabo
DL,
et al. Treatment of children with congenital cytomegalovirus infection with ganciclovir. Pediatr Infect Dis J. 2003;22:504–509.
+
Miles
S, Balden
E, Macpantay
L,
et al. Rapid serologic testing with immune-complex-dissociated HIV p24 antigen for early detection of HIV infection in neonates. New Eng J Med. 1993;328(5):297–302
+
Oliver
SE, Cloud
GA, Sánchez
PJ,
et al; National Institute of Allergy, Infectious Diseases Collaborative Antiviral Study Group. Neurodevelopmental outcomes following ganciclovir therapy in symptomatic congenital cytomegalovirus infections involving the central nervous system. J Clin Virol. 2009;46(suppl 4):S22–S26.
+
Pace
D, Pollard
AJ. Meningococcal disease: clinical presentation and sequelae. Vaccine. 2012;30:B-3-9.
+
Pass
RF, Anderson
B. Mother-to-child transmission of cytomegalovirus and prevention of congenital infection. J Pediatr Infect Dis Soc. 2014;3(Suppl 1):S2–S6.
+
Prendergrast
A, Tudor-Williams
G, Jeena
P. International perspectives, progress, and future challenges of paediatric HIV infection. Lancet. 2007;370(9581):68–80.
+
Plotkin
SA. The history of rubella and rubella vaccination leading to elimination. Clin Infect Dis. 2006;43(Supplement 3):S164–S168.
+
Razonable
RR, Paya
CV. Beta-herpesviruses in transplantation. Rev Med Microbiol. 2002;13:163–176.
+
Razonable
RR, Rivero
A, Brown
RA,
et al. Detection of simultaneous beta-herpes virus infection in clinical syndromes due to defined cytomegalovirus infection. Clin Transplant. 2003;17:114–120.
+
Read
JS, Newell
MK. Efficacy and safety of cesarean delivery for prevention of mother-to-child transmission of HIV-1. Cochrane Database Syst Rev. 2005;Oct 19;(4):CD005479.
+
Revello
M. Pathogenesis and prenatal diagnosis of congenital cytomegalovirus infection. J Clin Virol. 2004:29:71–83.
+
Sarnquist
C, Rahangdale
L, Maldonado
Y. Reproductive health and family planning needs among hiv -infected women in sub- saharan Africa. Current HIV Res. 2013;11(2):160–168.
+
Scarlatti
G. Mother-to-child transmission of HIV-1: advances and controversies of the twentieth centuries. AIDS Rev. 2004;6(2):67–78.
+
Suy
A, Hernandez
S, Thorne
C. Current guidelines on management of HIV-infected pregnant women: impact on mode of delivery. Eur J Obstet Gynecol Reprod Biol. 2008;139(2):127–32.
+
Tiemessen
CT, Kuhn
L. Immune pathogenesis of pediatric HIV-1 infection. Current HIV/AIDS Reports. 2006;3(1):13–19.
+
Wang
Q, Pang
S. An intercellular adhesion molecule-3 (ICAM-3) -grabbing nonintegrin (DC-SIGN) efficiently blocks HIV viral budding. FASEB J. 2008;22(4):1055–64.
+
White
SJ, Boldt
KL, Holditch
SJ, Poland
GA, Jacobson
RM. Measles, Mumps, and Rubella. Clin Obstet Gynecol. 2012;55(2):550–559.
+
Whitley
RJ, Alford
CA, Hirsch
MS,
et al. Vidarabine versus acyclovir therapy in herpes simplex encephalitis. New Eng J Med. 1986;314(3):144–149.
+++
Peripheral Neuropathy
++
++
++
Often progressive if underlying cause is not addressed; in acute peripheral neuropathy, symptoms appear suddenly and progress at a rapid pace; chronic PN presents subtly and progresses very slowly (NINDS, 2012)
Excellent prognosis- most patients, if underlying medical condition is identified and treated. For example: DM, nerve entrapment. Vitamin B-12 deficiency responds well to treatment, though nerve damage may be permanent. Pregnant women who experience PN during pregnancy usually return to normal postpartum or after a nutritional deficiency is corrected (Massey, 2008).
Poor prognosis-permanent damage can occur (i.e. Charcot-Marie-Tooth). Patients with uncontrolled diabetic peripheral neuropathy (DPN) may need limb amputation in later stages (Dixit, 2014). CIPN can affect long-term quality of life and can possibly be a reason to discontinue specific chemotherapy agents prematurely (Hershman, 2013).
No cure- most patients with hereditary neuropathies
Increasing age is a poor prognostic factor because the peripheral nervous system undergoes a decrease in density of small- and large-myelinated fibers, a decrease in the amplitude of nerve action potentials, and a decrease in nerve conduction velocity (Anastaci, 2013).
Sequelae- infection, poor healing, intrinsic foot muscle deterioration, decreased ankle ROM, tearing of plantar fascia, metatarsophalangeal joint and foot deformities (Cheuy, 2013), ulcerations, amputations (Duby, 2004)
Symptoms like skin ulcerations and chronic pain may slow the recovery from PN due to causing other problems like infection and immobility (Craig, 2003)
Disabilities are common and can include inability to complete ADL's safely such as bathing and dressing, as well as, inability to work at some jobs (NINDS, 2012)
+++
IV. Medical / Surgical Management
++
First step- treat underlying condition (i.e. diabetes) and/or remove external cause (i.e. alcohol) (Hughes, 2002)
General treatments include maintaining optimal weight, avoiding toxins, and correcting vitamin deficiencies (NINDS, 2012)
Medical Management
Monochromatic Infrared Photo Energy (MIRE) has shown to be effective at reducing neuropathic pain and improvement in foot sensation increasing quality of life for patients with PN with continuous use. (Harkless, 2006). In addition to decreases in neuropathic pain and increases in foot sensation, Anodyne (brand name for MIRE), has been shown to improve standing balance and decrease fall risk among patients with diabetic peripheral neuropathy. (Hao, 2008). With conflicting evidence for lack of benefit for individuals with DM (Laverly, 2008), MIRE has been supported to result in a significant decrease in pain associated with DPN (Lavery, 2008).
Surgical Management
Pharmacological Management
Tricyclic antidepressants)/ SSRIs /other antidepressants, anticonvulsants, opioids, topicals, (Duby, 2004)
Tricyclics- (amitriptyline; Elavil®), cause a balanced reuptake inhibition of serotonin and norepinephrine
Botulinum Toxin A- significant pain reduction was reported vs. placebo after intradermal injections to the dorsum of the foot (Ghasemi, 2014)
Selective serotonin reuptake inhibitors (SSRIs)- (Fluoxetine; Prozac®) selective inhibition of presynaptic reuptake of serotonin not norepinephrine, include Nortriptyline, Desipramine (Hershman, 2013).
Anticonvulsants- (carbamazepine) unspecific sodium channel blockers / reduce neuronal excitability in sensitized C-nociceptors
Serotonin-norepinephrine reuptake inhibitors (SNRIs)- (venlaflaxine;Effexor®, Tapentadol) serotonin and noradrenaline reuptake inhibitor specific for diabetic painful neuropathy (Chong, 2007; Afilalo, 2013). Duloxetine (Hershman, 2013).
Opioids- (NSAIDs, tramadol)modifies or inhibits pain impulses, inhibition of substance P
Prostacyclin Analogue- (beraprost sodium) antiplatelet and vasodilating properties (Shin, 2013)
Topicals- (Capsaicin) desensitization of type C nociceptive fibers by depleting substance P. Patch applied directly onto skin for quick delivery (Vinik, 2016).
NSAIDS-first line therapy for mild peripheral neuropathy (Nicholas, 2014)
Alpha-Lipoic Acid- 600 mg intravenous plus oral doses produced significant improvements in DPN symptoms vs. a placebo (Ziegler, 1995)
ARA-290- A non-erythropoietic peptide created from erythropoietin, has positive effects in diabetic small fiber neuropathy and metabolic control without the side effects of erythropoietin (Brines, 2015).
Alternative Management
Acupuncture treatment has also been shown to increase nerve conduction of the sural nerve in PN of undefined etiology (Shroder, 2007). Manual acupuncture is used in China to treat diabetic peripheral neuropathy, both as a stand-alone treatment and in conjunction with traditional approaches. Research has shown that the intervention has a positive effect on global symptom improvement and NCV in both treatment scenarios (Chen, 2013).
Acupuncture/Moxibustion (acu/moxa) is another alternative therapy that is used in China. Moxibustion is a form of heat therapy (burning of mugwort leaf). Research shows it significantly reduced neuropathic pain in patients with sensory neuropathy due to HIV (Nicholas, 2014).
Reflexology (Pressure application to specific regions or points on the body with the intentions of reflexively affecting other specific body organs or systems), applied to the feet has been associated with a significant decrease in symptoms of diabetic neuropathy and greater effect than pharmacologic treatment (Dalal, 2014).
+++
V. Preview of Therapeutic Management
++
Preferred Practice Patterns
5G Impaired Motor Function and Sensory Integrity Associated With Acute or Chronic Polyneuropathies.
5F Impaired Peripheral Nerve Integrity and Muscle Performance Associated with Peripheral Nerve Injury.
Physical Therapy
For prevention of muscle weakness: therapeutic exercise, aerobic training, resistance training
For neuropathic pain: TENS (Spallone, 2012), ice massage (Ownby, 2006)
For gait abnormalities: balance exercises, gait training, fitting for assistive devices, foot orthoses with consultation of orthotics specialist (Mueller, 2006)
For palliative and preventive care: TENS and MIRE/Anodyne (Harkless, 2006; Hao, 2008)
For improvement with IADL's: PT with consultation of occupational therapists.
For proprioceptive loss and balance deficits: Exercise can improve balance and decrease fall risk (Shoemaker, 2001). Whole Body Vibration, when coupled with balance and exercise training, has been shown to have greater positive effects on balance than exercise alone (Lee, 2013)
For pain and sensation deficits: Use of manual therapy and infrared can decrease pain and improve sensation deficits (Volkert, 2005).
For promotion of lower limb circulation: education on avoiding static posture and therapeutic exercise to increase circulation to lower limbs (Anastaci, 2013)
Patient education: guide patients in checking body for injuries (especially feet), check water temperature before bathing, inspect shoes for grit or wear, avoid prolonged pressure on sensitive points, education on proper footwear to reduce injury or ulceration (Anastaci, 2013)
Health and Wellness: control diabetes, limit alcohol, maintain balanced diet, exercise for weight reduction and improved circulation, healthy lifestyle and mobility
Precautions
Ulcerations, blunt force or traction to skin, extreme hot or cold, sudden numbness, tingling, or pain, falls
+++
VI. Consumer and Professional Resources
++
+
Alshahrani
A, Bussell
M, Johnson
E, Tsao
BE, Bahjri
K, The Effects of a Novel Therapeutic Intervention in Diabetic Peripheral Neuropathy Patients. Arch Phys Med Rehabil. 2016. doi: 10.1016/j.apmr.2015.12.026.
+
Afilalo
M, Morlion
B. Efficacy of Tapentadol ER for Managing Moderate to Severe Chronic Pain. Pain Physician. 2013 Jan; 16(1): 27–40.
+
Anastasi
JK, Capili
B, Chang
M. HIV Peripheral Neuropathy and Foot Care Management: A Review of Assessment and Relevant Guidelines. Am J Matern Child Nurs. 2013;113(12):34–39.
+
Brines
M, Dunne
AN, van Velzen
M, Proto
PL, Ostenson
CG, Kirk
RI, Petropoulos
IN, Javed
S, Malik
RA, Cerami
A, Dahan
A. ARA 290, a Nonerythropoietic Peptide Engineered from Erythropoietin, Improves Metabolic Control and Neuropathic Symptoms in Patients with Type 2 Diabetes. Molecular Medicine. 2014;20:658–66.
+
Bromberg
MB. An Electrodiagnostic Approach to the Evaluation of Peripheral Neuropathies, Phys Med Rehabil Clin N Am. 2013; 24(1): 153–168.
+
Çakici
N, Fakkel
TM, van Neck
JW, Verhagen
AP, Coert
JH. Systematic Review of Treatments for Diabetic Peripheral Neuropathy. Diabetic Medicine. January 1, 2016. doi:10.1111/dme.13083.
+
Cheuy
Victor A, Hastings
Mary K, Commean
Paul K, Ward
Samuel R, Mueller, Michael
J. Intrinsic Foot Muscle Deterioration Is Associated with Metatarsophalangeal Joint Angle in People with Diabetes and Neuropathy. Clinical Biomechanics. 2013 Dec 31;28(9):1055–60.
+
Chen
W, Yang
G-Y, Liu
B, Manheimer
E, Liu
J-P. Manual acupuncture for treatment of diabetic peripheral neuropathy: a systematic review of randomized controlled trials. PLOS ONE. 2013;8(9):e73764. doi:10.1371/journal.pone.0073764.
+
Chong
MS, Hester
J. Diabetic painful neuropathy: current and future treatment options. Drugs. 2007; 67 (4): 569–85
+
Craig
ASW, Richardson
JK. Acquired peripheral neuropathy, Phys Med Rehabil Clin N Am. 2003; 14(2): 365–386.
+
Dalal
K, Maran
VB, Pandey
RM, Tripathi
M. Determination of efficacy of reflexology in managing patients with diabetic neuropathy: a randomized controlled clinical trial. Evid Based Complement Alternat Med. 2014; 2014: 843036.
+
Dixit
S, Maiya
A. Diabetic peripheral neuropathy and its evaluation in a clinical scenario: A review. Postgrad. Med. J. 2014;60(1):33–40.
+
Donofrio
PD, Albers
JW. AAEM minimonograph 34: Polyneuropathy: Classification by nerve conduction studies and electromyography. Muscle and Nerve. 1990; 13(10): 889–903.
+
Duby
JJ, Campbell
RK, Setter
SM, White
JR, Rasmussen
KA. Diabetic neuropathy: an intensive review. American Journal of Health-System Pharmacy. 2004; 61 (2): 160–73, 175–6.
+
England
J, Asbury
AK. Peripheral neuropathy. Lancet, 2004; 363 (9427): 2151–2161.
+
England
JD, Gronseth
GS, Franklin
G, Carter
GT, Kinsella
LJ, Cohen
JA, Asbury
AK, Szigeti
K, Lupski
JR, Latov
N, Lewis
RA, Low
PA, Fisher
MA, Herrmann
DN, Howard
JF, Lauria
G, Miller
RG, Polydefkis
M, Sumner
AJ. Practice Parameter: evaluation of distal symmetric polyneuropathy: role of autonomic testing, nerve biopsy, and skin biopsy (an evidence-based review). Report of the American Academy of Neurology, American Association of Neuromuscular and Electrodiagnostic Medicine, and American Academy of Physical Medicine and Rehabilitation. Neurology. 2009; 72(2): 177–84.
+
Freeman
R. Autonomic peripheral neuropathy. Lancet. 2005; 365(9466): 1259–1270.
+
Ghasemi
M, Ansari
M, Basiri
K, Shaigannejad
V. The effects of intradermal botulinum toxin type a injections on pain symptoms of patients with diabetic neuropathy. J Res Med Sci. 2014;19(2):106–11.
+
Grisold
W, Cavaletti
G, Windebank
AJ. Peripheral neuropathies from chemotherapeutics and targeted agents: diagnosis, treatment, and prevention. Neuro-Oncology. 2012;14(Suppl 4):iv45–iv54.
+
Guillet
V, Guegen
N, Verny
C, Ferre
M, Homedan
C, Loiseau
D, Procaccio
V, Amati-Bonneau
P, Bonneau
D, Reynier
P, Chevrollier
A. Adenine nucleotide translocase is involved in a mitochondrial coupling defect in MFN2-related Charcot-Marie-Tooth type 2A disease. Neurogenetics. 2009. Accessed July 23,2009.
+
Hao
L, Nyland
J, Shelton
T. Effectiveness of the anodyne therapy system in treating diabetic peripheral neuropathy: a systematic review. Phys Ther Rev. 2008;13(6):395–404.
+
Harkless
LB, DeLellis
S, Carnegie
DH, Burke
TJ. Improved foot sensitivity and pain reduction in patients with peripheral neuropathy after treatment with monochromatic infrared photo energy-MIRE. Journal of Diabetes and its Complications. 2006; 20(2): 81–87.
+
Hershman
DL, Lacchetti
C, Loprinzi
CL. Prevention and Management of Chemotherapy Induced Peripheral Neuropathy in Survivors of Adult Cancers: American Society of Clinical Oncology Clinical Practice Guideline Summary. J Clin Oncol. 2014;10(6):421–424.
+
Hughes
BA. Peripheral neuropathy. BMJ: British Medical Journal. 2002; 324 (7335): 466–469.
+
Joa
KL, Kim
C. Clinical significance of the double-peak sensory response in nerve conduction study of normal and diabetic patients. Am J Phys Med Rehabil. 2013 Feb; 92(2): 111–7.
+
Lauria
G, Cornblath
DR, Johansson
O, McArthur
JC, Mellgren
SI, Nolano
M, Rosenberg
N, Sommer
C. EFNS guidelines on the use of skin biopsy in the diagnosis of peripheral neuropathy. European Journal of Neurology. 2005; 12: 747–758.
+
Lavery
LA, Murdoch
DP, Williams
J, Lavery
DC. Does anodyne light therapy improve peripheral neuropathy in diabetes? A double blind, sham-controlled, randomized trial to evaluate monochromatic infrared photoenergy. Diabetes Care. 2008; 31(2):316–21.
+
Lee
K, Lee
S, Song
C. Whole-body vibration training improves balance, muscle strength and glycosylated hemoglobin in elderly patients with diabetic neuropathy. Tohoku J Exp Med. 2013;231(4):305–314.
+
Malik
MM, Jindal
S, Bansal
S, Saxena
V, Shukla
US. Relevance of ankle reflex as a screening test for diabetic peripheral neuropathy. Indian J Endocrinol Metab. 2013;17(Suppl 1):S340–341. doi:10.4103/2230-8210.119641.
+
Marchettini
P. Painful peripheral neuropathies. Current Neuropharmacology. 2006;4(3):174–181.
+
Marrs
J, Newton
S. Updating your peripheral neuropathy 'know-how'. Clinical Journal of Oncology Nursing. 2003;7 (3): 299–303.
+
Massey
EW, Stolp
KA. Peripheral Neuropathy in Pregnancy. Phys Med Rehabil Clin N Am. 2008;19(1):149–162.
+
Mellion
ML, Silbermann
E, Gilchrist
JM, Machan
JT, Leggio
L, de la Monte
S. Small-Fiber Degeneration in Alcohol-Related Peripheral Neuropathy. Alcohol Clin Exp Res. 2014;38(7):1965–1972.
+
Meuller
MJ, Lott
DJ, Hastings
MK, Commean
PK, Smith
KE, Pilgram
TK. Efficacy and mechanism of orthotics devices to unload metatarsal heads in people with diabetes and a history of plantar ulcers. Phys Ther. 2006; 86(6): 833–42.
+
Nascimento
OJM. Leprosy neuropathy: clinical presentations. Arq Neuropsiquiatr. 2013;71(9B):661–666.
+
Nicholas
PK, Corless
IB, Evans
LA. Peripheral Neuropathy in HIV: An Analysis of Evidence-based Approaches. J Assoc Nurses AIDS Care. 2014;25(4):318–329.
+
Oh,
Shin J., Shoji
Hemmi, Yuki
Hatanaka. On-Nerve Needle Nerve Conduction Study in the Sural Nerve: A New Technique for Evaluation of Peripheral Neuropathy. Clin Neurophysiol. 2015;126(9):1811–16.
+
Olaleye
D, Perkins
BA, Bril
V. Evaluation of three screening tests and a risk assessment model for diagnosing peripheral neuropathy in the diabetes clinic. Diabetes Research and Clinical Practice. 2001; 54: 115–128.
+
Ownby
KK. Effects of ice massage on neuropathic pain in persons with AIDS. J Assoc Nurses AIDS Care. 2006;17(5): 15–22.
+
Ponirakis
G, Odriozola
M, Odriozola
S,
et al. NerveCheck: An inexpensive quantitative sensory testing device for patients with diabetic neuropathy. Diabetes Res Clin Pract. 2016;113:101–107.
+
Schröder
S, Liepert
J, Remppis
A, Greten
JH. Acupuncture Treatment Improves Nerve Conduction in Peripheral Neuropathy. Eur J Neurol. 2007;14(3):276–81.
+
Shin
S, Kim
KJ, Chang
H, Lee
B, Yang
W, Cha
B, Choi
D. The effect of oral prostaglandin analogue on painful diabetic neuropathy: A double-blind, randomized, controlled trial. Diabetes, Obesity and Metabolism. 2013; 15(2): 185–188.
+
Shoemaker
MJ. A Focused Exercise Regimen Improves Clinical Measures of Balance in Patients With Peripheral Neuropathy. Phys Ther. 2001;81(7):1362.
+
Singleton
JR, Marcus
RL, Jackson
JE, Lessard
KM, Graham
TE, Smith
AG, Exercise Increases Cutaneous Nerve Density in Diabetic Patients without Neuropathy. Ann Clin Transl Neurol. 2014;1(10):844–49.
+
Smith
AG, Singleton
JR. Impaired glucose tolerance and neuropathy. Neurologist. 2008; 14(1):23–9.
+
Spallone
V, Lacerenza
M, Rossi
A, Sicuteri
R, Marchettini
P. Painful diabetic polyneuropathy: approach to diagnosis and management. Clin J Pain. 2012; 28(8): 726–43.
+
Vinik
AI, Perrot
S, Vinik
EJ,
et al. Capsaicin 8% patch repeat treatment plus standard of care (SOC) versus SOC alone in painful diabetic peripheral neuropathy: a randomised, 52-week, open-label, safety study. BMC Neurol. 2016;16:251. doi:10.1186/s12883-016-0752-7.
+
Volkert
W, Hassan
A, Hassan
M. Effectiveness of monochromatic infrared photo energy and physical therapy for peripheral neuropathy: changes in sensation, pain, and balance -- a preliminary, multi-center study. Phys Occup Ther Geriatr. 2005;24(2):1–17.
+
Wood
WA, Wood
MA. Decompression of peripheral nerves for diabetic neuropathy in the lower extremity. The Journal of Foot and Ankle Surgery. 2003; 42(5): 268–275.
+
Zhao
W, Zeng
H, Zhang
X, Liu
F, Pan
J, Zhao
J, Zhao
J, Li
L, Bao
Y, Liu
F, Jia
W. A High Thyroid Stimulating Hormone Level Is Associated with Diabetic Peripheral Neuropathy in Type 2 Diabetes Patients. Diabetes Res Clin Pract. January 19, 2016. doi:10.1016/j.diabres.2016.01.018.
+
Ziegler
D. (2008). Treatment of diabetic neuropathy and neuropathic pain: How far have we come? Diabetes Care. 2008;31 Suppl 2:S255–261.
+
Ziegler
D, Hanefeld
M, Ruhnau
KJ, Meissner
HP, Lobisch
M, Schutte
K
et al.. Treatment of symptomatic diabetic peripheral neuropathy with the anti-oxidant alpha- lipoic acid. A 3-week multicentre randomized controlled trial (ALADIN Study). Diabetologia. 1995; 38: 1425–1423.
+++
Poliomyelitis / Post-polio Syndrome
++
Acute Poliomyelitis
Pathogenesis
Caused by an enterovirus transmitted via fecal-oral or oral-oral route among humans.
It replicates in oropharynx and intestinal tract (Chasens, 2000; Howard, 2005; McNaughton, 2003; Tebbens, 2005).
The virus remains in pharynx for 1-2 weeks and in feces >3-6 weeks (Halbritter, 2001).
80% to 90% of polio cases are accompanied by mild symptoms (Halbritter, 2001).
Paralysis occurs in 1 of 200 cases: t
The virus crosses into blood stream, invades the nervous system, and destroys anterior horn cells in the spinal cord and motor neurons of the brainstem. This will cause weakness or paralysis of related skeletal muscles (Chasens, 2000; Howard, 2005; McNaughton, 2003; Tebbens, 2005).
Prevalence
In 1988 there were an estimated 350,000 cases in 125 countries. Since then, the number of polio cases has decreased 99%. This reduction is the result of the global effort to eradicate the disease (World Health Organization, 2014).
As of January 2016, only Pakistan and Afghanistan remain polio-endemic.
Globally, only 72 cases were reported in 2015 (Patel, 2016).
As poliomyelitis nears eradication from the world, efforts need to be geared towards modifying or stopping immunization programs safely to prevent re-emergence (Minor, 2014).
Out of three strains, Type 2 wild poliovirus has been eradicated. It exists only in laboratories and in an attenuated form in the trivalent oral polio vaccine (tOPV).
A shift to the bivalent oral polio vaccine (bOPV) to combat both the Type 1 and Type 3 strains will begin the phased withdrawal of oral polio vaccine antigens (Patel, 2016).
Type 3 wild poliovirus has not been identified worldwide since 2012 (Jorba, 2016).
Vaccine-associated paralytic poliomyelitis (VAPP) may occur in individuals after OPV administration (Alexander, 2004).
Incidence of VAPP in the United States is 1 case per 900,000 doses of OPV administered (Burns, 2014).
Immunocompromised individuals are more likely to develop VAPP than those with normal immune systems (Alexander, 2004).
Vaccine-derived polioviruses (VDPVs) are types of polioviruses that are genetically different from wild polioviruses, divided into three types: circulating VDPVs (cVDPVs), immunodeficiency-associated VDPVs (iVDPVs), and ambiguous VDPVs (aVDPVs) (Burns, 2014).
cVDPVs are categorized when person-to-person transmission is detected in the environment (Burns, 2014).
iVDPVs are present in individuals with PID who have experienced prolonged VDPV infections (Burns, 2014).
aVDPVs are identified in patients with no known immunodeficiency or if the source of the VDPVs are unknown. (Burns, 2014).
Post Polio Syndrome (PPS)
Categorized by new neurological symptoms affecting polio survivors 15-30 years after an initial poliomyelitis infection (Gonzaleza, 2009).
Includes gradual or sudden onset of progressive and persistent new symptoms. The most common symptoms include
Muscle atrophy, generalized and muscular fatigue, progressive muscular weakness, pain from joint degeneration, and increasing skeletal deformity (Stolwijk-Swüste, 2007; National Institute of Health, 2015).
Cause unknown, but associated with severity during acute illness of limb, bulbar, or respiratory involvement; and overuse of muscles in intervening years (Howard, 2005; Jagannathan, 2002; Halbritter, 2001).
PPS can develop even if the patient experiences non-paralytic type of poliomyelitis (McKeever, 2012).
++
Acute Poliomyelitis
Signs/Symptoms:
Fever, fatigue, headache, nausea, vomiting (caused by virus invading GI tract); followed by, neck stiffness, limb pain, pharyngitis, myalgia, anorexia, severe muscle spasm, asymmetrical flaccid weakness or paralysis.
Bulbar poliomyelitis affects muscles of respiration, phonation, swallowing, facial expression and may cause death (Howard, 2005; McNaughton, 2003; Silver, 2001).
Hypertrophy in contralateral limb; compensation for affected muscles (Chasens, 2000).
The four types of poliomyelitis are differentiated by signs/symptoms. (McKeever, 2012)
Subclinical polio: flu-like symptoms, no CNS problems, no loss of PNS function
Abortive polio: low grade fever, headaches, mild CNS dysfunction, complete recovery
Non paralytic polio: stiff neck, CNS irritability, high grade fever
Paralytic polio: paralysis, stiff spine, and hypertension
Molecular diagnosis: technique of choice for identifying poliovirus serotype (Howard, 2005).
Differential diagnoses- must rule out factors leading to flaccid paralysis
Enteroviruses, neuropathy, heavy metal poisoning, myelitis, acute SC compression, trauma, infarction, myositis, myopathy, porphyria, diphtheria, Lyme disease, and disorders at the neuromuscular junction (Howard, 2005)
Because paralysis is a sign of polio, WHO monitors polio outbreaks by surveying cases of acute flaccid paralysis (AFP) (CDC, 2014).
WHO labs also test stool samples for polioviruses in individuals with suspected cases of polio (CDC, 2014).
Environmental surveillance is also utilized by testing sewage samples in areas with known polio outbreaks (CDC, 2014).
Post Polio Syndrome:
Signs/Symptoms:
New onset of muscle weakness; unaccustomed fatigue (occurs certain time of day; mostly late afternoon); joint and muscle pain; decreased endurance; skeletal deformities; muscle atrophy; fasciculations; bulbar or respiratory difficulties including aspiration; cold intolerance; sleep disorders; difficulty chewing and dysphagia; difficulty emptying the bladder; and urinary incontinence (Chasens, 2000; Gordon, 2002; Howard, 2005; McNaughton, 2003; Jagannathan, 2002; Ahlstrom, 2000; Koopman, 2010).
Pain
Pain reported for PPS is more nocioceptive in character, where neuropathic pain is usually due to a concomitant disease.
Women and younger patients report more frequent pain with PPS (Werhagen, 2010).
No specific diagnostic tests available (Fiorini, 2007)
Due to vague and non-specific subjective symptoms, PPS can be difficult to diagnose and it is a diagnosis of exclusion (Abrar, 2015).
Diagnostic criteria:
Symptoms listed above
Elimination of other possible causes such as: Parkinson's, radiculopathy, neuropathy, myopathy, spondylosis, ALS, and spinal stenosis
Meet PPS criteria
Prior known history of polio (usually documented by EMG)
Some initial strength improvements after paralysis
At least 15 years, typically 25-35 years, of stability (Chasens, 2000; McNaughton, 2003; Jubelt, 2000; Silver, 2001; Halbritter, 2001; Correa, 2008).
++
Poliomyelitis:
Non-progressive
Five phases of polio infection:
Signs of widespread denervation of muscle (month)
Motor neuron recovery and reinnervation (≈2 to 12 months)
Continued clinical recovery (≈2 to 8 years after infection)
Functional stability (≈8 to 40 years after infection)
Late deterioration (≈8 to 40 years after infection) (McNaughton, 2003).
Higher rates of mortality were observed in adult populations who contracted poliomyelitis between 1996-2012 (Mach, 2014).
Post Polio Syndrome:
25%-60% of polio survivors eventually develop PPS (Halbritter, 2001)
Survivors now between 50-60 yrs old
Symptoms and functional decline likely to increase.
Prognosis unaffected by sex, time since polio, symptom status, and residual weakness (Swust, 2005).
Fatigue is severe and persistent symptom that affects patients physically and psychologically (Tersteeg, 2011).
Prognosis can be affected by weight gain, muscle overuse, age, and medical comorbidities i.e. osteoporosis (Faraj, 2006; Laffont, 2010).
In patients with PPS, significant individual variation exists in the rate of decline of strength and walking capacity (Bickerstaffe, 2014).
In the assessment and treatment of gait abnormalities, muscle strength is only a weak to moderate predictor of gait performance (Flansbjer, 2013).
Carefully monitor symptoms and functional decline in this population, as there is a lack of predictive factors in substantial decline of mobility (Bickerstaffe, 2014).
+++
IV. Medical/Surgical Management:
++
Poliomyelitis
Once polio has occurred, no medications stop the progression.
To prevent worsening paralysis, bed rest and limited physical activity in acute phase (Howard, 2005).
Acute respiratory failure may require intubation, positive or negative pressure ventilation, or tracheostomy (Chasens, 2000, Howard 2005).
Vaccines:
Two vaccines:
Inactivated polio vaccine (IPV) "Salk" which was injected, and oral live attenuated "Sabin" polio vaccine (OPV) (Howard, 2005; McNaughton, 2003).
Worldwide eradication of virus not achieved possibly due to certain genetic factors and ineffectiveness of OPV in India (Paul, 2007).
The global risk of vaccine-associated paralytic poliomyelitis (associated with OPV) is 4.7 cases per million births (Platt, 2014).
Now that Type 2 eradication has been certified, the global transition from tOPV to bOPV has been approved and will occur between April 17th-May 1st of 2016 (Patel, 2016).
Sensitive surveillance and continued rapid response supplemental immunization activities (SIAs) are key to limiting further WPV spread (CDC, 2009)
Post Polio Syndrome:
No medications reverse the physiologic changes, but they can help relieve symptoms associated with post polio syndrome
For the treatment of fatigue:
Amantadine Hydrochloride (also an antiviral agent), Pyridostigmine Bromide, Pemoline.
For the treatment of depression:
Amitriptyline Hydrochloride, Fluoxetine Hydrochloride.
For the treatment of pain:
Amitriptyline Hydrochloride, NSAIDS (Chasens, 2000; McNaughton, 2003; Clinical Pharmacology, 2007; Silver, 2001).
IV immunoglobulin may decrease pain while increasing strength. (Kaponides, 2006)
For the treatment of musculoskeletal problems:
Surgery may be used for musculoskeletal problems (cervical stenosis, scoliosis, rotator cuff repair).
Possible techniques include: tendon transfer, muscle transplant, arthrodesis, limb lengthening, and joint replacement (Provelengios, 2009, Faraj, 2006).
Intravenous Immunoglobulin (IVIG) therapy decreases inflammatory cytokines and in turn may improve quality of life, functional outcomes, and pain ratings. (Gonzalez, 2012).
+++
V. Implications for Therapeutic Management
++
+++
VI. Consumer and Professional Resources
++
Information for those affected by polio or PPS, newsletters, locating a PPS support group, information for healthcare professionals www.post-polio.org
+
Abrar
A, Ahmad
A. Post Poliomyelitis Syndrome: A Rare Sequel of Acute Poliomyelitis. J Pak Med Assoc. 2015;65(3):327–329.
+
Ahlström
G, Karlsson
U. Disability and quality of life in individuals with postpolio syndrome. Disability and Rehabilitation. 2000; 22: 416–422.
+
Appelin
K, Lexell
J, Månsson
Lexell E. Occupations that People with Late Effects of Polio Perceive Difficult to Perform. Occup Ther Int. 2014;21(3):98–107.
+
Bickerstaffe
A, Beelen
A, Nollet
F. Change in physical mobility over 10 years in post-polio syndrome. Neuromuscular Disorders. doi:10.1016/j.nmd.2014.11.015.
+
Bocci
T, Acler
M, Vergari
M, Barbieri
S, Priori
A, Bertolasi
L. Transcranial direct current stimulation (tDCS) for fatigue in patients with post-polio syndrome. Clin Neurophysiol. 2013;124(10):e180–e180.
+
Boyer
FC, Auyeung
V, Luff
R, Holland
M, Hodgkiss
A, Weinman
J. Post-polio syndrome: Pathophysiological hypotheses, diagnosis criteria, medication therapeutics. Ann Phys Rehabil Med. 2010;53(1):34–41.
+
Burns
CC, Diop
OM, Sutter
RW, Kew
OM. Vaccine-Derived Polioviruses. J Infect Dis. 2014;210(suppl_1):S283–S293.
+
Center for Disease Control. Mmwr Morb Mortal Wkly Rep, 2009; 58(14): 357–62.
+
Center for Disease Control. Mmwr Morb Mortal Wkly Rep, 2014; 63(16): 356–361.
+
Chasens
ER, Umlauf
MG. Post-Polio Syndrome.
American Journal of Nursing. 2000; 100: 60–61, 63–65. Clinical Pharmacology [database online]. Gold Standard Incorporated, 2007. Available at:
http://cpip.gsm.com. Accessed April 14, 2007.
+
Correa
JC,
et al. Electromyographic and neuromuscular analysis in patients with post-polio syndrome. Electromyogr Clin Neurophysiol ]. 2008;48(8):329–33.
+
Davidson
AC,
et al. Prolonged benefit in post-polio syndrome from comprehensive rehabilitation: a pilot study. Disabil Rehabil ]. 2009;31(4):309–17.
+
Dencker
A, Sunnerhagen
KS, Taft
C, Lundgren-Nilsson
Å. Multidimensional Fatigue Inventory and Post-Polio Syndrome – A Rasch Analysis. Health Qual Life Outcomes. 2015;13(20). doi:10.1186/s12955-015-0213-9.
+
Faraj
A. Poliomyelitis: Orthopaedic Management. Current Orthopaedics. 2006;20(1):41–46.
+
Fiorini
M, Zanusso
G, Baj
A, Bertolasi
L, Toniolo
A, Monaco
S. Post-polio syndrome: Clinical manifestations and cerebrospinal fluid markers. Future Neurology. 2007;2(1):451–463.
+
Flansbjer
U-B, Brogårdh
C, Lexell
J. Muscle strength is only a weak to moderate predictor of gait performance in persons with late effects of polio. NeuroRehabil. 2013;33(3):457–464.
+
Gonzalez
H, Khademi
M, Borg
K, Olsson
T. Intravenous immunoglobulin treatment of the post-polio syndrome: sustained effects on quality of life variables and cytokine expression after one year follow up. J Neuroinflammation. 2012;9(1):167–178.
+
Gordon
PA, Feldman
D. Post-polio syndrome: Issues and strategies for rehabilitation counselors. Journal of Rehabiliation. 2002;68(2) 28–32.
+
Halbritter
T. Management of a patient with post-polio syndrome. Journal of American Academy of Nurse Practioners. 2001;13:12.
+
Halstead
L. A Brief History of Postpolio Syndrome in the United States. Arch Phys edD Rehabil. 2011;92(8):1344–1349.
+
Howard
RS. Poliomyelitis and the postpolio syndrome. British Medical Journal. 2005; 330:1314–1318.
+
Jagannathan
NR, Wadhwa
S. In vivo proton magnetic resonance spectroscopy (MRS) study of post polio residual paralysis (PPRP) patients. Magnetic Resonance Imaging. 2002; 20: 113–117.
+
Jones
K, Grenstad
T, Dreyer
V. Perceived disability, fatigue, pain and measured isometric muscle strength in patients with post-polio symptoms. Physiotherapy Research International. 2007; 12: 39–49.
+
Jorba
J, Diop
OM, Iber
J, Sutter
RW, Wassilak
SG, Burns
CC. Update on Vaccine-Derived Polioviruses — Worldwide, January 2015–May 2016. MMWR Morb Mortal Wkly Rep. 2016; 65:763–769.
+
Jubelt
B, Agre
JC. Characteristics and management of postpolio syndrome. Journal of the American Medical Association. 2000; 284: 44,412–414.
+
Kaonides
G, Gonzalez
H, Olsson
T, Borg
K. Effect of intravenous immunoglobulin in patients with post-polio syndrome -- an uncontrolled pilot study. J Rehabil Med. 2006; 38(2): 138–40.
+
Khan
F. Rehabilitation for postpolio sequelae. Australian Family Physician. 2004;33(8):621–624.
+
Khan
S, Gajapathi
M, Pandit
M. Additive effect of Pulsed Electromagnetic Field Therapy (PEMF) on Hip Flexor Contracture in Post Polio Syndrome (PPS). Indian J Physiother Occup Ther. 2013;7(2):191–196.
+
Klein
MG, Whyte
J, Esquenazi
A, Keenan
MA, Costello
R. A comparison of the effects of exercise and lifestyle modification on the resolution of overuse symptoms of the shoulder in polio survivors: a preliminary study. Arch Phys Med Rehabil. 2002; 83: 708–713.
+
Laffont
I, Julia
M, Tiffreau
V, Yelnik
A, Herisson
C, Pelissiee
J. Aging and sequelae of poliomyelitis. Ann Phys Rehabil Med. 2010;5(1):24–33.
+
Mach
O, Tangermann
RH, Wassilak
SG, Singh
S, Sutter
RW. Outbreaks of paralytic poliomyelitis during 1996-2012: the changing epidemiology of a disease in the final stages of eradication. J Infect Dis. 2014;210 Suppl 1:S275–S282.
+
McKeever
AE, Young
JM, Suplee
PD. Post-Polio Syndrome: A Rising Health Problem Among Women. Am J Nurse Pract. 2012;16(9/10):6–10.
+
McNaughton
HK, McPherson
KM. Problems occurring late after poliomyelitis: a rehabilitation approach. Critical ReviewsTM in Physical and Rehabilitation Medicine. 2003; 15: 295–308.
+
Minor
P. The polio endgame. Human Vaccines & Immunotherapeutics. 2014;10(7):2106–2108.
+
Oncu
J, Durmaz
B, Karapoalat
H. Short-term effects of aerobic exercise on functional capacity, fatigue, and quality of life in patients with post-polio syndrome. Clin Rehabil. 2009 Feb;23(2):155–63.
+
Patel
M, Orenstein
W. A World Free of Polio — The Final Steps. NEJM. 2016;374(6):501–503.
+
Paul
Y. Role of genetic factors in polio eradication: new challenge for policy makers. Vaccine. 2007; 25:50: 8365–8371.
+
Platt
LR, Estívariz
CF, Sutter
RW. Vaccine-associated paralytic poliomyelitis: a review of the epidemiology and estimation of the global burden. J Infect Dis. 2014;210 Suppl 1:S380–S389.
+
Provelengios
S. The Role of Pantalar Arthrodesis in the Treatment of Paralytic Foot Deformities. A Long-Term Follow-up Study. J Bone Joint Surg. 2009 Mar; 91(3): 575–83.
+
Sharma
SS, Sheth
MS, Vyas
NJ. Fatigue and Functional Capacity in Persons with Post-Polio Syndrome: Short-term Effects of Exercise and Lifestyle Modification Compared to Lifestyle Modification Alone. Disability, CBR & Inclusive Development. 2014;25(3):78–91.
+
Silver,
J. Post-Polio Syndrome: A Guide for Polio Survivors and Their Families. Yale University Press. 2001.
+
Spiliotopoulou
G, Fowkes
C, Atwal
A. Assistive technology and prediction of happiness in people with post-polio syndrome. Disabil Rehabil Assist Technol. 2012;7(3):199–204.
+
Stolwijk-QSwüste
JM, Beelen
A, Gustaff
J, Lannkhorst
N, Frans
N. Impact of age and co-morbidity on the functioning of patients with sequelae of poliomyelitis: a cross-sectional study. J Rehabil Med. 2007;39(1):56–62.
+
Swuste
J, Beelen
A, Lankhorst
G, Nollet
F, CARPA Study Group. The course of functional status and muscle strength in patients with late-onset sequelae of poliomyelitis: a systematic review. Arch Phys Med Rehabil. 2005; 86: 1693–1701.
+
Tebbens
RJD
et al.. A dynamic model of poliomyelitis outbreaks: learning from the past to help inform the future. American Journal of Epidemiology. 2005; 162:358–372.
+
Tersteeg
IM, Koopman
FS, Stolwijk-Swüste
JM, Beelen
A, Nollet
F. A 5-Year Longitudinal Study of Fatigue in Patients With Late-Onset Sequelae of Poliomyelitis. Arch Phys Med Rehabil. 2011;92(6):899–904.
+
Vreede
KS, Henriksson
J, Borg
K, Henriksson
M. Gait characteristics and influence of fatigue during the 6-minute walk test in patients with post-polio syndrome. J Rehabil Med. 2013;45(9):924–928.
+
Werhagen
L, Borg
K. Analysis of long-standing nociceptive and neuropathic pain in patients with post-polio syndrome. Journal of neurology. 2010; 257: 1027–1031.
+
Werhagen
L, Borg
K. Impact of pain on quality of life in patients with post-polio syndrome. J Rehabil Med. 2013;45(2):161–163.
+
Young
CA, Quincey
A-MC, Wong
SM, Tennant
A. Quality of life for post-polio syndrome: a patient derived, Rasch standard scale. Disability and Rehabilitation. 2016;0(0):1–6. doi:10.1080/09638288.2016.1260650.
+++
Prader Willi Syndrome
++
Prader-Willi syndrome (PWS) is a complex multi-system genetic disorder affecting growth, appetite, metabolism, cognitive function and behavior as well as a variety of other clinical manifestations. A major finding is insatiable hunger (hyperphagia) and slowed metabolism leading to life-threatening obesity (Cassidy, 2012).
PWS is a genetic syndrome that results in failure of expression of the paternal genes in chromosome 15q11.2-q13; types of defect include microdeletion (in 70%), uniparental maternal disomy 15 (in 25%), imprinting defects (in 2-5%) and structural chromosome rearrangement (in 1%); genetic defect seems to occur randomly, without known factors that increase risk (Verhoeven, 2003; Cassidy, 2012; Bittel, 2003; McCandless, 2010)
PWS is not associated with any specific ethnic group, geographic region, or socioeconomic class and affects males and females almost equally. It can occur in any conception and is not usually inherited (Lang, 2010)
Prevalence: one in 10,000 - 30,000 live births (Cassidy, 2012)
Pathogenesis
The genetic material on chromosome 15 may alter synthesis, release, metabolism, binding, intrinsic activity, or reuptake of specific neurotransmitters (Dimitropoulos , 2000)
Hypothalamic dysfunction seems to be central to many of the problems seen in PWS: (Verhoeven, 2003; Swaab, 1997; Burman, 2001)
Fetal hypothalamic systems may explain fetal hypotonia; decreased fetal movement; frequent abnormal fetal position at delivery; premature or post-mature onset of labor; increased incidence of assisted or cesarean section delivery; and high percentage of asphyxia during birth
Decreased levels of gonadotropins result in hypogonadism, incomplete onset of puberty and infertility in both sexes
Decreased levels of growth hormones result in short stature and small feet/hands
Abnormal control of body temperature and variety of sleep disturbances
Decreased number of oxytocin neurons (putative satiety neurons) in the hypothalamic paraventricular nucleus source of insatiable hunger and obesity
Neuroimaging studies reveal variable brain abnormalities of undetermined significance (Miller, 2007)
++
Signs and Symptoms (Cassidy, 2012; Thompson, 2006; Goldstone, 2004; Elena, 2012; Holm, 1993; Lang, 2010; Verhoeven, 2003)
Hypotonia a consistent finding in fetus and infant with secondary weak cry, diminished reflexes including poor suck reflex, decreased spontaneous movement and arousal; hypotonia gradually decreases with maturity
Developmental delay (in 90-100%)
Gross and fine motor delays
In population of 4650 kindergarten children, those identified as obese had lower gross motor skills, especially locomotor and balance skills (Roberts, 2012)
Language – expressive language a relative strength; articulation problems common
Cognitive - mean intelligence quotient (IQ) 60-70
Severe learning disabilities and academic performance lower than would be expected based on IQ
Hyperphagia and obesity
Seven nutritional phases identified
Phase 0 (in utero) – hypotonia, growth restriction and decreased movements
Phase 1a (birth to 15 months: median age 9 months) – hypotonia, difficulty feeding, possible failure to thrive
Phase 1b (5 to15 months with median age of onset 9 months) – steady growth with weight progressing at normal rate
Phase 2a (age of onset 2 years) – weight increases without atypical appetite or caloric intake
Phase 2b (age of onset about 4.5 years) – weight gain with increased interest in food
Phase 3 (age of onset about 8 years) – hyperphagia (insatiable appetite)
Phase 4 (in some adults) – insatiable appetite and weight gain decreases
Associated behavioral characteristics: food hoarding or foraging, eating inedibles, stealing (money or food)
Decreased resting energy expenditure secondary to decreased physical activity, decreased lean body mass and possibly hormonal influence
Elevated ghrelin levels (produced in stomach/regulated by hypothalamus) • Complications of obesity:
Dysmorphic features (may evolve over time): narrow bifrontal diameter, almond-shaped palpebral fissures, narrow nasal bridge, thin upper lip, down-turned corners of mouth, slender hands with hypoplastic ulnar bulge, tapered fingers and puffy dorsum of palm and fingers; hypopigmentation of hair, eyes and skin with specific genetic configuration
Behavioral and psychiatric disturbances (in 70-90%) starting in early childhood: temper tantrums, stubbornness, controlling and manipulative behaviors, compulsivity, difficulty with change in routine; repetitive and ritualistic behaviors (Gleaves, 2006)
Hypogonadism in both sexes with genital hypoplasia, incomplete pubertal development and infertility
Short stature and growth hormone deficiency; small hands and feet
Sleep disorders: reduced rapid eye movement latency, altered sleep patterns, central and obstructive sleep apnea; daytime sleepiness resembling narcolepsy (Cassidy, 2012); across a selection of research studies involving 224 children with PWS, prevalence of OSA was 79.91%; and narcolepsy was 35.71% (Sedky, 2014)
Physical findings:
Hip dysplasia (in approximately 10-20%); scoliosis (in 40-80%) without relationship to growth hormone supplementation; osteopenia and secondary increase in fractures (Shim, 2010)
Abnormal kinematics in knees secondary to obesity (stand in more valgus and have altered knee loading patterns during walking and jogging) (Briggs, 2017)
Genu valgum and flat feet in children who are obese (Jankowicz-Szymanska, 2016)
Increased base of support while walking, decreased balance, and decreased leg explosive strength (Pathare, 2013)
Other variable findings: strabismus (in 60-70%); leg edema and ulceration (especially with obesity); skin picking; altered temperature sensation; decreased saliva flow and thick saliva; high vomiting threshold; seizures (in 10-20%); central adrenal insufficiency; hypothyroidism (in up to 25%); impaired glucose tolerance and diabetes mellitus (up to 25% with mean age of onset 20 years); recurrent respiratory infections (up to 50%)
Diagnostic Testing (Cassidy, 2012; Kubota, 1996)
Important to recognize symptoms to facilitate early diagnosis and intervention (McCandless, 2011)
Clinical diagnostic criteria using a numeric scale is useful first step (Holm, 1993)
DNA methylation analysis will confirm diagnosis in 99% of cases
Once diagnosis confirmed, determination of molecular class is necessary for genetic counseling and genotype-phenotype correlation
+++
III. Prognosis and Sequelae
++
Trends of Progression – refer to nutritional phases previously outlined
Expected Sequelae (secondary impairments, structural and functional)
+++
IV. Medical/Surgical Management – no cure; management of complex symptoms; varies across lifespan
++
Medical/Pharmaceutical
Growth hormones (GH) treatment shown to have beneficial effects
Increase muscle mass and metabolism, normalize height and weight, increase energy levels, improve respiratory function, and increase bone density (Yearwood, 2011; Cassidy, 2012)
Prevent deterioration of certain cognitive skills short term; significantly improve abstract reasoning and visuospatial skills during 4 years of GH treatment; more benefit for children with more severe deficit (Elbrich, 2012)
One year of GH treatment significantly improved mental and motor development in infant and toddlers with PWS; treatment group median age of 2.3 years; control group median age of 1.5 years; assessment with Bayley Scales of Infant and Toddler Development II (Festen, 2008)
Cessation of GH treatment resulted in increased percent body fat, decreased IGF-1 and HDL levels, and decreased moderate-vigorous physical activity back to baseline after suggesting GH treatment may need to be continuous (Butler, 2013)
GH therapy for 24 months with 35 children with PWS resulted in sustained increases in lean body mass, decreases in percent body fat, improvements in physical strength and agility, and increased fat oxidation; slowed growth rate between 12 and 24 months suggests more research needed on the long term value of GH therapy (Myers, 2000)
Sex hormone replacement therapy may be considered (as early as 6 months) to address hypogonadism, pubertal deficiencies and osteopenia/osteoporosis with added benefit of improving muscle mass and strength in males (Yearwood, 2011; Cassidy, 2012)
Selective serotonin uptake inhibitors can be effective for psychoses; not mood stabilizers (Cassidy, 2012)
Medications (e.g., Fluoxetine) being tried to treat obesity and reduce binge eating episodes and caloric intake (Salehi, 2015)
Other medications may include stool softeners, fiber supplements or motility agents for gastrointestinal issues; medications for high blood pressure, diabetes, osteoporosis and other clinical symptoms, as indicated (Yearwood, 2011)
Evaluation of sleep disturbance
Dry mouth associated with decreased saliva production can be addressed with special toothpastes, gels, mouthwash and gum (Cassidy, 2012)
Surgical
Gastric bypass for morbid obesity (Donaldson, 1994)
Tonsillectomy and adenoidectomy shown to improve obstructive sleep apnea (OSA) but some residual OSA remained in majority of cases post-op (Sedky, 2014)
Orthopedic surgery for joint or spine deformities as indicated
+++
V. Key Aspects of Therapeutic Management
++
Multidisciplinary approach is essential, including primary care physician, genetic counselor, dietician, psychologist, physical therapist, occupational therapist, speech and language pathologist, special educator, and endrocrinologist (Lewis, 2000); focus on three areas – education, obesity management and behavioral management (Cassidy, 2012)
Patient and caregiver education addressed by all team members
High rehabilitation priorities for parents are eating issues and adaptive functioning deficits (Pituch, 2010)
Genetic counseling – majority of families have recurrence risk of less than 1% but various scenarios (Cassidy, 2012)
Individualized education program in a multidisciplinary team approach coordinated by special education and classroom teachers to address cognitive, behavioral, social-adaptive, language and mobility skills.
Nutritional support/consultation for adequate caloric intake and feeding in infancy and weight loss, adequacy of vitamin and mineral intake and other dietary issues in childhood/adolescence and adulthood.
Speech therapy for articulation abnormalities and speech/language delays
Physical therapy to address motor delays: prevent musculoskeletal problems; increase muscle strength, aerobic endurance, postural control, movement efficiency, and function, and address issues related to obesity (Lewis, 2000)
Children and adolescents with obesity exhibited increased strength and decreased percent body fat and body mass index after participating in resistance training protocols (Lewis, 2000)
Aquatic rehabilitation has been shown to improve walking endurance, lower extremity strength and pain while promoting physical activity in protective environment for joints (Fragal-Pinkham, 2009)
Prader-Willi Rehabilitation comprehensive treatment cycles (26 day program repeated 4 times per year) has shown significant weight loss; program consists of stationary bike exercise, step activities, mat exercises, walking, psychomotor or music therapy and recreational activities; diet of 1500 kilocalories with emphasis on raw fruits and vegetables and unrestricted water intake; appropriate rest times to accommodate fatigue and minimize stress (Grolla, 2011)
+
Bittel
DC, Kibiryeva
N, Butler
MG. Expression of 4 genes between chromosome 15 breakpoints 1 and 2 and behavioral outcomes in Prader-Willi syndrome. Pediatrics. 2006;118:1276–1283.
+
Briggs
MS, Bout-Tabaku
S, McNally
MP,
et al. Relationship between standing frontal-plane knee alignment and dynamic knee joint loading during walking and jogging in youth who are obese. Phys Ther. 2017;97(5):571–580.
+
Burman
P, Ritzén
EM, Lindgren
AC. Endocrine dysfunction in Prader-Willi syndrome: a review with special reference to growth hormone. Endocr Rev. 2001;22:787–799.
+
Butler
MG, Smith
BK, Lee
J,
et al. Effects of growth hormone treatment in adults with Prader-Willi syndrome. Growth Horm IGF Res. 2013;23(3):81–87.
+
Cassidy
SB, Schwartz
S, Miller
JL, Driscoll
DJ. Prader-Willi syndrome. Genetics Med. 2012;14(1):10–25.
+
Dimitropoulos
A, Feurer
ID, Roof
E,
et al. Appetitive behavior, compulsivity, and neurochemistry in Prader-Willi syndrome. Mental Retard Dev Disabil Res Rev. 2000;6:125–130.
+
Donaldson
M, Chu
C, Cooke
A, Wilson
A, Greene
S, Stephenson
J. The Prader-Willi syndrome. Arch Dis Childhood. 1994;70(1):58–63.
+
Elbrich
PC, Siemensma
Roderick FA, Tummers-de
Lind van WijngaardenDederieke AM,
et al. Beneficial effects of growth hormone treatment on cognition in children with Prader-Willi syndrome: a randomized controlled trial and longitudinal study. J Clin Endocr Metab. 2012;97(7):2307–2314.
+
Elena
G, Bruna
C, Benedetta
M, Stefania
DC, Giuseppe
C. Prader-willi syndrome: clinical aspects. J Obes. 2012;2012:1–13.
+
Festen
DA, Wevers
M, Lindgren
AC,
et al. Mental and motor development before and during growth hormone treatment in infants and toddlers with Prader-Willi syndrome. Clin Endocrino. 2008;68:919–925.
+
Fragala-Pinkham
M, Dumas
H, Barlow
C, Pasternak
A. An aquatic physical therapy program at a pediatric rehabilitation hospital: a case series. Pediatr Phys Ther. 2009;21(1):68–78.
+
Goldstone
AP. Prader-Willi syndrome: advances in genetics, pathophysiology and treatment. Trends Endocr Metab. 2004;15:12–20.
+
Greaves
N, Prince
E, Evans
DW, Charman
T. Repetitive and ritualistic behavior in children with Prader-Willi syndrome and children with autism. J Intel Disabil Res. 2006;50:92–100.
+
Grolla
E, Andrighetto
G, Dolcetta
D,
et al. Specific treatment of Prader-Willi syndrome through cyclical rehabilitation programmes. Disabil Rehabil. 2011;33(19/20):1837–1847.
+
Holm
VA, Cassidy
SB, Butler
MG,
et al. Prader-Willi syndrome: consensus diagnostic criteria. Pediatrics. 1993;91(2):398–402.
+
Jankowicz-Szymanska
A, Mikolajczyk
E. Genu valgum and flat feet in children with healthy and excessive body weight. Pediatr Phys Ther. 2016;28(2):200–2006.
+
Kubota
T, Sutcliffe
JS, Aradhya
S,
et al. Validation studies of SNRPN methylation as a diagnostic test for Prader-Willi syndrome. Am J Med Genet. 1996;66(1):77–80.
+
Lang
R, Smith
W, Ence
W, Langthorne
P. Rehabilitation issues for children with Prader-Willi syndrome. Dev Neurorehabil. 2010;13(1):1–2.
+
Lewis
C. Prader-Willi syndrome: a review for pediatric physical therapists. Pediatr Phys Ther. 2000;12(2):87–95.
+
McCandless
SE, Committee on Genetics: clinical report-health supervision for children with Prader-Willi syndrome. Pediatrics, 2011;127(1):195–204.
+
Myers
SE, Carrel
AL, Whitman
BY, Allen
DB. Sustained benefit after 2 years of growth hormone on body composition, fat utilization, physical strength and agility, and growth in Prader-Willi syndrome. J Pediatr. 2000;137(1):42–49.
+
Miller
JL, Couch
JA, Schmalfuss
I, He
G, Liu
Y, Driscoll
DJ. Intracranial abnormalities detected by three-dimensional magnetic resonance imaging in Prader-Willi syndrome. Am J Med Genet A. 2007;143A(5):476–483.
+
Pathare
N, Haskvitz
E, Selleck
M. Comparison of measures of physical performance among young children who are healthy weight, overweight or obese. Pediatr Phys Ther. 2013;25(3):291–296.
+
Pituch
K, Green
V, Sigafoos
J,
et al. Rehabilitation priorities for individuals with Prader-Willi Syndrome. Disabil Rehabil. 2010;32(24):2009–2018.
+
Roberts
D, Veneri
D, Decker
R, Gannotti
M. Weight status and gross motor skill in kindergarten children. Pediatr Phys Ther. 2012;24(4):353–360.
+
Salehi
P, Leavitt
A, Beck
AE, Chen
ML, Roth
CL. Obesity management in Prader-Willi syndrome. Pediatr Enocrinol Rev. 2015;12(3):297–307.
+
Sedky
K, Bennett
DS, Pumariega
A. Prader Willi syndrome and obstructive sleep apnea: co-occurrence in the pediatric population. J Clin Sleep Med. 2014;10(4):403–409.
+
Shim
J, Lee
S, Seo
S, Koo
K, Jin
D. The musculoskeletal manifestations of Prader-Willi syndrome. J Pediatr Orthoped. 2010;30(4):390–395.
+
Steinberg
N, Rubinstein
M, Nemet
D,
et al. Effects of a program for improving biomechanical characteristics during walking and running in children who are obese. Pediatr Phys Ther. 2017;29(4):330–340.
+
Swaab
DF. Prader-Willi syndrome and the hypothalamus. Acta Paediatr Suppl. 1997;423:50–54.
+
Thompson
A, Glasson
E, Bittles
A. A long-term population-based clinical and morbidity review of Prader-Willi syndrome in Western Australia. J Intellectual Disabil Res. 2006;50:69–78.
+
Verhoeven
WMA, Tuinier
S, Curfs
LMG. Prader–Willi syndrome: cycloid psychosis in a genetic subtype? Acta Neuropyschiatrica. 2003;15:32–37.
+
Wills
M. Orthopedic complications of childhood obesity. Pediatr Phys Ther. 2004;16(4):230–235.
+
Yearwood
E, McCulloch
M, Tucker
M, Riley
J. Care of the patient with Prader-Willi syndrome. Medsurg Nsg. 2011;20(3):113–122.
+++
Progressive Bulbar Palsy
++
Progressive bulbar palsy (PBP) is a chronic, progressive fatal disease
PBP is characterized by bulbar dysfunction and motor neuron degeneration (Knott, 2015)
ALS can include UMN involvement (pseudobulbar palsy or supranuclear palsy) and LMN involvement (bulbar palsy) (Kühnlein, 2008).
Pseudobulbar Palsy will most commonly present with UMN signs including increased tone and exaggerated reflexes (jaw jerk) (Knott, 2015)
Progressive Supranuclear Palsy is characterized by neurodegeneration in the basal ganglia, brainstem, prefrontal cortex, and cerebellum and can result in bulbar signs and symptoms (Zampieri, 2006)
About 75% of individuals with classic ALS eventually show some bulbar involvement
Approximately 25% of individuals with ALS have early symptoms that begin with bulbar involvement (NINDS, 2013; Kernich, 2009)
ALS with Bulbar symptoms at diagnosis has worse prognosis than spinal onset ALS (Zoccolella, 2008)
Primary bulbar palsy type ALS (ALS-PBP) is associated with decreased cognitive functioning
Patients with ALS-PBP scored significantly lower on the Hasegawa Dementia Scale- Revised and the Frontal Assessment Battery than control subjects and patients with limb type ALS (Morimoto, 2012)
PBP by itself (no arm/leg involvement) is extremely rare. (NINDS, 2013)
PBP can be classified into two different categories (Karam, 2010)
Demographics:
Mostly occurs after 40 years of age and more often in men than women (NINDS, 2013; Rubin, 2008).
It is commonly seen in late adulthood and seen to a lesser extent in younger adults and children.
Onset of the disease in patients of all ages is insidious.
PBP is idiopathic like other motor neuron diseases and it has no clear cause (NINDS, 2013; Rubin, 2008)
Pathogenesis
PBP is a LMN disease involving progressive atrophy and paralysis of the muscles of the lips, tongue, mouth, pharynx, and larynx due to lesions of the motor nuclei of the lower brain stem.
Motor nuclei of cranial nerves V, VII, IX, X, and XII gradually atrophy while the glial cells overgrow (gliosis)
There is also subcortical involvement of the corticobulbar tracts.
Most cases will progress to ALS. (Karam, 2010).
Genetic Factors
Fazio-Londe-Syndrome and Brown-Vialetto-Van Laere syndrome (BVVL) occur in children and young adults (Hawkins, 1990)
Both are allelic disorders and involve mutations of the SLC52A2 and SLC52A3 genes (Manole 2014).
For FLS and BVVL, riboflavin treatment greatly increases survival rate (Bosch, 2012)
Fazio-Londe-Syndrome (FLS):
Patients have less reports of sensory deafness
Greater mortality
Survive three years from diagnosis, suggesting more severe genetic recessive genotype (Sathasivam, 2008)
Autosomal recessive or autosomal dominant inheritance (Manole, 2014).
Brown-Vialetto-Van Laere syndrome (BVVL) BVVL:
Periods of gradual deterioration, interrupted with periods of stability or abrupt worsening of symptoms
Those with BVVL generally survive up to 5-10 years after the diagnosis (Sathasivam, 2008).
Suggested disease is genetically heterogeneous and autosomal recessive inheritance.
Possible X chromosome mutation (Galllai, 1981; Hawkins, 1990; Megarbane, 2000; Ramussen, 1994).
Possible autoimmune origin has been suggested (Megarbane, 1990).
Mutation in the SLC52A2 gene (a riboflavin transporter that has a role in neuron maintenance and homeaostasis) is thought to cause interruption of the energy pathway in motor neurons therefore leading to BVVL and further, Progressive Bulbar Palsy. (Timmerman, 2014).
The development of PBP can occur secondary to:
Motor Neuron Disease, Guillain Barre Syndrome, Syringobulbia, Amyotrophic Lateral Sclerosis (ALS), Multiple Sclerosis (MS), Poliomyelitis, Diphtheria, Myasthenia Gravis, Neurosyphilis
++
Symptoms
Progressive weakening of the muscles innervated by the cranial nerves V, VII, IX, X, XII (more rarely CN's III and VI), and corticobulbar tracts (Gallai, 1981)
Nasal regurgitation, drooling, difficulty chewing, facial weakness, and tongue weakness (Lapiedra, 2002, Megarbane, 2000)
Dystussia, a cough deficit, puts patients at risk for aspiration and respiratory failure (Britton, 2014)
Laryngeal dysfunction is common in individual's with Bulbar ALS
Changes in voice to a soft, weak, low-pitched, and monotonous voice occur from flaccid paresis of laryngeal muscles (Kühnlein, 2008)
Pharyngeal muscle weakness leading to dysphagia (NINDS, 2016)
Respiratory difficulty may occur before limb weakness (Karam, 2010; Lapiedra, 2002; NINDS, 2013)
Signs: lesions of both upper and lower motor neurons may be present
Slurred speech, swallowing difficulties (Kent, 2013)
Speech changes (NINDS, 2013; Lapiedra, 2002) most commonly dysarthria (93%), dysphagia (86%) (Kühnlein, 2008)
Dysarthria is 8x more common than dysphagia as an initial symptom (Traynor, 2000)
Oropharyngeal assessment by speech therapist to evaluate for dysphagia (Medscape, 2013)
Videofluoroscopic swallowing evaluation to check for swallowing problems (Medscape, 2013)
Emotional lability
There may be a pseudobulbar emotional response, with labile or inappropriate emotion
Pseudobulbar Affect- a neurologic condition characterized by sudden outbursts of uncontrollable crying or laughing (Krivickas, 2003)
Vocal cord paralysis (Ertekin, 2000), and resulting dysphonia (Knott, 2015)
Laryngospasms (Kühnlein, 2008)
Spasticity of the upper extremities (Megarbane, 2004)
Opthalamoplegia is a pseudobulbar or supranuclear sign (Megarbane, 2004; Armstrong, 2010)
Fasciculation of the tongue and lip muscles (Lapiedra, 2002)
Atrophy of tongue (NINDS, 2013)
Upper airway clearance initiation and effectiveness diminished (Garcia-Pachon, 1994)
Abnormal reflexes:
Hyporeflexic, and eventually absent, swallowing, jaw jerk and gag (Megarbane, 2004; Ertekin, 2000; Leighton, 1994)
Hyperactive mandibular reflex (Ertekin, 2000)
Hyporeflexic diaphragmatic recoil (Ertekin, 2000)
Hyperactive esophageal reflex (Ertekin, 2000, Leighton, 1994)
Variable reflex demonstration of UMN and LMN activity (Ertekin, 2000; Lapiedra, 2002; NINDS, 2013)
Based on the above clinical signs and symptoms the following should be considered for differential diagnoses (Krivickas, 2003):
Myasthenia Gravis, Multiple Sclerosis, Foramen Magnum Tumor, Brainstem Glioma, Stroke, Syringobulbia, Head and Neck Cancer, Polymyositis, Kennedy's Disease
Diagnostic Procedures
Few diagnostic tests exist for PBP (Evans, 2001).
Diagnosis of PBP is made in the absence of non-bulbar clinical signs.
Due to similarities in symptoms, a physician will rule out other neurologic disorders before making the diagnosis (Shackel, 2004; NINDS, 2013).
A clinical examination and diagnostic tests are used to establish the diagnosis and include:
Electrodiagnostic tests such as electromyography (EMG), and nerve conduction velocity (NCV).
Blood and urine studies such as high-resolution serum protein electrophoresis, and thyroid and parathyroid hormone levels.
Spinal Tap and X-rays, including MRI and or myelogram of the cervical spine
Voxel Based Morphometry (Padovani, 2006)
Diffusion Tensor Imaging (Padovani, 2006)
Videofluroscopy and pulse oximetry during swallowing (Rocha JA 2005)
Muscle and nerve biopsies may be performed
Used to exclude possible diagnoses other than motor neuron disease (Krivickas, 2003)
Nerve biopsies can be performed to determine nerve disease or nerve regeneration. (NINDS, 2016)
MRI
CSF laboratory testing can rule out MS (Krivickas,2003)
DNA Testing can rule out Kennedy's Disease (Krivickas,2003)
++
Progressive Bulbar Palsy usually advances in the following manner
Atrophy and fasciculations of the bulbar muscles
The tongue appears wasted and has prominent fasciculations, which produce a writhing appearance.
Orbicularis oris affected at the same time as the tongue.
Orbicularis oculi and other facial muscles affected later.
The palate, extrinsic muscles of the pharynx and larynx atrophy.
Protrusion, both lateral and vertical, of the tongue is weakened and eventually lost (Lapiedra, 2002).
Early speech impairments are followed several months later by dysphagia including troubles swallowing, aspiration, and fatigue during eating (Rocha, 2005; Ball, 2001).
In ALS, initial impairments in verbal communication ability are correlated with a more poorly perceived quality of life (Felgoise, 2015).
PBP may become indistinguishable from ALS as it progresses (Hawkins, 1990).
Females are more likely to have isolated PBP before progressing to ALS (Karam, 2010; Kollewe, 2011)
The progression to ALS is similar for both PBP-N and PBP-A (Karam, 2010)
ALS patients with bulbar onset have a more rapid progression than those who have limb onset (Turner, 2010)
Death most often is caused by pneumonia and usually occurs 1-3 years after onset of PBP, (Rubin, 2008)
PBP usually progresses to aspiration pneumonia or respiratory arrest (Lapiedra, 2002; Kernich 2009).
33 months is average life expectancy for persons with bulbar symptoms (Turner, 2010)
Progressive bulbar symptoms result in decreased quality of life and life expectancy for individuals with ALS (Kühnlein, 2008)
Mental illnesses commonly develop in patients with ALS/ PBP including depression and anxiety disorders
80% of patients with PBP-N also developed depression (Karam, 2010)
The most common predictive factors of Hospital Anxiety and Depression scores identifying disorders were negative social support and high bulbar scores on the ALS Severity Scale (Goldstein, 2006)
Self esteem Scale scores were best predicted by pre-illness marital intimacy scores (Goldstein, 2006)
PBP commonly results in denutrition (Lapiedra, 2002).
+++
IV. Medical Management
++
Palliative care and management of symptoms are major concerns in the treatment of Bulbar Palsy (Kühnlein, 2008)
Medications to treat depression:
Antidepressants may be prescribed for patients who have impaired swallowing
Amitriptyline – used to relieve symptoms of depression, loss of interest in daily activities, appetite changes, fatigue, and thoughts of death or suicide.
Zoloft – used to treat depression and panic disorder.
Medications to aid sleep
Medications to treat excessive saliva (NINDS, 2013; Kernich, 2009)
Amitriptyline
Glycopyloate
Atropine (Kühnlein, 2008)
Botulinum toxin –jaw spasms, drooling (NINDS, 2013)
Transdermal patches that secrete scopolamine (hyoscine hydrobromide) over a 72 hour period (Kühnlein, 2008)
Steroid Therapy
Commonly used in children with BVVL or Fazio-Londe-Syndrome.
Steroid therapy can help improve activity, thus making normal feeding possible and increasing patient morale (Gallai, 1981; Hawkins, 1990).
Riboflavin Supplementation (Manole, 2014)
Oral riboflavin supplementation in children with BVVL showed promising results, with mean age at start of treatment = 6.9 years.
When high dose therapy begins early in the disease process, significant clinical and biochemical improvements have been found.
Clinical improvements include improved strength, motor function, respiration, hearing, and vision (Bosch, 2012)
Significant return of motor function and muscle strength, even to the point of removing respiratory support, has occurred in a few patients (Timmerman, 2014).
Long term effects are unknown and symptoms often returned when supplementation stopped.
Percutaneous Endoscopic Gastrostomy
PEG/ G-tube provides nutrition to patients who can no longer swallow. (Kernich, 2009)
This does not exclude the risk of aspiration, because saliva aspiration can still occur (Kent, 2013)
Management of bowel-bladder
Increasing fluid intake can help to improve bowel/bladder problems.
Physician may prescribe a stool softener, laxative, or bulk former.
Management of respiration/ventilation
Invasive ventilation or tracheostomy may be needed to assist with respiration, especially in the later stages of the disease
Assisted respiration at night using a respirator may help to prevent respiratory arrest during sleep (Gallai, 1981).
Management of Nutrition
Frequent assessments of nutrition should be conducted on patients with bulbar symptoms. Malnutrition can be prevented with a diet of adequate calories, dietary texture modification, and placement of a feeding tube (Raheja, 2016).
Management of Communication
A palatal lift may help to improve speech.
Computer programs with electronic devices are available to help people communicate and control their environment.
Picture boards or hand signals can be helpful communication tools (Kent, 2013)
Personal Care
+++
V. Therapeutic Management
++
Physical Therapist Preferred Practice Pattern
Role of Physical Therapy and Occupational Therapy
Help maintain functional activities and mobility, improve strength, and prevent or alleviate joint stiffness and pain that results from muscle wasting, cramps, or spasticity
Help the patient maintain function, mobility, and independence (ADLs & IADLs)
Evaluate and monitor the progression of the disease through clinical testing and outcome measures (Takeuchi, 2008; Bello-Haas, 1998)
Possible outcomes measures: ALSFRS-R, modified Norris scale, grip strength with hand dynamometer, ALSAQ-5 (Hashizume, 2012), ALSSQOL-R (Simmons, 2015)
Assistive Devices can help the patient to remain as independent as possible and may include:
Education
Educate caregivers on energy conservation methods for safe, efficient transfers (Medscape, 2013)
Instruct caretakers in passive range of motion exercises, and body positioning techniques to help with transfers.
Educate and perform airway clearance techniques and respiratory maintenance (Garcia-Pachon, 1994).
Perform home evaluation and advise on equipment use and home alterations.
Role of Speech Pathologist
Assess and advise patient on the use of swallowing techniques, and proper food selection and preparation.
Education
Provision of moist soft food and the avoidance of dry crumbly and mixed texture foods (Kent, 2013)
Maintaining an upright position and keeping chin-tucked to reduce the risk of aspiration (Kent, 2013)
As dysphagia progresses the SLP and dietician play a major role in education of the possible implementation of a enteral feeding system
Communication strategies can be taught and suggest appropriate augmentative equipment
Reducing background noise, facing the individual, not rushing the individual, and using close-ended questions (Kent, 2013)
Augmentative and alternative communication devices can be used to enhance a patients ability to communicate non-verbally
Low-tech devices such as alphabet boards or high-tech devices such as computer programs can be used depending on the progression of symptoms affecting speech (Barber, 2015)
Role of Respiratory Therapist
Instruct the patient in lung strengthening exercises to delay disease progression at the initial onset of respiratory difficulty
Instruct the patient regarding lung health and provide airway clearance when the muscles used for swallowing begin to weaken later in the disease process
Potentially provide artificial ventilation or suctioning of the lungs because of fluid build-up from pneumonia in late stages of the disease
Non-invasive positive pressure ventilation is the initial preferred treatment for patients with respiratory symptoms (Simmons, 2013).
Non-invasive ventilation systems can add up to 48 days of survival in patients with late stage ALS and good or moderate bulbar functioning
Social Worker/Psychiatrist
+++
VI. Consumer and Professional Resources
++
Support Groups:
ALS forums: http://www.alsforums.com/.
ALS Advocacy Support Community
MedHelp Forums – Neurology Community: progressive bulbar palsy sharing
Motor Neuron Disease Association
National Neuroscience Institute- MND/ALS Support Group c/o Department of Neurology
Patient Support (The Rilutek Care Connection)
Organizations:
+
Agrawal
P, Pandey
A, Sompura
S, Khandelwal
G, Yadav
S. A rare case report showing direct association between hepatitis B and bulbar palsy. J Assoc Physicians India. 2014;62(3):267–268.
+
Armstrong
RA. Visual signs and symptoms of progressive supranuclear palsy. Clin Exp Optom. 2011;94(2):150–160.
+
Ball
LJ, Willis
A, Beukelman
DR, Pattee
GL. A protocol for identification of early bulbar signs in amyotrophic lateral sclerosis. J Neurol Sci. 2001;191:43–53.
+
Barber
C. Management of bulbar symptoms in motor neurone disease: a community speech and language therapist perspective. J Neurosci Nurs. February 2015;11(1):41–46.
+
Bello-Haas,
V. Physical therapy for a patient through six stages of amyotrophic lateral sclerosis. Phys Ther. 1998; 78: 1312–1324.
+
Bosch
AM, Stroek
K, Abeling
NG, Waterham
HR, Ijlst
L, Wanders
RJ. The Brown-Vialetto-Van Laere and Fazio Londe syndrome revisited: natural history, genetics, treatment and future perspectives. Orphanet J Rare Dis. 2012;7:83. doi: 10.1186/1750-1172-7-83.
+
Britton
D, Benditt
JO, Merati
AL. Associations between laryngeal and cough dysfunction in motor neuron disease with bulbar involvement. Dysphagia. 2014;29(6):637–46.
+
De Carvalho
M, Turkman
A, Swash
M. Motor responses evoked by transcranial magnetic stimulation and peripheral nerve stimulation in the ulnar innervations in amyotrophic lateral sclerosis: the effect of upper and lower motor neuron lesion. J Neurol Sci. 2003;210:83–90.
+
Ertekin,
C. Mechanisms of dysphagia in suprabulbar palsy with lacunar infarct. Stroke. 2000;6:1370–6.
+
Evans,
J, Shaw
P.J. (2001). Motor Neuron Disease – Clinical Features and Pathogenesis. The Pharmaceutical Journal. 276: 681–684.
+
Felgoise
SH, Zaccheo
V, Duff
J, Simmons
Z. Verbal communication impacts quality of life in patients with amyotrophic lateral sclerosis. Amyotroph Lateral Scler Frontotemporal Degener. 2016;17(3-4):179–183.
+
Gallai,
V., Hockaday,
J.M., Hughes,
D.J.,
et al. Ponto-Bulbar Palsy with Deafness (Brown-Vialetto-Van Laere Syndrome. J Neurol Sci. 1981;50:259–275.
+
García-Pachón,
E. Clinical significance of upper airway dysfunction in motor neurone disease. Thorax. 1994; 49: 896–900.
+
Goldstein
LH, Atkins
L, Landau
S, Brown
RG, Leigh
PN. Longitudinal predictors of psychological distress and self-esteem in people with ALS. Neurology. 2006;67(9):1652–8.
+
Hawkins
SA, Nevin
NC, Harding
AE. Pontobulbar palsy and neurosensory deafness (Brown-Vialetto-Van Laere syndrome) with possible autosomal dominant inheritance. Journal of Medical Genetics. 1900;27(3):176–179.
+
Hashizume
A, Katsuno
M, Banno
H, Suzuki
K, Suga
N, Mano
T, Atsuta
N, Oe
H, Watanabe
H, Tanaka
F, Sobue
G. Longitudinal changes of outcome measures in spinal and bulbar muscular atrophy. Brain. 2012;135(Pt 9):2838–48.
+
Karam
C, Scelsa
S, MacGowan
D. The clinical course of progressive bulbar palsy. Amyotrophic Lateral Sclerosis. 2010;11:364–368.
+
Kent
A. Progressive supranuclear palsy. Nursing Standard. 2013;27(51):48–57.
+
Kernich
CA. Motor Neuron Disease. The Neurologist. 2009; 15(1):49–50.
+
Kollewe
K, Munte
TF, Samii
A, Dengler
R, Petri
S, Mohammadi
B. Patterns of cortical activity differs in ALS patients with limb and/or bulbar involvement depending on motor tasks. J Neurol. 2011;258:804–810.
+
Krivickas
LS. Amyotrophic lateral sclerosis and other motor neuron diseases. Phys Med Rehabil Clin N Am. 2003;14(2):327–45.
+
Lapiedra
RC, Lopez
LAM, Gomez
GCE. Progressive Bulbar Palsy: a case report diagnosed by lingual symptoms. J Oral Pathol Med. 2002;31: 277–279.
+
Leighton
S. Swallowing in motor neuron disease. J R Soc Med. 1994; 87: 801–5.
+
Manole
A, Fratta
P, Houlden
H. Recent advances in bulbar syndromes: genetic causes and disease mechanisms. Curr Opin Neurol. 2014;27(5):506–514.
+
Kühnlein
P, Gdynia
H-J, Sperfeld
A-D,
et al. Diagnosis and treatment of bulbar symptoms in amyotrophic lateral sclerosis. Nat Clin Pract Neurol. 2008;4(7):366–374.
+
Mégarbané
A, Desguerres
I, Rizkallah
E, Delague
V, Nabbout
R, Barois
A, Urtizberea
A. Brown-Vialetto-Van Laere Syndrome in a large inbred Lebanese family: Confirmation of autosomal recessive inheritance? Am J Med Genetics. 2000;92(2):117–121.
+
Mohammadi
B, Kollewe
K, samii
A, Krampfl
K, Dengler,
R, Munte
TF. Decreased brain activation of tongue movements in amyotrophic lateral sclerosis with bulbar involvement but not Kennedy syndrome. J Neurol. 2009;256:1263–1269.
+
Morimoto
N, Kurata
T, Sato
K, Ikeda
Y, Sato
S, Abe
K. Frontal dysfunctions of ALS-PBP patients in relation to their bulbar symptoms and rCBF decline. J Neurol Sci. 2012;319(1-2):96–101.
+
Noto
Y, Misawa
S, Kanai
K,
et al. Awaji ALS criteria increase the diagnostic sensitivity in patients with bulbar onset. Clin Neurophysiol. 2012;123(2):382–385.
+
Padovani,
A. Diffusion tensor imaging and voxel based morphometry study in early progressive supranuclear palsy. Journal of Neurology Neurosurgery Psychiatry. 2006; 77:457–63.
+
Radunovic
A, Annane
D, Rafiq
MK, Mustfa
N. Mechanical ventilation for amyotrophic lateral sclerosis/motor neuron disease. Cochrane Database Syst Rev. 2013;3:CD004427.
+
Raheja
D, Stephens
HE, Lehman
E, Walsh
S, Yang
C, Simmons
Z. Patient-reported problematic symptoms in an ALS treatment trial. Amyotroph Lateral Scler Frontotemporal Degener. 2016;17(3-4):198–205.
+
Rocha
JA, Reis
C, Simões
F, Fonseca
J, Mendes
Ribeiro J. Diagnostic investigation and multidisciplinary management in motor neuron disease. J Neurol. 2005;252(12):1435–1447.
+
Sathasivam
S. Brown-Vialetto-Van Laere syndrome. Journal of Rare Diseases. 2008; 3:9.
+
Simmons
Z. Patient-Perceived Outcomes and Quality of Life in ALS. Neurotherapeutics. 2015;12(2):394–402.
+
Simmons
Z. Rehabilitation of motor neuron disease. Handb Clin Neurol. 2013;110:483–498.
+
Takeuchi,
Y. Walking capacity evaluated by the 6-minute walk test in spinal and bulbar muscular atrophy. Muscle and Nerve. 2008; 38: 964–971.
+
Timmerman
V, Jonghe
PD. Promising riboflavin treatment for motor neuron disorder. Brain. 2014;137(1):2–3.
+
Traynor
BJ, Codd
MB, Corr
B, Forde
C, Frost
E, Hardiman
OM. Clinical features of Amyotrophic lateral sclerosis according to the El Escorial and Airlie House Diagnostic Criteria.
Arch Neurol.[Archives of Neurology Full Text] 2000;57:1171–1176.
+
Turner
MR, Scaber
J, Goodfellow
JA, Lord
ME, Marsden
R, Talbot
K. The diagnostic pathway and prognosis in bulbar-onset amyotrophic lateral sclerosis. Journal of the Neurological Sciences. 2010;294(1–2):81–85.
+
Urban
PP, Bogt
T, Hopf
HC. Corticobulbar tract involvement in amyotrophic lateral sclerosis: a transcranial magnetic stimulation study. Brain. 1998;121:1099–1108.
+
Urban
PP, Wicht
S, Hopf
HC. Sensitivity of transcranial magnetic stimulation of cortico-bulbar vs. cortico-spinal tract involvement in Amyotrophic Lateral Sclerosis (ALS). J Neurol. 2001;248:850–855.
+
Zampieri
C, Fabio
RPD. Progressive Supranuclear Palsy: Disease Profile and Rehabilitation Strategies. Phys Ther. 2006;86(6):870–880.
+
Zoccolella
S, Beghi
E, Palagano
G, Fraddosio
S, Guerra
V, Samarelli
V, Lepore
V, Simone
I L, Lamberti
P, Serenga
L, Logroscino
G. Analysis of survival and prognostic factors in amyotrophic lateral sclerosis: a population based study. J Neurol Neurosurg Psychiatr. 2008;79(1):33–7.
++
Rett syndrome (RTT) is a neurodevelopmental disorder effecting girls (primarily) that manifests in a degeneration of developmental skills in infants who appeared to be developing typically; characterized by loss of hand function and unusual stereotypical hand motions, loss of previously acquired language and motor skills, respiratory symptoms (apnea or hyperventilation), seizures, slowed head growth and autistic-like behaviors.
Incidence is estimated at one in every 10,000-22,000 females in the United States (IRSF)
Pathogenesis
Genetic mutation causes failure of neuronal maturation in diverse areas of nervous system (Hunter, 2007)
Typical or classic form of RTT, in 95-97% of cases, is caused by mutation in the Methyl-CpG-binding protein 2, or MECP2 gene on the X chromosome that controls the expression of many other genes and is important in brain development.
Although it is a genetic disorder, it is typically a spontaneous mutation and not inherited.
Mutation in MECP2 gene is also found in individuals who do not have RTT clinical manifestations, including asymptomatic female carriers.
Since girls have two X chromosomes, the time of onset and severity of RTT symptoms are determined, in part, on which of the X chromosomes are active in any given cell.
Boys with the aberrant MECP2 gene on their one X chromosome have no compensatory genetic mechanism and are typically severely involved and die soon after birth.
Atypical forms of RTT can vary in genetic composition and clinical symptoms
++
Signs and Symptoms –
Classic form of the disorder has been described in four phases (US Department of Health and Human Services, 2015):
Early onset phase: developmental delays become apparent during first year; hypotonia and stereotyped hand movements (in 42%); often not noticed immediately but video demonstrates that all infants showed problems with early development from birth to age 6 months (Einspieler, 2005).
Rapid Destructive Phase: rapid regression of previously acquired skills with purposeful hand movements and speech some of the earliest skills lost; stereoptypic hand movements and breathing problems emerge.
Plateau Phase: regression slows and other problems may seem to lessen or even improve; seizures and movement problems common; some may spend most of lives at this stage.
Late Motor Deterioration Phase. Individuals in this stage may become stiff, lose functional abilities, even becoming. Musculoskeletal problems like scoliosis may continue to worsen and interfere with mobility, positioning, and comfort; cardiopulmonary problems may increase because of general lack of physical activity and restricted mobility; stereotypic hand movements and breathing problems seem to become less common as the individual ages.
Other common symptoms in complex disorder
Gross and fine motor regression (early) and delays
Gait abnormalities – lack of accurate feedforward control and diminished impulse to move forward (Isaias, 2014); shaky, unsteady, stiff, wide-based gait and/or toe walking (Nuel, 2010; Hunter, 2007)
Inability to grasp, reach or intentionally touch things
Hypotonia in early stage (common) and hypertonia in later stages (variable)
Gastrointestinal dysmotility, including gastroesophageal reflux and reflux disease, constipation (Baikie, 2014)
Abdominal bloating in 42.4%, more frequent in individuals older than 20 years (Mackay, 2017); aerophagia (air swallowing); abdominal discomfort; gallbladder problems such as gallstones (Hunter, 2007)
Poor growth and weight gain (Leonard, 2013)
Cardiopulmonary problems – cardiac arrhythmias and more generalized problems secondary to lack of physical activity, severe scoliosis constricting lung volume, etc.
Autonomic breathing disturbances - from a large international database study (413 families): breath-holding for 68.8% and hyperventilation for 46.4%; hyperventilation more prevalent and frequent in those younger than 7 years of age; onset of breathing irregularities usually during early childhood; caregivers perceived that daily life was considerably impacted in just over one third of those with breath-holding (35.8%) hyperventilation (35.1%). Only 31 individuals had received medical treatment (Mackay, or 2017)
Behavioral and emotional aspects, including mood fluctuations; signs of fear/anxiety, inconsolable crying and screaming at night; repetitive mouth and tongue movements; grimacing; social withdrawal (Mount, 2002; Lee, 2013; Hunter, 2007)
Autism-like behaviors or autism as a co-occurring condition
Severe cognitive impairment and learning disabilities (Berger-Sweeney, 2011)
Seizure disorder:
In study of 165 patients with RTT: epilepsy in 79%; drug-resistant in 30%; differs among the various phenotypes and genotypes with respect to age at onset, drug responsiveness, and seizure semiology (Pintaudi, 2010);
In study investigating database of 1248 girls/women with RTT: epilepsy in 68.1%; uncontrolled in 32.6%; mean age of onset 4.68 years; younger age of onset correlated to severity (Nissenkorn, 2015)
Excessive saliva and drooling; difficulty swallowing and chewing food which may cause choking and aspiration; bruxism (grinding teeth) not usually at night (Hunter, 2007)
Slowing head growth beginning at approximately 5-6 months; microcephaly
Poor circulation with possible blue discoloration arms and legs
Vomiting and strabismus noted as part of regression in early stage (Lee, 2013)
Intense eye gaze (Hunter, 2007)
Disrupted sleep patterns (Hunter, 2007)
Diagnostic Testing
Genetic testing of a blood sample can identify if a child has one of the known mutations of the MECP2 gene but other disorders have a mutated MECP2 gene so confirmation by clinical presentation is also necessary.
Diagnostic Statistical Manual (DSM)-IV was published in 1994; the genetic cause of RTT was identified in 1999. Without knowledge of the genetic cause, RTT was listed in the DSM-IV as a subcategory of autistic spectrum disorder. DSM-V, published in 2013 (American Psychiatric Association), removed RTT from under the ASD umbrella but specific diagnostic criteria for RTT are not included.
RettSearch members, representing the majority of the international clinical RTT specialists, reached consensus on revised and simplified diagnostic criteria (Neul, 2010). Various levels and options are involved for both typical and atypical forms of RTT, but the main criteria for the typical type are:
A pattern of development, regression, then recovery or stabilization
Partial or complete loss of purposeful hand skills such as grasping with fingers, reaching for things, or touching things on purpose
Partial or complete loss of spoken language
Repetitive hand movements, such as wringing the hands, washing, squeezing, clapping, or rubbing
Gait abnormalities, including walking on toes or with an unsteady, wide-based stiff-legged gait.
+++
III Prognosis and Sequelae
++
Trends of Progression
Phases of RTT summarized above
Since RTT is relatively rare and diagnostic criteria are relatively recent, long term outcomes and life expectancy are not well understood, but despite the severity of the disease there are women with RTT living into their 40's and 50's (Hunter, 2007)
In a Danish study of 27 women with RTT over the age of 30 years with confirmed MECP2 mutation, 63% unable to walk outside their homes; 11% unable to walk at all; 67% could not transfer from sitting to standing without support; profound difficulties communicating; 85.1% could consistently point with their hand or eyes to things of interest (Schönewolf-Greulich, 2016)
Expected Sequelae – possible worsening of scoliosis, musculoskeletal contractures, decreased functional mobility, cardiovascular problems
+++
IV Medical/Surgical Management
++
+++
V Key Aspects of Therapeutic Management
++
Multidisciplinary team approach essential because of broad range and severity of symptoms
Involvement of caregivers/family as part of team; caregiver education (Hunter, 2007)
RTT Caregiver Inventory Assessment – valid and reliable inventory of physical, emotional and social burden for caregivers (Lane, 2017)
Rett Syndrome Behavior Questionnaire (RTTBQ) can help identify emotional and behavioral aspects (Mount, 2002)
Physical therapy to address deterrents to functional mobility and help prevent secondary problems:
Transfers, gait (including anticipatory strategies), balance, coordination, positioning, strength, endurance, joint mobility
Assistive technology including appropriate seating systems, assistive gait devices, supported standing
Hippotherapy; aquatic therapy (Hunter, 2007)
Direct, indirect and consultative roles, as appropriate within educational environment (e.g., school bus, adapted physical education, energy conservation, playground)
Speech and language pathology to promote expressive and receptive language or evaluate for augmentative communication strategies/devices
Occupational therapy for support with activities of daily living (ADL) and fine motor function (e.g., dressing, bathing, writing, feeding/eating)
Special education for support in educational environment
Psychologist or psychiatrist to help manage emotional/behavioral aspects
+
American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. 5th edition. Washington, DC: American Psychiatric Association; 2013.
+
Baikie
G, Ravikumara
M, Downs
J,
et al. Gastrointestinal dysmotility in Rett syndrome. J Pediatr Gastroenterol Nutr. 2014;58(2):237–44.
+
Berger-Sweeney
J. Cognitive deficits in Rett syndrome: what we know and what we need to know to treat them. J Neurobiol Learn Mem. 2011;96(4):637–46.
+
Downs
J, Bergman
A, Carter
P,
et al. Guidelines for management of scoliosis in Rett syndrome based on expert consensus and clinical evidence. Spine. 2009;34(17):E607–617.
+
Einspieler
C, Kerr
AM, Prechtl
HFR. Is the early development of girls with Rett disorder really normal? Pediatr Res. 2005;57:696–700.
+
Isaias
IU, Dipaola
M, Michi
M,
et al. Gait initiation in children with Rett syndrome. PLoS One. 2014;9(4):e92736. doi: 10.1371/journal.pone.0092736. eCollection 2014.
+
Jefferson
A, Leonard
H, Siafarikas
A,
et al. Clinical guidelines for management of bone health in Rett syndrome based on expert consensus and available evidence. PLoS One. 2016;11(2):e0146824.
+
Kaufmann
WE, Stallworth
JL, Everman
DB, Skinner
SA. Neurobiologically-based treatments in Rett syndrome: opportunities and challenges. Expert Opin Orphan Drugs. 2016;4(10):1043–1055.
+
Lane
JB, Salter
AR, Jones
NE,
et al. Assessment of caregiver inventory for Rett syndrome. J Autism Dev Disord. 2017;47(4):1102–1112.
+
Lee
JYL, Leonard
H, Piek
JP, Downs
J. Early development and regression in Rett syndrome. Clin Genetics. 2013;84(6):572–576.
+
Leonard
H, Ravikumara
M, Baikie
G,
et al. Assessment and management of nutrition and growth in Rett syndrome. J Pediatr Gastroenterol Nutr. 2013;57(4):451–460.
+
Mackay
J, Downs
J, Wong
K, Heyworth
J, Epstein
A, Leonard
H. Autonomic breathing abnormalities in Rett syndrome: caregiver perspectives in an international database study. J Neurodev Disord. 2017;28(9):15.
+
Mount
RH, Charman
T, Hastings
RP, Reilly
S, Cass
H. The Rett Syndrome Behavior Questionnaire (RTTBQ): refining the behavioral phenotype of Rett syndrome. J Child Psychol Psychiatry. 2002;43(8):1099–1110.
+
Neul
JL, Kaufmann
WE, Glaze
DG, Christodoulou
J,
et al. Rett syndrome: revised diagnostic criteria and nomenclature. Ann Neurol. 2010;68(6):944–950.
+
Nissenkorn
A, Levy-Drummer
RTT, Bondi
O,
et al. Epilepsy in Rett syndrome: lessons from the Rett networked database. Epilepsia. 2015;56(4):569–576.
+
Pintaudi
M, Caleva
MG, Bignoli
A,
et al. Epilepsy in Rett syndrome: clinical and genetic features. Epilepsy Behav. 2010;19:296–300.
+
Schonewolf-Greulich
B, Stahlhut
M, Larsen
JL, Syhler
B, Bisgaard
AM. Functional abilities in aging women with Rett syndrome: the Danish cohort. Disabil Rehabil. 2017;39(9):911–918.
+
Weaving
LS, Ellaway
CJ, Geez
J, Christodoulou
J. Rett syndrome: clinical review and genetic update. J Med Genet. 2005;42:1–7.
++
First described by R. D. Reye in 1963. He linked the presence of a viral illness, an early symptom, with the use of aspirin and other salicylates (Langford, 2002).
Common viral infections that are associated with Reye's: Influenza A, Influenza B, varicella, gastroenteritis, herpes simplex, poliomyelitis, Epstein-Barr, measles, and rubella (Cooper, 2003; Starko, 1980).
While the etiology, epidemiology and natural history of Reye's syndrome remain unknown, the failure of mitochondria function is thought to contribute to this disorder (da Costa, 2011).
Symptoms show striking similarities to salicylate toxicity, and epidemiologic studies have noted high rates of salicylate consumption by patients with Reye's syndrome (Starko, 1980).
Conflicting evidence about the link between Reye's and salicylates; no specific dose of salicylate has been linked with causality (Glasgow, 2006).
Clinical differentiation difficult; various inborn metabolic disorders that require extensive laboratory testing for diagnosis, can present with manifestations that mimic those of Reye's syndrome (Belay, 1999).
Cause of disease remains unclear; even after years of research and discussion, it still cannot be proved irrefutably that the administration of acetylsalicylic acid to children with viral infections is a causative (Tanret, 2011).
Four concepts of pathogenesis (Crocker, 1981):
Intrinsic toxin – affects mitochondrial metabolism and other cellular metabolic functions
Extrinsic toxin – can alter response during recovery of a virus
According to the Mayo clinic "Exposure to certain toxins — such as insecticides, herbicides and paint thinner — also may contribute to Reye's syndrome."
Genetic susceptibility to Reye's syndrome
Primary defect in lipid ammonia metabolism in which cerebral edema and coma occur following viral illness
Decreased incidence: discovery of inborn errors in metabolism and identification of toxins that have manifestations similar to those of Reye syndrome, dramatic decrease in the use of aspirin among children (Saradhai, 2010)
Study results indicate a defective isoprenoid pathway as a possible cause of altered mitochondrial function (Kurup, 2003).
Classified as an acute encephalopathy in children <18 years of age (Glasgow, 2006).
Biphasic disorder without a known etiology; not contagious (Wei, 2005).
Found to show a monophasic early clinical course (Maegaki, 2006).
Considered a medical emergency because of the rapid deterioration following onset.
Viral-like illness, followed abruptly by vomiting and encephalopathy (Starko, 1980).
Child shows signs of recovery from the initial viral illness when acute symptoms of Reye's begin without warning. In most cases aspirin was reportedly used during the illness (Rice, 2003)
Syndrome affects many organs, but the brain and liver are primary targets.
Unspecific pathological changes occurring during Reye's syndrome: cerebral edema without inflammation, which can increase inter-cranial pressure (ICP); fatty deposits in the liver; edema and deformation of mitochondria in hepatocytes and cerebral neurons; elevations in amino acids, free fatty acids, and short-chain fatty acids (Trauner, 1980; D'Antiga, 2006; Starko, 1980).
Fatty infiltration of liver with concurrent encephalopathy, hyperammonemia and increased ICP; selective hepatic dysfunction; fatty infiltration of the kidneys (renal tubular cells), brain and myocardium (Maheady, 1989; Glasgow, 2006).
Additionally, elevated levels of ammonia (NH3) cause the blood to become acidic and blood sugar levels can be lowered to hypoglycemic levels.
This condition was very common in the 1970s and reached its highest level of 555 cases in the US in 1980. Since then, incidence rates have continuously dropped to less than 2 cases per year since 1994 (Cooper, 2003).
Improved diagnostic techniques and criteria have contributed to decrease in cases; many known to be metabolic disorders (Orlowski, 2002)
Between 1981 and 1997 there were 1207 reported cases of Reye's syndrome; 93 percent Caucasian and 52% female (Belay, 1999); no apparent gender bias in the development of the syndrome.
++
Reye's syndrome has various symptoms that do not appear in any set sequences, nor do they all have to be present in every patient
Abrupt deterioration after viral illness occurs with profuse vomiting and altered consciousness (Glasgow, 2006).
Signs/Symptoms:
5 stages of progression through Reye's syndrome (if untreated) (Cooper, 2003).
Stage I: vomiting, sleepiness, lethargy
Stage II: restlessness, irritability, combativeness, delirium, confusion, increased respiratory and heart rates, dilated pupils with slow response
Stage III: obtundation and comatose with decorticate posturing
Stage IV: deep coma, decerebrate posturing, fixed pupils (nonresponsive to light), abnormal reflexes
Stage V: seizures, flaccid paralysis, unresponsive pupils, no normal reflexes, cannot breathe independently
Other symptoms that may be seen with Reye's syndrome include diplopia, hearing loss, muscle function loss or paralysis of the arms and legs, and/or speech difficulties.
Related to increased intracranial pressure: vomiting with no nausea, changes in mental status, lethargy, confusion, and combative (Rice, 2003).
Upper respiratory tract prodrome characterized by headache (Starko, 1980).
Hyperreflexia, seizures, and coma may also occur in the later stages of Reye's syndrome (Cooper, 2003; Rice, 2003).
Infants may present with diarrhea instead of the trademark vomiting. Irregular breathing is also common
Important differential diagnosis in adults: drug overdose, diabetes, head trauma, poisoning, encephalitis, meningitis, kidney and liver failure, or psychiatric illness; in infants: SIDS; and in children under age 6: certain metabolic disorders (Rice, 2003).
Hypoglycemia, hyperthermia, and acidosis may occur from other metabolic changes (Cooper, 2003).
Hepatomegaly (Samaha,1974).
Elevation of liver enzymes and abnormal coagulation tests (Maegaki, 2006).
++
According to the National Reye's Syndrome Foundation's emergency room information a differential diagnosis must be done to rule out the following: meningitis, encephalitis, diabetes, drug overdose, poisoning, head trauma, renal or hepatic failure, metabolic disorder
Diagnostic tests, labs and other diagnostic measures: complete blood count, serum ammonia, liver functional studies, liver biopsy, lumbar puncture and examination of cerebrospinal fluid (CSF) (Lindh, 2009):
Diagnostic Criteria established by CDC
Apparent liver dysfunction (liver biopsy with fatty changes and increased ammonia levels), <9 white blood cells per mm CSF, brain biopsy showing non-inflammatory edema (Cooper, 2003; Costa, 2011)
Must be associated with either fatty changes in the liver or a 3x or greater increase of transaminases or ammonia in serum and no other reasonable explanation for cerebral or hepatic abnormalities (D'Antiga, 2006).
SGOT (SAT) and SGPT (ACT) are two commonly used liver function tests that are commonly used in the diagnosis of Reye's syndrome. Their results can be obtained quicker than many other tests and are preferred in this emergency situation (National, No Date).
Low serum glucose levels
Increased serum fatty acid and lactate levels
Elevated SGOT and SGPT, in the absence of jaundice, in excess of 200 units combined with elevated blood ammonia confirm a diagnosis of Reye's syndrome. (Samaha, 1974) (National Reye's Syndrome Foundation)
Absence of jaundice, despite increased liver aminotransferase levels, rules out acute hepatic failure and hepatic encephalopathy (Maheady, 1989).
Prolonged thrombin time and relatively normal cerebrospinal fluid are found in almost every case (Samaha, 1974).).
Individual Reye Score of 17.8 or more out of 25 is associated with aspirin induced Reye's Syndrome while 12 or less is not considered a form of Reye's Syndrome (Hardie, 1996).
Reye's patients frequently show CT neuroimaging abnormalities within 2 days of onset (Maegaki, 2006).
Diffusion-weighted images (DWI) are the most sensitive technique for identification of cytotoxic edema in the acute stages of encephalitis, encephalopathy, and status epilepticus (Maegaki, 2006).
The DWI shows selective early susceptibility of the thalamus and midbrain to the "Reye's endotoxin" but does not show the nature of the disease (Johnsen, 2006).
Should rule out exposure to hepatotoxic substance as it shows a similar clinical picture to Reye's syndrome (Samaha, 1974).
+++
Abnormal Test Results with Reye's Syndrome (Dezateux, 1986):
++
Liver Function Studies: Asparte aminotransferase and alanine aminotransferase elevated to twice normal levels; bilirubin levels are usually normal
Liver Biopsy: Fatty droplets uniformly distributed throughout liver cells
Cerebrospinal Fluid (CSF) Analysis: White blood cell count lower than normal; if patient is in coma, there will be increased CSF pressure
Coagulation Studies: Prolonged prothrombin time and partial prothrombin time
Blood Values: Elevated serum ammonia level; normal or low (15% of cases) serum glucose level; increased serum fatty acid and lactate levels (Dezateux, 1986).
++
Prognosis for patients diagnosed with Reye's syndrome varies, based on the severity of CNS depression. ICP monitoring and early treatment of increased ICP has assisted in decreasing mortality rates from 90-20%. If death occurs, it is usually a result of cerebral edema or respiratory arrest.
++
++
++
The CDC considers patients who cannot be classified, because they have been treated with curare or other medications that alter level of consciousness, as stage VI (Sanadhai, 2010)
++
Early diagnosis and aggressive treatment leads to a better chance of full recovery. This is because Reye's syndrome is a progressive disease during the acute stage, especially if treatment is not started. Children who have made a recovery can still have significant neurological deficits.
Transfer to or consultation with a children's or teaching hospital is recommended (National, No Date).
Stages III-V are considered the late stages with poor chance of recovery without serious neurological damage.
Rapid progression through coma stages and high peak ammonia concentrations worsen prognosis (Dezateux, 1986).
Prognoses are often very poor, commonly consisting of neurological or liver damage or early death depending on the degree of central nervous system depression. (Maegaki, 2006).
Time is of the utmost importance.
Early diagnosis and initiation of treatment greatly improves prognosis (Glasglow, 2001).
Prognosis worsens the higher the stage and the younger the child, with death occurring in 85 percent of patients who have progressed to stage V (Sanadhai, 2010)
Dr. Crocker states, "Both death and neurologic sequelae are attributed to the insult to the nervous system" (Crocker, 1981)
Serum ammonia levels are also a predictor of neurologic damage, levels above 45mg/dl greatly increase the chance of neurologic sequelae (Belay, 1999)
Mortality rates for Reye's syndrome have only decreased from 50% to 20%.
10-20 percent have persistent neurologic damage ranging from slight to profound (Sanadhai, 2010)
+++
IV. Medical and Surgical Management
++
There is no cure. Treatment is supportive care (D'Antiga, 2006).
Treatment developed for each of the 5 stages of Reye's syndrome:
When used before evidence of irreversible brain stem damage, peritoneal dialysis may offer hope in reversing high mortality by correcting metabolic acidosis, providing glucose, and removing dialyzable poisons (Samaha, 1974).
Other treatments commonly used include cooling blankets for hyperthermia, fresh frozen plasma for increased prothrombin time, IV mannitol to help decrease intracranial pressure, barbiturate induced coma (most commonly phenobarbital), and behavior management (Trauner, 1980) (Jacobs, 1986).
Patients should be kept as calm and quiet as possible; unnecessary movement or stimulation could raise ICP (Sanadhai, 2010).
Nursing procedures should occur with patient in head up, midline position under full sedation and paralysis (Dezateux, 1986).
+++
V. Therapeutic Management
++
Neuromuscular Patterns: (APTA, 2017)
Pattern 5A: Primary Prevention/Risk Reduction for Loss of Balance and Falling
Pattern 5C: Impaired Motor Function and Sensory Integrity Associated With Nonprogressive Disorders of the Central Nervous System - Congenital Origin or Acquired in Infancy or Childhood
Pattern 5D: Impaired Motor Function and Sensory Integrity Associated With Nonprogressive Disorders of the Central Nervous System - Acquired in Adolescence or Adulthood
Pattern 5E initially, may progress Pattern 5I Impaired Arousal, Range of Motion, and Motor Control Associated with Coma, Near Coma, or Vegetative State
Musculoskeletal Patterns: (APTA, 2017)
Cardiopulmonary Pattern: (APTA, 2017)
The emergent nature of Reye's syndrome may not allow for physical therapy treatment if the patient has not been diagnosed and stabilized. However, it may be useful to treat extensor spasms and rigidity that occur to make the patient more comfortable.
Therapists should be aware of what stage the patient is in and take appropriate precautions (Williams, 2010):
Chest physical therapy may be required due to decreased clearance and ventilation (Trauner, 1980; Crocker, 1981).
Following Reye's syndrome a physical therapist may continue treatment for rigidity, as well as improving motor skills, regression in developmental status, motor tics, evaluation for learning disabilities, assess fall risk, and education for family members on support options
Contraindications: changing head and neck positions rapidly and in extreme ranges of motion, painful stimuli, coughing/struggling against intubation (patient needs to be sedated), use of any anesthetics that increase blood volume (may increase intracranial pressure), loud noise levels (Hubbert, 1979).
Precautions for patients who have previously suffered from Reye's Syndrome and may suffer from the following residual effects (National, 2008):
Prevention:
Education plays an enormous role in preventing Reye's syndrome among children and adolescents; directed towards educating adolescents and parents of older children about Reye's syndrome and cautioning against the use of aspirin to treat influenza, upper respiratory infections or varicella (Maheady, 1989)
+++
VI. Consumer and Professional Resources:
++
The National Reye's Syndrome Foundation:
Services provided by the National Reye's Syndrome Foundation:
National Reye's Syndrome Foundation
426 N. Lewis Street
PO Box 829
Bryan, OH 43506-0829
Phone: 1-800-233-7393 or 1-419-924-9000 Fax: 1-419-924-9999
Email: [email protected] Website: www.reyessyndrome.org
MDJunction – Reye's Syndrome Support Group
A community of patients, family members and friends dedicated to dealing with Reye Syndrome, together.
Website: http://www.mdjunction.com/reye-syndrome
Peace Health – Reye's Syndrome Helpline
Provides guidance to families affected by Reye's syndrome. Helps increase public awareness. Fund-raising.
Phone: 1-800-233-7393
Email: [email protected]
Website: http://www.reyessyndrome.org
National Institute of Neurological Disorders and Stroke
Provides information and access to current clinical trial information regarding Reye's Syndrome.
NIH Neurological Institute
P.O. Box 5801
Bethesda, MD 20824
Phone: (800) 352-9424 or (301) 496-5751
Website: http://www.ninds.nih.gov/disorders/reyes_syndrome/reyes_syndrome.htm
+
Belay
ED, Bresee
JS, Holman
RC,
et al. Reye's syndrome in the United States from1981 through1997. New Eng J Med. 1999;340(18):1377–1382.
+
Crocker
J, Bagnell
P. Reye's syndrome: a clinical review. Canadian Med Assoc J [serial online]. 1981;124(4):375.
+
Cooper
C. Would you recognize this syndrome? RN Mag. 2003;66:49–52.
+
Costa
PS, Ribeiro
GM, Vale
TC, Casali
TG, Leite
FJ. Adult Reye-like syndrome associated with serologic evidence of acute parvovirus B19 infection. Braz J Infect Dis. 2011;15(5):482–483.
+
D'Antiga
L, Zancan
L, Vecchi
M,
et al. Reye's syndrome: a disregarded lesson from the past. Ital J Pediatr. 2006;32:180–183.
+
Dezateux
CA, Dinwiddie
R, Helms
P,
et al. Recognition and early management of Reye's syndrome. Arch Dis Child. 1986;61:647651.
+
Glasglow
J, Middleton
B. Reye syndrome—insights on causation and prognosis. Arch Dis Child. 2001;85(5):351–353.
+
Glasgow
J. Reye's syndrome: the case for a causal link with aspirin. Drug Safety. 2006;29(12):1111–1121.
+
Hardie
R, Newton
L, Bruce
J. The changing clinical pattern of Reye's syndrome. Arch Dis Child. 1996;74:400–405.
+
Hubbert
CH. Critical care and anesthetic management of Reye's syndrome. South Med J. 1979;72:684–686.
+
Jacobs
H, Lynch
M, Cornick
J, Slifer
K. Behavior management of aggressive sequela after Reye's syndrome. Arch Phys Med Rehabil. 1986;67(8):558–563.
+
Jenkins
J, Glasgow
J, Crean
P,
et al. Reye's syndrome: assessment of intracranial monitoring. BMJ (Clinical Research Edition). 1987;294(6568):337–338.
+
Johnsen
SD, Bird
CR. The thalamus and midbrain in Reye syndrome. Pediatr Neurol. 2006;34:405–407.
+
Kurup
RK, Kurup
PA. The isoprenoid pathway and the pathogenesis of Reye's syndrome. Pediatr Pathol Mol Med. 2003;22:423–434.
+
Langford
NJ. Aspirin and Reye's syndrome: is the response appropriate? J Clin Pharm Ther. 2002;27:157–160.
+
Lindh
W, Pooler
M, Tamparo
C, Dahl
B. Delmar's Clinical Medical Assisting. Boston: Cengage Learning; 2009.
+
Lovejoy
FH, Smith
AL, Bresnan
MJ, Wood
JN, Victor
DI, Adams
PC. Clinical staging in Reye syndrome. Am J Dis Child. 1974;128(1):36–41.
+
Maegaki
Y, Kondo
A, Okamoto
R,
et al. Clinical characteristics of acute encephalopathy of obscure origin: a biphasic clinical course is a common feature. Neuropediatr. 2006;37:269–277.
+
Maheady
DC. Reye's syndrome: Review and update. J Pediatr Health Care. 1989;3(5):246–250.
+
Orlowski
J, Hanhan
U, Fiallos
M. Is aspirin a cause of Reye's syndrome? A case against. Drug Saf. 2002;25(4):225–231.
+
Rice
S. Reye's syndrome isn't just child's play. Nursing Mag. 2003;33:1–2.
+
Saradhai
P, Babu
V, Nivethithai
P, Nikhat
S. Pediatric Reye's syndrome: a review. Drug Invention Today [serial online]. 2010;2(2):163–166.
+
Samaha
FJ, Blau
E, Berardinelli
JL. Reye's syndrome: clinical diagnosis and treatment with peritoneal dialysis. Pediatrics. 1974;53:336–340.
+
Starko
KM, Ray
GC, Dominguez
LB, Stromberg
WL, Woodall
DF. Reye's syndrome and salicylate use. Pediatrics. 1980;66:859–864.
+
Tanret
I, Duh
D. The Reye syndrome. J Pharm Belg. 2011;(1):13–15.
+
Trauner
DA. Treatment of Reye syndrome. Ann Neurol. 1980;7:2–4.
+
Wei
C, Chen
H, Lee
P, Chen
C, Ma
C, Hwu
W. Reye's syndrome developing in an infant on treatment of Kawasaki syndrome. J Paediatr Child Health. 2005;41:303–304.
+
Williams
L, Kluwer
W. Professional Guide to Pathophysiology. 3rd ed. Philadelphia, PA: Lippincott Williams Wilkins; 2010.
+++
Seizure Disorder (Epilepsy)
++
A seizure is a transient disruption of brain function caused by excessive electrical discharge in the brain (Cross, 2004).
Epilepsy is defined as a neurologic condition in which an abnormality in the brain causes recurrent seizure activity (Gambrell, 2006).
Epilepsy affects approximately 65 million people worldwide, including over 2 million people in the United States (Epilepsy Foundation).
There are 150,000 new cases of epilepsy in the United States every year (Epilepsy Foundation).
Epilepsy affects 0.5% to 1% of children up to age 16. In children with developmental disabilities, the incidence of epilepsy increases by 30% to 50% (Wolf, 2006).
Epilepsy is the 4th most common neurologic disease in the elderly, after migraine, stroke, and Alzheimer's disease (Epilepsy Foundation).
++
++
Central nervous system (CNS) and systemic infections including meningitis and encephalitis, neurological/developmental, metabolic disorders, trauma, toxicological, neoplasms, congenital brain defects, and idiopathic (Friedman, 2006).
Non-epileptic seizures can result from alcohol withdrawal, fever, hypoxia, drug overdose, or poisoning (Gambrell, 2006). Other conditions which can precipitate seizures include hypoglycemia associated with insulin use; hyperglycemia; hyponatremia; hypocalcemia; medications, including all barbiturates and benzodiazepines, and phenothiazines, antidepressants, theophylline, and antibiotics (French, 2008).
Seizures that occur in the elderly are often secondary to other conditions such as stroke, infection, degenerative disease, dementia (Sirven, 2005).
Causes of neonatal seizures include hypoxic-ischemic encephalopathy, low gestational age, jaundice, intrapartum fever, the need for cardiopulmonary resuscitation, intracranial hemorrhage, developmental defects, and drug withdrawal. Brain alkalosis following birth asphyxia can play a key role in the triggering of seizures (Helmy, 2011).
Events that may trigger epileptic seizures: stress, poor nutrition, missed medication, skipping meals, flickering lights, illness, fever, allergies, lack of sleep, heat and humidity.
++
++
++
++
Patients may report aura that occurs just before a seizure, which may include varying sensations such as tingling, visual disturbances, or smells (Unnwongse, 2010).
++
++
Vary by type of seizure; generally, involves repetitive, transient episodes of altered consciousness, memory lapses, confusion, changes in speech and sensory or motor symptoms (Sirven, 2005).
Three main categories of seizures:
Partial - Limited to one cerebral hemisphere. Found in 60% of those with epilepsy. These include:
Simple partial - consciousness maintained; wide range of manifestations depending on the area of the brain affected; abnormal muscle contraction, staring episodes, visual disturbances, numbness, tingling, or hallucinations (PubMed Health, 2012).
Complex partial - typically start with some degree of impaired consciousness, followed by automatic uncontrolled movements
Secondarily generalized - starts as a partial seizure then spreads throughout the brain, becoming generalized
Generalized - Occurs in both cerebral hemispheres simultaneously. These include:
Absence - characterized by brief episodes of staring and impaired consciousness that may occur many times per day; also known as petit mal seizures. In nearly 70% of cases, these seizures stop by the age of 18.
Myoclonic - rapid jerking of body or limbs; very brief
Clonic - consist of involuntary rhythmic jerking movements of arms and legs, sometimes on both sides of the body. The movements cannot be controlled by restraining or repositioning the arms or legs.
Tonic - characterized by rigidity or stiffness, deviation of the eyes, dilation of the pupils, altered respiratory patterns. Consciousness is usually preserved. Tonic seizures most often occur during sleep and usually involve all or most of the brain, affecting both sides of the body.
Tonic-clonic - previously known as grand mal seizures; These commonly include muscle rigidity, violent involuntary muscle contractions, and loss of consciousness; may include difficulty breathing, loss of bowel or bladder control, and drowsiness or confusion after the seizure (PubMed Health, 2012).
Atonic - characterized by loss of tone. Eyelids may droop, head may nod, person may drop things and often the person will fall to the ground. These seizures are also called drop attacks. (Cross, 2004; Wolf, 2006; Epilepsy.com, 2010)
Unclassified or Cryptogenic: generalized convulsive seizures but undetermined focal or generalized onset (Jallon, 2001)
Seizures may also be either symptomatic or idiopathic in nature:
Symptomatic: A distinguished cause (such as head injury, brain tumor, identifiable abnormality) (Beghi, 2005)
Idiopathic: genetic cause (usually controlled by medication); not associated with increased risk of death
Seizures can further be classified as epileptic seizures or psychogenic nonepileptic seizures. Psychogenic nonepileptic seizures are behavioral events of psychological distress and are not due to neurological or other medical disorders (Hendrickson, 2014).
Pediatric Seizures may include jerking of the eyes, ocular fixation, eyelid fluttering, sucking, smacking, drooling, and "swimming, rowing, or pedaling" movements (NINDS, 2012).
Febrile seizure - seizure in association with a fever in the absence of CNS infection or acute electrolyte imbalance in a child older than 6 months of age without prior afebrile seizures; most common between ages 6 months and 6 years.
Infantile Spasms - characterized by a sudden contraction of flexor or extensor muscle groups of the head, trunk, or extremities. Onset tends to be between ages 4 and 7 months of age.
Lennox-Gaustaut syndrome - an electroclinical epilepsy syndrome characterized by the triad of electroencephalogram showing diffuse slow spike-and-wave discharges and paroxysmal fast activity, multiple intractable seizure types, and cognitive impairment (Wolf, 2006; Saneto, 2009).
Cognitive and psychiatric disorders are common co-morbidities of those with epilepsy while subjective forgetfulness is also common (El-Menshawi, 2014).
++
++
History - Difficult birth; head injury, meningitis, encephalitis; family history of epilepsy
Events of seizure - events preceding seizure; time of day, warning signs, bowel/bladder control or tongue biting during seizure; confusion, normal speech, headache, or muscle ache after seizure; a reliable witness or video is helpful to describe the events (Lipman, 2006).
A minimum of two unprovoked seizures are required for the diagnosis of epilepsy (Bromfield, 2006).
Laboratory studies include: blood testing for glucose, serum electrolytes, and calcium (Dunn, 2005); liver and renal function tests, urinalysis, toxicology screening (Sirven, 2005)
The best method to diagnose and classify the seizure is to observe the seizure and the associated electroencephalogram recording simultaneously (Epilepsy Foundation).
Lumbar puncture is used in neonates and patients with altered mental status; showing signs of meningeal irritation; with suspected CNS infection or cancer (Friedman, 2006 and Sirven, 2005).
Functional MRI (fMRI) is a dynamic test that can show the exact location of activity during seizure (RSNA, 2008); MRI is the preferred modality for clinical neuro-imaging
Adults presenting with their first, non-provoked seizure should have immediate neuro-imaging if possible; children without risk factors may be discharged without immediate neuro-imaging (American College of Emergency Physicians, 2014).
Patients at a higher risk for acute intracranial pathology should also receive immediate neuro-imaging (American College of Emergency Physicians, 2014).
A serum prolactin measurement is useful in distinguishing a generalized tonic-clonic seizure from a complex partial seizure from a pseudoseizure if done 10 to 20 minutes after the onset of the event (Chen, 2005).
Diagnostic instruments such as the 13-item questionnaire INCLEN Diagnostic Tool for Epilepsy have demonstrated high levels of sensitivity (85.8%) and specificity (95.3%) for diagnosing pediatric patients with epilepsy (Konanki, 2014).
Differential diagnosis: Conditions that mimic seizures include transient ischemic attack, stroke, syncope, migraine, and sleep disorders (Sirven, 2005).
++
++
A younger age at onset and longer duration of epilepsy is associated with a poor prognosis. Perinatal risk, age at onset, central nervous system infection, and antiepileptic drug use are prognostic indicators for not remaining seizure free for one year (Duman, 2017).
Over 60% of newly diagnosed patients will enter remission upon treatment (Kwan, 2004). For patients who discontinue their medication after remission, the incidence of seizure recurrence ranges from 12-66 % (French, 2008).
Approximately 25% to 35% of all infants with neonatal seizures exhibit cognitive impairment, motor impairment, or both. Seizures that occur in conjunction with prenatal asphyxia, intracranial infection, and prematurity with prolonged hypoglycemia have poor prognosis, with possible permanent neurologic sequelae such as cerebral palsy or cognitive impairment. Seizures that occur with subarachnoid hemorrhage without asphyxia or because of metabolic disorders have good prognosis if treatment begins early (Ronin, 2007; Mruk, 2015).
Epilepsy in childhood is associated with an increased risk of childhood death, but this risk is almost entirely confined to those with associated neurodevelopmental disorders. The risk of unexpected death in children with idiopathic epilepsy is extremely small (Nesbitt, 2012).
Psychogenic nonepileptic seizures are associated with poorer health-related quality of life in comparison to epileptic seizures after a study using somatization and alexithymia measures (Wolf, 2015).
++
++
Potential for learning disabilities, cognitive dysfunction, injuries from falls during seizures or seizures while driving a vehicle, permanent brain damage from ongoing uncontrolled seizures. Functional limitations vary greatly from minor to disabling. (NINDS, 2012).
Patients with epilepsy are at risk for two life-threatening conditions: status epilepticus and sudden unexplained death (NINDS, 2012).
Status epilepticus - continuous seizure activity lasting >30 minutes can lead to permanent brain damage. Mortality can be up to 33% with this condition (Gambrell, 2006).
Sudden unexplained death in epilepsy (SUDEP) – death of a person with epilepsy that occurs with no known cause, often during or after a seizure (Devinsky, 2011).
+++
IV. Medical/Surgical Management
++
++
Medication with antiepileptic drugs (AEDs) alone is successful in controlling seizures in 75-80% of those with epilepsy (Cross, 2004).
There are over 20 different antiepileptic drugs with the most common side effects being nausea, vomiting, and diarrhea (Cross, 2004; Gambrell, 2006). Many cause drowsiness or fatigue. Special care is needed for use during pregnancy to decrease chances of birth defects, minimize potential teratogenic effects, and decrease maternal mortality (Clarke, 2007).
Examples include valproate, benzodiazepines, gabapentin, phenytoin, pregabalin, and topiramate (Glauser, 2013).
Chronic use of antiepileptic drugs (AEDs) such as phenytoin, phenobarbital, carbamazepine, and primidone or non–enzyme-inducing AEDs such as valproate, are known to be associated with accelerated rate of bone loss. Therefore, vitamin D and calcium supplements should be used by patients who take AEDs to prevent bone loss (Lazzari, 2013).
Febrile seizures caused by birth asphyxia lead to severe alkalosis of brain tissue which can be treated by a therapeutic approach of applying air with CO2 in declining steps during the initial phase of recovery (Helmy, 2011).
A ketogenic diet of strict high fat and low carbohydrate intake may reduce seizures in children with intractable epilepsy (Martin, 2016).
Exposure to an auditory stimulus such as music can help reduce the occurrence of epilepsy and other seizure types; in some cases, complete cessation of seizures has occurred (Bodner, 2012).
++
++
About 30-40% of patients with epilepsy will have seizures that are resistant to multiple drug therapy. These patients may be candidates for surgery (Unnwongese, 2010).
Implantation of a vagus nerve stimulator may reduce the frequency of seizures in patients who are refractory to treatment with medication (Arya, 2013; Meneses, 2012).
Surgical options include temporal lobectomy or surgical resection of any affected area (Gambrell, 2006; Marks, 1998; Edelvik, 2013; West, 2015).
+++
V. Therapeutic Management
++
Possible Preferred Practice Pattern: 5A: Primary prevention/ risk reduction for loss of balance and falling, 5B: Impaired Neuromotor Development, 5C: Impaired Motor Function and Sensory Integrity Associated with Nonprogressive Disorders of the Central Nervous System - Congenital Origin or Acquired in Infancy or Childhood, 5D: Impaired Motor Function and Sensory Integrity Associated with Nonprogressive Disorders of the Central Nervous System - Acquired in Adolescence or Adulthood, 5E: Impaired Motor Function and Sensory Integrity Associated with Progressive Disorders of the Central Nervous System (APTA, 2003)
Therapeutic management of infants and children with seizure disorders with largely depend on the individual child's secondary activity and functional limitations.
Education and training with appropriate assistive device or technology for increased safety during ambulation and standing activities
Address delayed motor skills and impaired balance to improve performance of work, school, and play activities
Education on lifestyle changes: swimming with supervision, driving privileges, safety awareness, and injury prevention (Dunn, 2005).
Promote a consistent exercise program to promote a healthy lifestyle that can reduce the impact of epilepsy co-morbities such as depression and anxiety in addition to other health-related benefits (Arida, 2012).
Precautions and contraindications for physical therapy
Be aware of impaired arousal, attention, and cognition.
Do not apply electrotherapy near implanted stimulators.
Aquatic therapy – monitor for safety in the case of a seizure and for possible triggers of light flickering on water surface and changes in body temperature
Ask patients about any environmental conditions known to trigger seizures for the individual patient, such as bright light, patterned photic stimulation, unexpected auditory stimuli, fatigue, increased body temperature (NINDS, 2012; Schwartz, 2003).
In the event of a seizure, turn patient on his or her side and keep airway open, but do not put anything into the patient's mouth. Move nearby obstacles or protect the patient's head from injury with a pillow, but do not try to hold the person still. Time the seizure. Seek medical assistance if needed (Gambrell, 2004).
Patient and Family Education:
Identification of potential risk factors in the home to prevent risk of injury in the case of loss of consciousness
Instruction in proper monitoring of exercise intensity and cool down techniques to prevent overheating
Instruction in the safe use of assistive device for patients with decreased mobility or postural control (APTA, 2003).
Most states require a seizure-free period before a patient may resume driving; inform patient and family if unaware and work to accommodate their schedule
Standardized tests:
CNS Vital Signs (CNSVS) computerized battery (Gualtieri, 2006)
Strengths and Difficulties Questionnaire (SDQ) (He, 2013)
+
American Physical Therapy Association (APTA, 2017). Guide to Physical Therapist Practice. 2nd ed. Alexander, Va. 2003.
+
Arida
RM, Guimarães
de Almeida AC, Cavalheiro
EA, Scorza
FA. Experimental and clinical finding from physical exercise as a complementary therapy for epilepsy. Epilepsy Behav. 2013;26(3):273–278.
+
Arya
R, Greiner
HM, Lewis
A,
et al. Vagus nerve stimulation for medically refractory absence epilepsy. Seizure. 2013;22(4):267–270.
+
Beghi
E, Leone
M, Solari
A. Mortality in special groups mortality in patients with a first unprovoked seizure. Epilepsia. 2005;46:40–42.
+
Bodner
M, Turner
RP, Schwacke
J, Bowers
C, Norment
C. Reduction of seizure occurrence from exposure to auditory stimulation in individuals with neurological handicaps: a randomized controlled trial. PLoS One. 2012;7(10):1–8.
+
Bromfield
EB, Cavazos
JE, Sirven
JI. An Introduction to Epilepsy [Internet]. West Hartford, CT: American Epilepsy Society; 2006. Chapter 2, Clinical Epilepsy. Available from:
https://www.ncbi.nlm.nih.gov/books/NBK2511/ Accessed October 24, 2017.
+
Chen
DK, So
YT, Fisher
RS. Use of serum prolactin in diagnosing epileptic seizures: report of the Therapeutics and Technology Assessment Subcommittee of the American Academy of Neurology. Neurology. 2005:65:668–675.
+
Clarke
SD. Epilepsy in pregnancy. Br J Midwifery. 2007;15:740–5.
+
Cross
C. Seizures: regaining control. RN. 2004:67 44–51.
+
Devinsky
O. Sudden, unexpected death in epilepsy. N Engl J Med. 2011;365(19):1801–1811.
+
Dorsey
ER. Quality of life in epilepsy, multiple sclerosis, and beyond. Annals Neurol. 2007;62:307–308.
+
Duman
P, Varoglu
AO, Kurum
E. The long-term prognosis of epilepsy patients with medically treated over a period of eight years in Turkey. Pak J Med Sci. 2017;33(4):1007–1012.
+
Duncan
JS, Sander
JW, Sisodiya
SM, Walker
MC. Adult epilepsy. Lancet. 2006; 367(9516):1087–100.
+
Dunn
M, Breen
D, Davenport
R, Gray
A. Early management of adults with an uncomplicated first generalized seizure. J Emerg Med. 2005; 22: 237–242.
+
Edelvik
A, Rydenhag
B, Olsson
I,
et al. Long-term outcomes of epilepsy surgery in Sweden: a national prospective and longitudinal study. Neurology. 2013:81:1244–1251.
+
El-Menshawi
EE, Mohamed
MA, Zaher
AA. Forgetfulness among people with epilepsy. Egypt J Neurol Psychiatry Neurosurg. 2014;51(4):427–431.
+
Epilepsy Foundation. Epilepsy and seizure information for patients and health professionals. Available at:
www.epilepsy.com. Accessed March 31, 2017.
+
French
JA, Pedley
TA. Initial management of epilepsy. N Engl J Med. 2008;359(2):166–176.
+
Friedman
M, Sharieff
G. Seizures in children. Pediatr Clin North Am. 2006;53:257–277.
+
Gambrell
M, Flynn
N. Seizures 101. Nursing. 2004;34:36–42.
+
Glauser
T, Ben-Menachem
E, Bourgeois
B,
et al. Updated ILAE evidence review of antiepileptic drug efficacy and effectiveness as initial monotherapy for epileptic seizures and syndromes. Epilepsia. 2013;54(3):551–563.
+
Gualtieri
CT, Johnson
LG. Reliability and validity of a computerized neurocognitive test battery, CNS Vital Signs. Arch Clin Neuropsychol. 2006;21(7):623–643.
+
He
JP, Burstein
M, Schmitz
A, Merikangas
KR. The Strengths and Difficulties Questionnaire (SDQ): the factor structure and scale validation in U.S. adolescents. J Abnorm Child Psychol. 2013;41(4):583–595.
+
Helmy
MM, Tolner
EA, Vanhatalo
S, Voipio
J, Kaila
K. Brain alkalosis causes birth asphyxia seizures, suggesting therapeutic strategy. Ann Neurol. 2011;69:493–500.
+
Hendrickson
R, Popescu
A, Dixit
R, Ghearing
G, Bagic
A. Panic attack symptoms differentiate patients with epilepsy from those with psychogenic nonepileptic spells (PNES). Epilepsy Behav. 2014:37:210–214.
+
Jallon
P, Loiseau
P, Loiseau
J. Newly Diagnosed Unprovoked Epileptic Seizures: Presentation at Diagnosis in CAROLE Study. Epilepsia. 2001;42(4):464–475.
+
Konanki
R, Mishra
D, Gulati
S,
et al. INCLEN diagnostic tool for epilepsy (INDT-EPI) for primary care physicians: development and validation. Indian Pediatr. 2014;51(7):539–543.
+
Kwan
P, Sander
JW. The natural history of epilepsy: an epidemiological view. J Neurol Neurosurg Psychiatry. 2004;75(10):1376–1381.
+
Lazzari
AA, Dussault
PM, Thakor-James
M,
et al. Prevention of bone loss and vertebral fractures in patients with chronic epilepsy – antiepileptic drug and osteoporosis prevention trial. Epilepsia. 2013:54(11):1997–2004.
+
Lipman
M. Epilepsy: one disease, many causes. Consum Rep Health. 2006;18: 11.
+
Luciano
A. Results of Treatment Changes in Patients with Apparently Drug-Resistant Chronic Epilepsy. Ann Neurol. 2007;62:375–81.
+
Marks
M, Garcia
P. Management of seizures and epilepsy. Am Fam Physician. 1998;57:1589–1600.
+
Martin
K, Jackson
CF, Levy
RG, Cooper
PN. Ketogenic diet and other dietary treatments for epilepsy. Cochrane Database Syst Rev (Online). 2016;2:CD001903.
+
Meneses
MS, Rocha
SFB, Cristiane
S,
et al. Vagus nerve stimulation may be a sound therapeutic option in the treatment of refractory epilepsy. Arq Neuropsiquiatr. 2013;71(1):25–30.
+
Mruk
AL, Garlitz
KL, Leung
NR. Levetiracetam in neonatal seizures: a review. J Pediatr Pharmacol Ther. 2015;20(2):76–89.
+
Nesbitt
V, Kirkpatrick
M, Pearson
G, Colver
A, Forsyth
R. Risk and causes of death in children with a seizure disorder. Dev Med Child Neurol. 2012;54:612–617.
+
Ronen
GM, Buckley
D, Penney
S,
et al. Long-term prognosis in children with neonatal seizures: a population-based study. Neurology. 2007;69(19):1816–1822.
+
Saneto
R, Anderson
G. Onset of action and seizure control in Lennox-Gaustaut Syndrome: focus on Rufinamide. Ther Clin Risk Manage. 2009;5:271–280.
+
Schwartz
TH. Optical Imaging of Epileptiform Events in Visual Cortex in Response to Patterned Photic Stimulation. Cerebral Cortex. 2003;12(12):1287–1298.
+
Sirven
J, Ozuna
J. Diagnosing epilepsy in older adults: what does it mean for the primary care physician. Geriatrics. 2005;60:30–35.
+
Unnwongse
K, Wehner
T, Foldvary-Schaefer
N. Selecting patients for epilepsy surgery. Curr Neurol Neurosci Rep. 2010;10:299–307.
+
West
S, Nolan
SJ, Cotton
J,
et al. Surgery for epilepsy. Cochrane Database Syst Rev. 2015;(7):CD010541.
+
Wolf
LD, Hentz
JG, Ziemba
KS,
et al. Quality of life in psychogenic nonepileptic seizures and epilepsy: the role of somatization and alexithymia. Epilepsy Behav. 2015;43:81–88.
+
Wolf
S, McGoldrick
P. Recognition and management of pediatric seizures. Pediatr Ann. 2006;35:332–344.
++
Spinal cord tumors are divided into three categories based upon anatomic location: intradural intramedullary, intradural extramedullary, and extradural. (Chamberlain, 2015).
Intradural-intramedullary tumors (IMSCT):
Intramedullary spinal cord tumors are the rarest of primary spinal cord tumors.
IMSCT's comprise 20-30% of all primary intradural spinal cord tumors and 8-10% of total spinal cord tumors are IMSCT's (Chamberlain, 2015; Samartzis, 2015).
IMSCTs can include:
Ependymomas
Astrocytomas
Hemingoblastomas.
IMSCT's, like other tumors can be either malignant or benign. Tumors originating from other parts of the body can metastasize to the spinal cord itself or to the vertebrae and cause injury to the spinal cord (Chamberlain, 2009; Mayo Clinic, 2011).
Intradural-extramedullary tumor:
These tumors occur outside the spinal cord but inside the dura
These are the most common type of primary intraspinal tumors in adults, accounting for about 45% of all spinal tumors and 70-80% of primary intradural spinal cord tumors (Samartzis, 2015).
Some types of extramedullary tumors can be a result of drop down metastases by the CSF from types of malignant brain tumors.
Three dominant types:
Meningiomas
Develop in the spinal cord's arachnoid membrane, and are typically benign. Meningiomas are also most commonly found in women in the thoracic spine (Iacoangeli, 2012; Zadnik, 2013).
Schwannomas
Develop in the nerve roots that extend from the spinal cord (Chamberlain, 2009; Mayo Clinic, 2011).
Neurofibromas
Extradural-extramedullary tumor (epidural spinal cord compression):
Extradural tumors are the most common of all types of spinal cord tumors and are normally caused by systemic metastases.
They are most likely to be tumors of the spinal column and typically occur in the anterior spine or vertebral bodies.
These tumors usually result in epidural spinal cord compression. The most common tumor sources that cause epidural spinal cord compression are breast cancer (20%), lung cancer (13%), lymphoma (11%), and prostate cancer (9%) (Chamberlain, 2015; Samartzis, 2015).
The cause of most spinal tumors is unclear:
They may be related to genetic defects, inherited or spontaneous, or induced by something in the environment.
Occasionally spinal tumors are linked to known inherited syndromes, such as neurofibromatosis 2 and von Hippel-Lindau disease (Mayo Clinic, 2011).
Syringomyelia, a fluid-filled cavity in the spinal cord, has been shown with the development of IMSCTs., occurring most frequently with ependymomas (Samartzis, 2015)
+++
Incidence and Prevalence
++
Cancer affects 39.6% of people in the United States at some point in their lifetime.
Spinal cord injury (SCI) from tumors accounts for 26–45% of non-traumatic SCI
Primary spinal cord tumors comprise 2-4% of primary cancers of the CNS. Primary spinal cord tumors are 10 times less common than cranial tumors (Samartzis, 2015).
From 2007-2011, there were more than 340,000 brain and CNS tumors. Tumors in the spinal cord or cauda equine totaled 10,051 (Ostrom, 2014)
Of the 11,712 primary spinal tumors reported in the US between 2004 and 2007, the majority were non-malignant
The spinal cord is a potential site of metastases for 10% of primary cancers. Some of the most common sources of metastasis to the spinal cord are:
Peak age of incidence for spinal cord tumors from 2004-2007 according to the CDC:
Non-malignant tumors age 70-79 years.
Malignant tumors age 60-69 years
Children age 0-9 was the lowest rate at 0.23 per 100,000 persons.
+++
II. Diagnostic Procedures
++
For any patient with known cancer history and a new-onset of back pain, without a known etiology, strongly consider spinal metastasis until proved otherwise.
Clinical signs and symptoms often precede the diagnosis an average of several months to 2 years because of the slow-growing nature of intradural-extramedullary and intramedullary spinal cord tumors (Duong, 2012; Martin, 2003). The clinical presentation of most differential diagnoses is more "acute and abrupt" than the signs and symptoms of most intradural spinal cord tumors (Abul-Kasim, 2008).
The following signs suggest spinal tumors:
Progressive, unexplained, or nocturnal back or radicular pain
Segmental neurological deficits
Unexplained neurological deficits referable to the spinal cord or nerve roots, and
Unexplained back pain in patients with primary tumors in the lungs, breasts, prostate, kidneys, or thyroid or with lymphoma (Patchell, 2013).
Myelopathy can be caused by radiation to the spinal cord, with hallmark positive Lhermitte's sign (A sudden shock sensation brought on by neck flexion).
Increasing weakness is likely to occur in the arms with specifically cervical tumors. Increasing weakness can also occur in the legs with cervical tumors, as well as in thoracic and conus tumors. Bladder, bowel, sexual dysfunction, or any combination of the three often occurs early in all tumor types. Patients could also develop poor balance (Duong, 2012).
Rapid progression of paralysis and decreased walking ability likely indicates a malignant tumor rather than a benign intramedullary tumor.
++
Signs and Symptoms by type:
++
++
The imaging of choice is MRI with and without contrast. If patients are unable to tolerate MRI, CT scans may be performed to detect spinal cord tumors (Abul-Kasim, 2008; Chamberlain, 2015).
Computed tomography (CT), magnetic resonance imaging (MRI), angiography, and biopsy may be performed to confirm the cellular diagnosis and extent of the tumor. (John Hopkins, 2013)
Post-contrast axial MRI looking at sequences of the spinal cord may be helpful for differential diagnosis. (Adamec, 2011)
Diffusion tensor-fiber tracking can help to define the margins of an intramedullary spinal cord tumor in pediatric patients. (Choudhri, 2014)
Before an MRI, spinal x-rays, possibly taken for other reasons, may show bone destruction/erosion, vertebral pedicle widening, or paraspinal tissue distortion, especially if the tumor is metastatic. (Patchell, 2013)
++
Monitoring spinal tumors clinically and radiologically can lead to early treatment when symptoms and radiologic progression are present, leading to a more favorable and functional prognosis (Aboukais, 2015)
Predictors of good surgical outcome for intramedullary spinal cord tumors include (Chamberlain, 2009; Samartzis, 2016):
Functional status before surgery
Low histological grade of the tumor
Non-critical location of the tumor
Small size of the tumor
Decreased age of the patient
No difficulty in the surgical resection.
Survival Rates for individuals with Spinal Cord and Cauda Equina malignant tumors: (Ostrom, 2014)
1 yr, 88.9%
2 yrs, 84.4%
5 yrs, 79.5%
10 yrs, 75.6%
Prognosis after surgery:
With a mild preoperative neurological deficit and total excision achieved in 90% of ependymomas, functional improvement was obtained (Chiba, 2008; Özkan, 2015).
Functional outcomes after surgery depend on duration of symptoms, preoperative neurological conditions, and tumor localization with respect to the spinal cord axis (Jadvyga, 2010).
In cases of intramedullary spinal cord tumors, early diagnosis and early surgery, before the paralysis becomes severe are important to obtain good functional outcomes (Nakamura, 2008; Raco, 2005)
Total resection may be achieved and is often curative (Chamberlin, 2009; 2015).
Total resection of high-grade astrocytomas is unlikely due to their growth into the spinal cord (Raco, 2005; Chamberlain, 2015)
Differing from high-grade tumors, total excision was complicated and functional outcomes were poor.
It is vital that extra attention be given to tumors that have feeding arteries on the ventral side and associated with von Hippel-Lindau disease (Chiba, 2008)
Microsurgical resection of cervically-located intramedullary astrocytomas have a better prognosis than those in the thoracolumbar region (Ardeshiri, 2013).
Resection of benign intradural spinal cord tumors has been independently associated with these negative outcomes (Kalakoti, 2015):
inpatient mortality
unfavorable discharge
prolonged length of hospital stay
high-end hospital charges
neurological complications
DVT
pulmonary embolism
wound complications
wound infection
cardiac complications
Impairments caused by treatment
One specific myelopathy is caused by a late effect of radiation to the cord 6-36 months post-radiation.
Impairments caused by chemotherapy (Schagen, 2013):
+++
IV. Medical and Surgical Management
++
+++
V. Therapeutic Management
++
++
Pattern 4I: Impaired Joint Mobility, Motor Function, Muscle Performance, and Range of Motion Associated With Bony or Soft Tissue Surgery (APTA, 2015)
General PT options: physical therapist can provide musculoskeletal treatment, range of motion, and stretching, that will be needed after surgery for prevention of contractures, education (Backus, 2009).
Pattern 5A: Primary Prevention/Risk Reduction for Loss of Balance and Falling (APTA, 2015)
General PT options: pre-gait/ gait training activities, transfers, and balance training, education (Backus, 2009).
Pattern 5E: Impaired Motor Function and Sensory Integrity Associated with Progressive Disorders of the Central Nervous System (APTA, 2015).
Pattern 5H: Impaired Motor Function, Peripheral Nerve Integrity, and Sensory Integrity Associated with Nonprogressive Disorders of the Spinal Cord (APTA, 2015).
Pattern 6B: Impaired Aerobic Capacity/Endurance Associated With Deconditioning (APTA, 2015)
General PT options: gait endurance (treadmill), strengthening activities, aquatic exercises (Backus, 2009).
Pattern 7A: Primary Prevention/Risk Reduction for Integumentary Disorders (APTA, 2015)
Pattern 7E: Impaired Integumentary Integrity Associated With Skin Involvement Extending Into Fascia, Muscle, or Bone and Scar Formation (APTA, 2015)
General PT options: wound care, skin management for the prevention of infection/ulcers/adhesions, education (Backus, 2009)
Overall general PT options: (APTA, 2015; APTA, 2017)
Patient or Client Instruction
Biophysical Agents
Functional Training in Self-Care and in Domestic, Education, Work, Community, Social, and Civic Life
Manual Therapy Techniques
Motor Function Training
Motor control
Motor coordination
Balance
Therapeutic Exercise
Contraindications/precautions
Ultrasound: Use for palliation of pain in end-stage disease may be allowed (Goodman, 2009).
Diathermy
Low Level Laser Therapy
"Avoiding modalities such as heat or cold or any topical agents over skin being irradiated is important for skin protection, because poor circulation inhibits normal heat and cold dissipation".
When irradiation is completed and skin is healed, based on skin integrity and circulation, heat, cold, or transcutaneous nerve stimulation (TNS) may be used.
Health care team contributions
Keeping lines of communication open between the oncologist, neurologist, nurses, occupational therapists, psychologists, and locations of discharge such as nursing homes, SNF, and family members will promote the best treatment for the patient (New, 2005pg257).
Patients should be evaluated postoperatively by physical therapists and occupational therapists to determine the extent of therapy needed and whether the patient is a candidate for inpatient or outpatient services (Tredway, 2014).
+++
VI. Consumer & Professional Resources
++
Online Communities and Support
++
What Next and Cancer Survivors Network are all online communities and resources for individuals with cancer.
Cancer Support Community™ provides information about cancer and support for patients and their families.
Road to Recovery is a specialized cancer program that provides patients without a way to their cancer treatment a ride (American Cancer Society).
"I Can Cope classes can help you and your loved ones learn about cancer and how to take care of yourselves. The more you know about cancer, the better you can handle your treatment and decisions about your care (American Cancer Society)."
Look Good…Feel Better: A volunteer cosmetologists helps women with cosmetic and skin care application to deal with changes that occur from cancer treatments. (American Cancer Society).
Reeve Foundation Peer and Family Support Program is a peer to peer mentoring program designed to help newly paralyzed persons and their families cope with their new life (Christopher and Dana Reeve Foundation).
The Spinal Cord Tumor Association provides numerous resources to patients recently diagnosed with a spinal cord tumor. They also have discussion boards and frequently asked questions about the disease (Spinal Cord Tumor Association).
Cancer Support Community provides online message boards with over 20 different topics. These are open 24 hours a day, 7 days a week for those affected by cancer to receive advice, support, and offer tips based on their own experiences.
+
Aboukais
R, Baroncini
M, Zairi
F, Bonne
N, Schapira
S, Vincent
C, Lejune
J. Prognostic value and management of spinal tumors in neurofibromatosis type 2 patients.
Acta Neurochir (Wien). 2013; 155(5):771–777.
[PubMed: 23381342]
+
Abul-Kasim
K, Thurnher
MM, Mckeever
P, Sundgren
PC. Intradural spinal tumors: current classification and MRI features.
Neuroradiology. 2008; 50(4):301–14.
[PubMed: 18084751]
+
Adamec
I, Brinar
V, Habek
M. Spinal cord tumor versus transverse myelitis.
Spine J. 2011; 11(12):1143–1145.
[PubMed: 22082692]
+
American Physical Therapy Association (APTA). Guide to Physical Therapist Practice 3.0. 2015: DOI: 10.2522/ptguide3.0_ 978-1-931369-85-5. Accessed May 20, 2015.
+
Ardeshiri
A, Chen
B, Hütter
B-O, Chen
B, Hutter
B, Oezkan
N, Wanke
I, Sure
U, Sandalcioglu
E. Intramedullary spinal cord astrocytomas: the influence of localization and tumor extension on resectability and functional outcome.
Acta Neurochir (Wien). 2013; 155(7):1203–1207.
[PubMed: 23700256]
+
Armstrong
TS, Gilbert
MR. Metastatic brain tumors: diagnosis, treatment, and nursing interventions.
Clin J Oncol Nurs. 2000; 4(5):217–225.
[PubMed: 11111453]
+
Backus
D, Bensimon
L, Gassaway
J, LaBarbera
J, McDowell
S, Mumma
S, Natale
A, Taylor
S, Zanca
JM. Classification of SCI rehabilitation treatments SCIRehab: the physical therapy taxonomy.
J Spinal Cord Med. 2009; 32(3):270–82.
[PubMed: 19810629]
+
Chamberlain
MC, Grimm
S. Adult primary spinal cord tumors. Expert Rev Neurother. 2009; 10(Oct9):1487–95.
+
Chamberlain
MC. Neoplastic myelopathies.
Continuum. 2015; 21(1):132–45.
[PubMed: 25651222]
+
Chiba
K, Ishii
K, Matsumoto
M, Nakamura
M, Takaishi
H, Toyama
Y, Tsuji
T, Watanabe
K. Surgical treatment of intramedullary spinal cord tumors: prognosis and complications. Spinal Cord. 2008 Apr; 46(4):282–6.
+
Choudhri
AF, Whitehead
MT, Jr
PK, Montgomery
BK, Boop
FA. Diffusion tensor imaging to guide surgical planning in intramedullary spinal cord tumors in children.
Neuroradiology (2014) 56:169–174
[PubMed: 24395215]
+
Dolecek
TA, Propp
JM, Stroup
NE, Kruchko
C. CBTRUS Statistical Report: Primary Brain and Central Nervous System Tumors Diagnosed in the United States in 2005–2009.
Neuro Oncology. 2012; 14(suppl 5): v1–v49.
[PubMed: 23095881]
+
Duong
LM, McAthy
BJ, McLendon
RE, Dolecek
TA, Kruchko
C, Douglas
LL, Ajani
UA. Descriptive Epidemiology of Malignant nonmalignant primary spinal cord, spinal meninges, and cauda equina tumors, United States, 2004-2007.
Cancer. 2012; 118(17);4220–4227
[PubMed: 22907705]
+
Fąfara-leś
A, Kwiatkowski
S, Maryńczak
L, Kawecki
Z, Adamek
D, Herman-Sucharska
I, Kobylarz
K. Torticollis as a first sign of posterior fossa and cervical spinal cord tumors in children.
Childs Nerv Syst. 2014; 30(3):425–30.
[PubMed: 23955178]
+
Iacoangeli
M, Gladi
M, Di Rienzo
A,
et al. Minimally invasive surgery for benign intradural extramedullary spinal meningiomas: experience of a single institution in a cohort of elderly patients and review of the literature.
Clin Interv Aging. 2012;7:557–564.
[PubMed: 23271902]
+
Ito
T, Sawakami
K, Ishikawa
S, Hirano
T, Endo
N, Kakita
A, Takakashi
H. Progression of paralysis is the most useful factor for differentiating malignant from benign intramedullary tumors.
Spinal Cord. 2013; 51(4):319–321.
[PubMed: 23208538]
+
Kalakoti
P, Missios
S, Menger
R, Kukreja
S, Konar
S, Nanda
A. Association of risk factors with unfavorable outcomes after resection of adult benign intradural spine tumors and the effect of hospital volume on outcomes: an analysis of 18, 297 patients across 774 US hospitals using the National Inpatient Sample (2002-2011). Neurosurg Focus. 2015; 39(2):E4:1–13.
+
Martin
DS, Woolsey
RM. Intradural Extramedullary Tumors. In: Lin
VW, Cardenas
DD, Cutter
NC. Spinal Cord Medicine: Principles and Practice. New York: Demos Medical Publishing; 2003. Available from:
http://www.ncbi.nlm.nih.gov/books/NBK9254. Accessed March 11, 2013.
+
New
PW. Functional outcomes and disability after nontraumatic spinal cord injury rehabilitation: results from a retrospective study.
Arch Phys Med Rehabil. 2005; 86:250–61.
[PubMed: 15706551]
+
New
P, Tan
M. Survival after rehabilitation for spinal cord injury due to tumor: a 12-year retrospective study.
J Neurooncol. 2011; 104:233–238.
[PubMed: 21104296]
+
Nzokou,
A, Weil,
A, Shedid,
D. Minimal Invasive Removal of Thoracic and Lumbar Spinal Tumors Using a Nonexpandable Tubular Retractor. Journal of Neurosurgery. 2013; 6:708–715.
+
Ostrom
QT, Gittleman
H, Liao
P, Rouse
C, Chen
Y, Dowling
J, Wolinsky
Y, Kruchko
C, Barnholtz-Sloan
J. CBTRUS statistical report: primary brain and central nervous system tumors diagnosed in the United States in 2007-2011.
Neuro-oncology. 2014; 16 Suppl 4:iv1–63.
[PubMed: 25304271]
+
Özkan
N, Jabbarli
R, Wrede
KH, Sariaslan
Z, Stein
KP, Dammann
P, Ringelstein
A, Sure
U, Sandalcioglu
EI. Surgical management of intradural spinal cord tumors in children and young adults: A single-center experience with 50 patients.
Surg Neurol Int. 2015; 6(Suppl 27):S661–7.
[PubMed: 26713174]
+
Raco
A, Esposito
V, Lenzi
J, Piccirilli
M, Delfini
R, Cantore
G. Long-term follow-up of intramedullary spinal cord tumors: a series of 202 cases.
Neurosurgery. 2005; 56(5):972–81.
[PubMed: 15854245]
+
Raj
VS, Lofton
L. Rehabilitation and treatment of spinal cord tumors.
J Spinal Cord Med. 2013;36(1):4–11.
[PubMed: 23433329]
+
Samartzis
D, Gillis
CC, Shih
P, O'Toole
JE, Fessler
RG. Intramedullary Spinal Cord Tumors: Part I—Epidemiology, Pathophysiology, and Diagnosis.
Global Spine J 2015;5(5):425–435.
[PubMed: 26430598]
+
Samartzis
D, Gillis
CC, Shih
P, O'Toole
JE, Fessler
RG. Intramedullary Spinal Cord Tumors: Part II—Management Options and Outcomes.
Global Spine J. 2016;6(2):176–185.
[PubMed: 26933620]
+
Sandalcioglu
I, Gasser
T, Asgari
S,
et al. Functional outcome after surgical treatment of intramedullary spinal cord tumors: experience with 78 patients.
Spinal Cord. 2005; 43(1):34–41
[PubMed: 15326473]
+
Schagen
SB, Wefel
JS. Chemotherapy-related changes in cognitive functioning. EJC Supplements. 2013; 11(2):225–232.
+
Schneider
C, Hidalgo
ET, Schmitt-Mechelke
T, Kothbauer
KF. Quality of life after surgical treatment of primary intramedullary spinal cord tumors in children: Clinical article. J Neurosurg Pediatrics 13:170–177, 2014.
+
Tredway
TL. Minimally invasive approaches for the treatment of intramedullary spinal tumors.
Neurosurg Clin N Am. 2014; 25(2):327–36.
[PubMed: 24703450]
+
Zadnik
PL, Gokaslan
ZL, Burger
PC, Bettegowda
C. Spinal cord tumours: advances in genetics and their implications for treatment.
Neurology. 2013; 9(5):257–266.
[PubMed: 23528542]
+++
Spinal Muscular Atrophy
++
Spinal muscular atrophy (SMA) is an autosomal- recessive disorder affecting lower motor neurons, specifically anterior horn cells of the spinal cord and, at times, the motor nuclei of the lower cranial nerves (Whitehead, 2002; Brian, 2004).
Caused by homozygous deletion, rearrangement, or mutation of the survival motor neurons (SMN1) gene on chromosome 5q12.2.
SMN1 & SMN2 are responsible for making SMN proteins for the maintenance of anterior horn cells (Whitehead, 2002; Oskoui, 2007).
When SMN 1, the largest maker of proteins, portion of the gene is deleted then SMN2 is relied on. However, most of SMN proteins made by SMN2 are not functional. The amount of SMN protein made in someone with SMA depends on how many copies of SMN2 they have. The number of SMN2 copies correlates with severity of the disease presentation.
SMN protein is expressed in the spinal cord and brain; however there is preservation of the corticomotoneuronal function. There seems to be an adaptive response by increased corticomotoneuronal projections to surviving spinal motoneurons (Farrar, 2012).
This mutation and insufficient amount of SMN protein leads to anterior horn cell loss resulting in muscular weakness and atrophy (Guillot, 2007)
Equal occurrence in genders (Iannaccone, 2007)
++
++
No cure for SMA currently available (Lunn, 2008).
The second most common fatal autosomal recessive disorder after cystic fibrosis (estimated incidence of 1 in 6,000 to 1 in 10,000 live birth (D'Amico, 2011).
The overall prognosis for SMA is poor due to progressive disease nature. The earlier the onset the poorer the outcome. The spectrum of debilitation associated with SMA ranges from almost complete paralysis to mild muscle weakness (Bach, 2002).
Outcome measures: gross motor functional measures seem to be the most reliable as compared to muscle strength testing. Pulmonary function tests in children require special training but are useful to monitor disease progression as mortality is associated with respiratory failure (Iannaccone, 2002).
Poor nutrition is a common resulting from bulbar weakness; decreased physical activity increases risk factor s for obesity (Burnett, 2009).
Decreased feeding is often a first sign of progressive weakness. The child often encounters prolonged feeding, coughing with feeding, weight loss, and extreme fatigue after (Mercuri, 2012).
Current research emphasizes search for cure, prevention of disease, improving accurateness of diagnosis, and exploration of genetic implications (Prior, 2007).
+++
IV. Medical/Surgical Management:
++
Overall management depends on severity and type of SMA.
Maintaining optimal respiratory function is a priority (Iannaccone, 2003). Careful observation and prevention of respiratory infection.
Respiratory muscle assistive devices aid in ventilation and prevent chest deformities (i.e., pectus excavatum) (Bach, 2008); Non-invasive ventilation aids (i.e. biphasic positive airway pressure) prolong necessity for tracheostomy (Burnett, 2009).
Non-Invasive ventilation can be used as early as neonatal period (Mercuri, 2012).
Pharmaceuticals: valproic acid (VPA) and phenylbutyrate prescribed to increase SMN protein levels and offset deterioration of motor neurons (Wirth, 2006).
Gastrostomy placement is often used when swallowing dysfunctions are present (Sproule, 2012)
Surgical techniques: Contracture releases, spinal fusion to correct scoliosis, and tracheostomy.
Tracheostomy provides lifetime ventilatory support, exacerbates airway secretion production, hinders speech development and creates swallowing difficulties (Bach, 2008).
Nutritional management: Amino acid diets have not proved to have significant effects on survival (Oskoui, 2007); Caloric requirements need to be assessed frequently; Gastrostomy tube required in cases with significant failure to thrive; Clinicians aim for a body weight somewhat less than typical ranges for height and age (Burnett, 2009).
Pharmaceutical research is currently in progress. The goal is to develop a medication that can slow, stop, or reverse the progression of the degradation of the motor neurons. There have been several clinical trials that showed no improvement in functional scales and were discontinued. Several other drug compounds have shown significant improvement in animal models and will move to the human phase of clinical trials soon (Kaufmann, 2008; Pruss, 2010; Markowitz, 2012).
+++
V. Implications of Therapeutic Management:
++
Preventative measures address musculoskeletal and cardiopulmonary systems, specifically, scoliosis and respiratory function. Postural education and bracing are used to address the scoliosis component while chest physiotherapy or airway clearance techniques are used to address the respiratory function (MDA, 2003).
General physical therapy options include ROM and strengthening to slow the deterioration of muscles, endurance training to address fatigue, facilitation of functional mobility and ADL. The buoyancy environment of aquatic therapy allows patients with SMA to work on ROM, muscle strengthening and respiratory endurance (MDA, 2007, O'Hagen, 2007); Milder forms of SMA respond to gait training.
In the most severe cases of SMA (type I), PT treatment should be aimed at improving quality of life for the child and their family.
Assistive devices or adaptive equipment include supine, prone, and dynamic standers; rolling walkers (anterior or posterior); wheelchairs; splints, braces or ankle-foot orthoses; adaptive tricycles.
Physical therapy assessments allow ambulatory patients (SMA III) to maintain independence and mobility by determining appropriate assistive devices, adaptive equipment, and energy conservation techniques (Wang, 2007).
Comprehensive multidisciplinary approach involves specialists in neurology; orthopedic surgery; critical care; physical medicine; physical, occupational and respiratory therapy; and clinical nutrition working together as team to provide total care (Iannaccone, 2007).
Starting physical therapy at an early age in individuals with SMA may enhance central adaptive processes. This may potentially promote stabilization and improvement of motor function. Additionally, physical therapy treatment may be complemented by pharmacological treatments that attempt to increase central plasticity (Farrar, 2012).
The 6 Minute Walk Test has been shown to be a viable clinical outcome measure for ambulatory SMA patients as it correlates with the Hammersmith Functional Motor Scale-Expanded and is sensitive to fatigue related changes (Montes, 2010).
In patients undergoing soft tissue releases, rapid and aggressive physical therapy may improve outcomes (Wang, 2007). Hammersmith Functional Motor Scale – tool developed to track motor changes in patients with type II and type III, particularly those with decreased mobility. Learning to use this tool could be particularly useful when working with these children, so that progress can be systematically tracked and documented (Main, 2003). This was an outcome tool used in the recent drug trials (Markowitz, 2012).
+++
VI. Consumer and professional resources
++
+
Bach
J. The use of mechanical ventilation is appropriate in children with genetically proven spinal muscular atrophy type I: the motion for. Pediatr Respir Rev. 2008;9:45–50.
+
Bach
J, Baird
J, Plosky
D, Navado
J, Weaver
B. Spinal muscular atrophy type 1: management and outcomes.
Pediatr Pulmonol. 2002;34:16–22.
[PubMed: 12112792]
+
Bach
J, Niranjan
V, Weaver
B. Spinal muscular atrophy type 1: a noninvasive respiratory management approach.
Chest. 2000;117:1100–1105.
[PubMed: 10767247]
+
Brian
H, Virginia
C, Patrick
I. Spinal muscular atrophy: survival pattern and functional status. Pediatrics. 2004; 114: 548–553.
+
Burnett
B, Crawford
T, Sumner
C. Emerging treatment options for spinal muscular atrophy.
Curr Treat Options Neurol. 2009;11:90–101.
[PubMed: 19210911]
+
Chng
S, Wong
Y, Hui
J,
et al. Pulmonary function and scoliosis in children with spinal muscular atrophy types II and III.
J Paediatr Child Health. 2003;39(9):673–676.
[PubMed: 14629498]
+
D'amnico
A, Mercuri
E, Tiziano
F, Bertini
E. Spinal muscular atrophy. Orphanet J Rare Dis. 2011;(6)1:art. no. 71.
+
Elsheikh
B, Prior
T, Zhang
X, Miller
R, Kolb
SJ, Moore
D,
et al. An analysis of disease severity based on SMN2 copy number in adults with spinal muscular atrophy.
Muscle Nerve. 2009;40:652–656.
[PubMed: 19760790]
+
Evans
G, Drennan
J, Russman
B. Functional classification and orthopaedic management of spinal muscular atrophy. J Bone Joint Surg. 1981;63:516–522.
+
Farrar
MA, Vucic
S, Lin
CS-Y,
et al. Dysfunction of axonal membrane conductances in adolescents and young adults with spinal muscular atrophy.
Brain. 2011;134:3185–3197.
[PubMed: 21926101]
+
Guillot
N, Cuisset
J, Cuvellier
J, Hurtevent
J, Joriot
S, Vallee
L. Unusual clinical features in infantile spinal muscular atrophies.
Brain Dev. 2007;30:169–178.
[PubMed: 17804187]
+
Iannaccone
S, AmSMART
Group. Outcome measures for pediatric spinal muscular atrophy. Arch Neurology. 2002;59:1445–1450.
+
Iannaccone
S, Hyman
L. Reliability of 4 outcome measures in pediatric spinal muscular atrophy. Arch Neurology. 2003;60:1130–1136.
+
Iannaccone
S. Modern management of spinal muscular atrophy.
J Child Neurol. 2007;22:974–978.
[PubMed: 17761652]
+
Kaufmann
P, Iannaccone
ST. Clinical trials in spinal muscular atrophy.
Phys Med Rehabil Clin N Am. 2008;19(3):653–660.
[PubMed: 18625422]
+
Lomen-Hoerth
C, Slawnych
MP. Statistical motor unit number estimation: from theory to practice.
Muscle Nerve. 2003;28(3):263–272.
[PubMed: 12929186]
+
Lunn
MR, Wang
CH. Spinal muscular atrophy.
Lancet. 2008;371:2120–2133.
[PubMed: 18572081]
+
Main
M, Kairon
H, Mercuri
E, Muntoni
F. The Hammersmith Functional Motor Scale for Children with Spinal Muscular Atrophy: a scale to test ability and monitor progress in children with limited ambulation.
Eur J Paediatr Neurol. 2003;7(4):155–159.
[PubMed: 12865054]
+
Markowitz
J. Spinal muscular atrophy: a clinical and research update.
Pediatr Neurol. 2012;46(1):1–12.
[PubMed: 22196485]
+
Medical Research Council: Nerve Injuries Research Committee. Aids to the investigation of peripheral nerve injuries.
Brain. 2010;133:2838–2844.
[PubMed: 20928945]
+
Mercuri
E, Bertini
E, Iannaccone
S. Childhood spinal muscular atrophy: controversies and challenges.
The Lancet Neurol. 2012;11(5):443–452.
[PubMed: 22516079]
+
Montes
J, McDermott
MP, Martens
WB,
et al. Six-Minute Walk Test demonstrates motor fatigue in spinal muscular atrophy.
Neurology. 2010;74:833–838.
[PubMed: 20211907]
+
Muscular Dystrophy Association. Spinal muscular atrophy fact sheet page. Available at:
http://www.mda.org. Accessed January 19, 2007.
+
O'Hagen
JM, Glanzman
AM, McDermott
MP,
et al. An expanded version of the Hammersmith Functional Motor Scale for SMA II and III patients.
Neuromuscul Disord. 2007;17:693–697.
[PubMed: 17658255]
+
Oskoui
M, Levy
G, Garland
CJ,
et al. The changing natural history of spinal muscular atrophy type 1.
Neurology. 2007;69:1931–1936.
[PubMed: 17998484]
+
Prior
T. Spinal muscular atrophy diagnostics.
J Child Neurol. 2007;22:952–956.
[PubMed: 17761649]
+
Prior
TW, Nagan
N. Spinal muscular atrophy: overview of molecular diagnostic approaches. Curr Protoc Hum Genet. 2016;88:9.27.1–9.27.13.
+
Pruss
RM, Giraudon-Paoli
M, Morozova
S,
et al. Drug discovery and development for spinal muscular atrophy: lessons from screening approaches and future challenges for clinical development.
Future Med Chem. 2010;2(9):1429–1440.
[PubMed: 21426138]
+
Sugarman
EA, Nagan
N, Zhu
H,
et al. Pan-ethnic carrier screening and prenatal diagnosis for spinal muscular atrophy: clinical laboratory analysis of >72 400 specimens.
Eur J Hum Genet. 2012;20(1):27–32.
[PubMed: 21811307]
+
Sproule
DM, Hasnain
R, Koenigsberger
D, Montgomery
M, DeVivo
D, Kaufmann
P. Age at disease onset predicts likelihood and rapidity of growth failure among infants and young children with spinal muscular atrophy type 1 and 2.
J Child Neurol. 2012;27(7):845–851.
[PubMed: 22467740]
+
Wadman
RI, Vrancken
AFJE, van den Berg
LH, van der Pol
WL. Dysfunction of the neuromuscular junction in spinal muscular atrophy types 2 and 3.
Neurology. 2012;79:2050–2055.
[PubMed: 23115209]
+
Wang
CH, Finkel
RS, Bertini
ES,
et al. Consensus statement for standard of care in spinal muscular atrophy.
J Child Neurol. 2007;22:1027–1049.
[PubMed: 17761659]
+
Whitehead
S, Jones
K, Xing
Z, Xiaodong
C, Terns
R, Terns
M. Determinants of the interaction of the spinal muscular atrophy disease protein SMN with the dimethylargine-modified box H/ACA small nucleolar ribonucleoprotein GAR1.
J Biol Chem. 2002;277:48087–48093.
[PubMed: 12244096]
+
Wee
D, Kong
L, Sumner
C. The genetics of spinal muscular atrophies.
Curr Opin Neurol. 2010;23:450–458.
[PubMed: 20733483]
+
Wirth
B, Brichta
L, Schrank
B,
et al. Mildly affected patients with spinal muscular atrophy are partially protected by an increased SMN2 copy number. Hum Genet. 2006;439:156–170.
++
- Syringomyelia is a rare chronic, progressive, disorder of the spinal cord.
- It most frequently affects children and young adults between the age of 30-40 (Heiss, 2012; Hilton, 2003; Rai, 2015).
- Syringomyelia is not a single disease, but rather a descriptive term used for any fluid filled cavity in the spinal cord (Michals, 1996), of which there are four types.
Normally a syrinx, or fluid filled cavity, develops paramedian in the spinal cord, almost exclusively in the cervical region, but may extend up into the medulla oblongata (syringobulbia) or down to the thoracic and/or lumbar region.
The development of the syrinx can have damaging effects on the grey matter that encompasses it as well as ascending and descending tracts, and the decussating fibers of the lateral spinothalamic tract, impairing bilateral pain and temperature sensations within the region. (Smith, 2016)
It can also form within the central canal, which is commonly termed hydromyelia. The accumulation of cerebral spinal fluid in the syrinx causes increased pressure inside the spinal cord, expanding the cavity and compressing the adjacent nerve fibers (Vandertop, 2014).
- Syringomyelia is usually associated with congenital malformations, such as Chiari Malformation, but could also be secondary to spinal cord trauma or tumors. (Hudson, 2008)
- Four Types:
Type I: Syringomyelia with obstruction of the foramen magnum and dilation of the central canal
Type II: Syringomyelia without obstruction of the foramen magnum (idiopathic)
Type III: Syringomyelia with other diseases of the spinal cord
Type IV: Pure hydromyelia with or without hydrocephalus
The four types are also known as communicating, noncommunicating, atrophic cavitation, and neoplastic cavitation. (Rai, 2015)
- Known Causes (Vandertop, 2014)
The most common known causes of syringomyelia are congenital neural tube defects or Chiari malformations.
Klippel-Feil is a rare genetic cause of syringomyelia.
Due to the nature of this disorder any type of CSF disturbance in the spinal cord has a chance to develop into syringomyelia.
Syringomyelia can also be acquired related to these mechanisms:
Hydrocephalus, post-infection, post-inflammatory, post-traumatic, and stenosis of the spinal cord.
Some patients presenting with shoulder dysfunction or impairments should be evaluated for cervical spinal pathology, especially after trauma to the area. (Zhang, 2016)
Studies have shown that those with Chiari I malformations with syringomyelia have abnormal spinal canal diameters in comparison to those with Chiari I malformations without syringomyelia; this can increase the flow of CSF velocity through the subarachnoid space in the upper cervical spine, thus affecting CSF dynamics. (Struck, 2016)
Idiopathic syringomyelia in comparison to the control subjects also involves abnormal cervical spinal canal diameters, that can influence the pathogenesis of syringomyelia and the fluid dynamics of CSF. (Struck, 2016)
- Incidence (Brodbelt, 2003; Rai, 2015)
There is limited evidence on the prevalence of syringomyelia in the United States.
However, the estimated prevalence is 9 per 100,000, with incidence rate of .44 new cases per year. This suggests that 22,000 Americans are affected with this condition each year.
Type I seems to be the most common type with about half of patients presenting alongside a Chiari malformation. The other three types have varying frequency with trauma slightly higher than the others.
It is estimated that 0.3-3% of the population with spinal cord injury are affected by post-traumatic syringomyelia. (Ushewokunze, 2010)
- Etiological Theories:
No theories as to the origination of this disease process have been supported by evidence.
One theory is that normal CSF flow is prevented by a congenital failure of the opening of the outlets of the fourth ventricle.
Others have suggested that edema is a major pathogenic factor, induced by angiomatus malformations, neoplasms, trauma, arachnoiditis, etc.
Hydrodynamic theories propose that fluid is forced out of the fourth ventricle into the central canal due to increased arterial or respiratory pressure effects.
When trauma occurs, the subarachnoid channels can become fibrotic at the location of the injury, which inhibits the flow of CSF. (Ushewokunze, 2010)
Craniospinal CSF pressure dissociation theory
Proposes that the main cause of syringomyelia is due to a localized subarachnoid obstruction which can induce a form of mechanical stress on the spinal cord caused by non-uniform contraction and expansion of the surrounding veins and capillaries. This mechanical stress then leads to tissue damage allowing fluid to accumulate. (Levine, 2004)
The volume change theory evolved from the craniospinal CSF pressure dissociation theory.
Pathophysiologically the volume change in the cervical spinal canal increases more with cervical flexion than cervical extension. The subarachnoid spaces of the cerebrum and spinal column are blocked during positional changes from flexion to extension and the spinal canal compensates by forming a central syrinx, leading to deformation. Since the cervical and lumbar spinal cord exhibit similar enlargements, the same process of syrinx formation can occur at cervical and lumbar levels (Rai, 2015).
Any of these theories may lead to central spinal cord degeneration.
Anterior white commissure fibers are interrupted, namely the bilateral fibers of lateral spinothalamic tract as they cross
If untreated, it will extend out and compress the anterior horn cells, causing ipsilateral lower motor neuron lesion symptoms at the level of the extension.
++
- A clinical neurologic evaluation including measuring central motor conduction times should be performed (Robinson, 1990).
- Use of highly sensitive clinical screening tests along with imaging provides optimal diagnosis (Hudson, 2008).
- Signs and Symptoms
Most signs and symptoms will be directly related to the location, size, and extent of the cavity.
Some of the most common, depending on size and location of syrinx, include:
The most obvious sign of syringomyelia is a cape-like loss of pain and temperature along the back and arms, referred to as dissociative anesthesia. Proprioception and motor function are still intact as this point. (Snoddy, 2017; Vandertop, 2014; Hudson, 2008; Hilton, 2003)
Later development of paresis and muscle atrophy of the bilateral hands and arms with loss of tendon reflexes as the syrinx grows larger. (Snoddy, 2017)
Other symptoms can include:
Weakness and ataxia of the upper extremities, hyperreflexia, spastic hypertonia, Charcot's joints, hypoesthesia, and a loss of bowel and bladder control can also occur. The patient may complain of a headache and inconsistent chronic pain. (Wang, 2017; Hudson, 2008; Hilton, 2003)
Syringomyelia is the most common cause of Charcot joints (neuropathic arthropathy) in upper extremities, specifically the shoulder and the elbow. It is important to take a thorough history and evaluation to correctly identify the underlying cause of the neuroarthropic joint. (Snoddy, 2017)
Can include exertional head and neck pain, nystagmus, cerebellar signs (ataxia, dysmetria, and dysdiadokokinesia) hydrocephalus, scoliosis, disc problems, pinched nerves, fasciculations, paraplegia, quadriplegia, chronic pain, general weakness of back/shoulders/extremities, and respiratory failure. (Wang, 2017)
Single fiber EMG studies have shown that the most severe abnormalities are located more distally. (Schwartz, 1980)
- Diagnostic Imaging (Schwartz, 1999; Hudson, 2008)
MRI
CT scans
Less reliable because of degradation of the spinal cord image due to the presence of surrounding bone.
CT is capable of exposing subarachnoid adhesions, but it lacks reliability in detecting syringes.
- Tests
The Queckenstedt test, Valsalva maneuver, and cough test have all been theorized to create a pressure differential within the subarachnoid spaces superior and inferior to the obstruction. These tests can increase CSF pressure enough to elicit the patient's symptoms (Heiss, 2012).
++
- Generally progressive
- Short-term and long-term outcomes depend completely on the size and location of the lesion.
- MRI diagnosis of syringomyelia in a timely manner with earlier surgical intervention to retard the disease's progression is associated with greater prognosis than delayed/incorrect imaging. (Hudson, 2008)
- If diagnosed and treated quickly the prognosis is fairly good.
Due to the pathophysiology of syringomyelia, much of the potential damage can be avoided mainly through surgical intervention. However, due to the progressive course of this disease some of the neurological deficits may return. (Konar, 2016; Ushewokunze, 2010)
- Syringomyelia is considered to be "incurable" due to the risk that it could come back.
- Recent evidence has shown that patients with a deviated syrinx, long syrinx, and a long duration of morbidity after foramen magnum decompression have poorer prognosis than those without these post-operative morphology (Nagoshi, 2014).
+++
IV. MEDICAL/SURGICAL MANAGEMENT
++
- Surgery is the only viable treatment for syringomyelia, which involves drainage or decompression of the syrinx cavity with diversion of fluid to the subarachnoid space or peritoneal cavity (Robinson, 1990; Hudson, 2008).
Surgical management through shunt tubing might not be a viable long-term solution for Type II syringomyelia. However, the short-term results of shunting the subarachnoid space and the syrinx cavity shows considerable promise. Yet, continued long-term follow-up seems to be necessary for consistent positive long-term outcomes. (Soo, 2014)
Shunting of CSF and decompression of Chari Malformation can alleviate the blockage in the spinal cord and halt disease progression, helping to reduce the effects of the syrinx on associated symptoms. Delaying surgical management may cause symptoms to worsen, with possible disability. (Hilton, 2003)
A spinal cordectomy, which is a transection of the damaged spinal cord, is considered the last treatment option for post-traumatic syringomyelia, since it is an irreversible procedure. Research has shown improvements in spasticity, pain levels, and ascending neurological deficits, as well as overall improved quality of life. (Konar, 2017)
An artificial conduit to reconstruct the CSF channels can be effective in the long-term in reducing symptoms in patients with post-traumatic syringomyelia. If performed in conjunction with other surgical techniques, it can stabilize the symptoms and provide even greater outcomes. (Ushewokunze, 2010)
- There is no known medical intervention for the disease itself, only treatment of associated symptoms. (Hudson, 2008)
+++
V. IMPLICATIONS FOR THERAPEUTIC MANAGEMENT
++
- Physical Therapy Neuromuscular Preferred Practice Pattern (APTA, 2017)
5H: Impaired Motor Function, Peripheral Nerve Integrity, and Sensory Integrity Associated with Nonprogressive Disorders of the Spinal Cord
5E: Impaired Motor Function and Sensory Integrity Associated with Progressive Disorders of the Central Nervous System
- Precautions to Therapeutic Management
- Physical Therapy (Vandertop, 2014; Zhang, 2016, Smith 2016)
Help with functional activities such as: bed mobility, transfers, gait abnormalities
Prevention and loosening of contractures
Treatment of spasticity
Education on assistive devices
Improving balance and coordination
Strengthening
Helping with functional ADLs
Assist with pain management.
Use of aquatic therapy can reduce pain and improve strength in this population. (Smith, 2016)
- Occupation Therapy
- Nursing
Bowel and bladder control
Postoperative care: neurological, respiratory, cardiac (Hilton, 2003)
- Speech/Language Pathologist
- Psychologist
+++
VI. CONSUMER AND PROFESSIONAL RESOURCES
++
- Mayo Clinic:
- American Syringomyelia & Chiari Alliance Project:
- MD Junction:
Website dedicated to providing alternative care options and support groups to individuals suffering from syringomyelia. Allows individuals to ask questions and receive feedback from medical doctors anonymously.
http://www.mdjunction.com/syringomyelia
- MD Guidelines:
- Chiari & Syringomyelia Foundation:
Website offers information on education, research and advocacy for patients, caregivers, family members and others on what syringomyelia is, its effects and how a person can help.
http://www.csfinfo.org
- The American Center for Spine & Neurosurgery:
- National Organization for Rare Diseases:
Website dedicated to many different rare diseases. It encompasses a report of syringomyelia conducted in part by a pediatric neurosurgeon It provides caregivers and health professionals more information on the disorder, clinical presentation, and how to treat it.
http://rarediseases.org/rare-diseases/syringomyelia
+
Brodbelt
AR, Stoodley
MA. Post-traumatic syringomyelia: a review.
J Clin Neurosci. 2003;10(4):401–408.
[PubMed: 12852875]
+
Heiss
J, Snyder
K, Peterson
M, Patronas
N, Butman
J, Smith
Rene, Devroom
Hetty, Sansur
Charles, Eskioglu
Eric, Kammerer
William, Oldfield
Edward. Pathophysiology of primary spinal syringomyelia.
J Neurosurg Spine. 2012;17(5):367–380.
[PubMed: 22958075]
+
Hilton
EL, Henderson
LJ. Neurosurgical considerations in posttraumatic syringomyelia. AORN J. 2003;77(1):136–150.
+
Hudson
BR, Cook
C, Goode
A. Identifying myelopathy caused by thoracic syringomyelia: a case report.
J Man Manip Ther. 2008;16(2):82–88.
[PubMed: 19119392]
+
Konar
SK, Maiti
TK, Bir
SC, Nanda
A. Spinal cordectomy: A new hope for morbid spinal conditions.
Clin Neurol Neurosurg. 2017;152:5–11.
[PubMed: 27846400]
+
Levine
DN. The pathogenesis of syringomyelia associated with lesions at the foramen magnum: a critical review of existing theories and proposal of a new hypothesis.
J Neurol Sci. 2004;220(1–2):3–21.
[PubMed: 15140600]
+
Michals
EA, Ramsey
RG. Radiology review: Syringomyelia.
Orthop Nurs. 1996;15(5):33–40.
[PubMed: 8954460]
+
Nagoshi
N, Iwanami
A, Toyama
Y, Nakamura
M. Factors contributing to improvement of syringomyelia after foramen magnum decompression for Chiari type I malformation.
J Orthop Sci. 2014;19(3):418–423.
[PubMed: 24633622]
+
Rai
SK, Rai
PK. Volume change theory for syringomyelia: A new perspective.
Asian J Neurosurg. 2015;10(4):245–51.
[PubMed: 26425150]
+
Robinson
LR. Little JW. Motor-evoked potentials reflect spinal cord function in post-traumatic syringomyelia.
Am J Phys Med Rehabil. 1990;69(6):307–10.
[PubMed: 2264950]
+
Robinson
LR. Bisnaire D. Syringomyelia and syringobulbia: pathophysiology, surgical treatment and nursing implications. J Neurosci Nursing. 1990;22(2):69–75.
+
Smith
R, Jones
G, Curtis
A, Murphy
H, Flint
G. Are Established Methods of Physiotherapeutic Management for Long-term Neurological Conditions Applicable to "Orphan" Conditions such as Syringomyelia?
Physiother Res Int. 2016;21(1):4–21.
[PubMed: 25475405]
+
Soo
TM, Sandquist
L, Tong
D, Barrett
R. Surgical treatment of idiopathic syringomyelia: Silastic wedge syringosubarachnoid shunting technique. Surg Neurol Int. 2014;5:114.
+
Schwartz
E, Falcone
S, Quencer
R, Green
B. Posttraumatic Syringomyelia: Pathogenesis, Imaging, and Treatment. ARJ Am J Roentgenol. 1999;173(2):487–492.
+
Schwartz
MS, Stålberg
E, Swash
M. Pattern of segmental motor involvement in syringomyelia: A single fibre EMG study.
J Neurol Neurosurg Psychiatry. 1980;43(2):150–155.
[PubMed: 7359152]
+
Snoddy
MC, Lee
DH, Kuhn
JE. Charcot shoulder and elbow: a review of the literature and update on treatment.
J Shoulder Elbow Surg. 2017;26(3):544–552.
[PubMed: 28111181]
+
Struck
AF, Carr
CM, Shah
V, Hesselink
JR, Haughton
VM. Cervical spinal canal narrowing in idiopathic syringomyelia.
Neuroradiology. 2016;58(8):771–775.
[PubMed: 27194170]
+
Wang
J, Alotaibi
NM, Samuel
N, Ibrahim
GM, Fallah
A, Cusimano
MD. Acquired Chiari Malformation and Syringomyelia Secondary to Space-Occupying Lesions: A Systematic Review.
World Neurosurg. 2017;98:800–808.e2.
[PubMed: 27894943]
+
Ushewokunze
SOS, Gan
YC, Phillips
K, Thacker
K, Flint
G. Surgical treatment of post-traumatic syringomyelia.
Spinal Cord. 2010;48(9):710–713.
[PubMed: 20309005]
+
Zhang
D, Melikian
R, Papavassiliou
E. Chiari I Malformation presenting as shoulder pain, weakness, and muscle atrophy in a collegiate athlete.
Curr Sports Med Rep. 2016;15(1):10–12.
[PubMed: 26745162]
++
Transverse myelitis (TM) is a neurological disorder caused by focal inflammation of the spinal cord (SC) (Kerr, 2002)
Transverse: involvement across one level of SC; Myelitis: inflammation of the SC
Myelin destruction: regional dysfunction of motor, sensory and autonomic pathways (Krishnan, 2004)
Can also affect white matter indiscriminately and destroys axons and cell bodies as well (Choi, 1996).
Transverse myelitis can be divided into 2 sub-groups (Scott, 2011; Jacob, 2008).
Incidence
It is a rare syndrome with an incidence of between 1 and 8 new cases per million per year (Krishnan, 2004).
No gender related or genetic predisposition.
It can develop at any age, however there is a bimodal peak between the ages of 10 to 19 years and 30 to 39 years with 28% of cases reported in children (Frohman, 2010; Krishnan, 2004; Beh, 2013)
Spinal cord involvement is usually central, uniform, and symmetric (Awad, 2011).
Demyelination, axonal loss, and gray matter involvement are characteristics of transverse myelitis (Awad, 2011).
The thoracic spinal cord is the most frequent site of involvement and the lesion typically spans multiple vertebral segments (Choi, 1996; Scott, 2011).
The main etiologies of transverse myelitis-like syndromes to rule out include multiple sclerosis, parainfectious myelitis, NMO, and systemic disease. However, after several years of follow up 15-36% of patients remain with the diagnosis of "Idiopathic TM" (Scott, 2011)
May be idiopathic or a result of: direct injury to the spinal cord, systemic or autoimmune disease, infectious disease, insufficient blood flow to the spinal cord, or as an isolated problem (Agmon-Levin, 2009).
Infectious agents associated with TM:
Viral: HSV-1, HSV-2, HIV, influenza, rabies, HAV, HBV, HCV (NINDS, 2007), Varicella zoster virus (VZV) (Lee, 2016), measles, rubella, mumps, Epstein-Barr virus (Agmon-Levin, 2009), Longitudinal Extensive Transverse Myelitis is associated with dengue fever.(Larik, 2012; DeSousa, 2014)
Bacterial: bacterial pneumonia, syphilis, TB, Lyme disease (Chan, 2006) rare--S. paratyphi B (Pourhassen,2009), mycoplasma (Agmon-Levin, 2009)
Vaccinations for: influenza, cholera, typhoid, poliomyelitis (Lahat, 1998), HBV (Kerr, 2002) and MR (Lim, 2004)
30% of pediatric cases are preceded with immunizations one month prior to disease onset (Awad, 2011).
Although rare, Guillain-Barre syndrome has been associated with acute transvers myelitis and could mask the signs of acute transvers myelitis and allow it to go untreated if the diseases are not tested for separately (Tolunay, 2016).
Systemic autoimmune and connective tissue diseases linked:
Multiple sclerosis, neuromyelitis optica (Devic's syndrome), systemic lupus erythematosus, Sjogren's syndrome, sarcoidosis, rheumatoid arthritis, dermatitis, vasculitis, antiphospholipid syndrome, and Behcet's disease caused Brown Sequard (Chan, 2006; Harzheim, 2004; Agmon-Levin, 2009; Brinar 2006),
TM (monofocal inflammation) may be the presenting feature of MS (multifocal disease) (Krishnan, 2004)
Vascular pathologies:
Thrombotic infarction of SC leading to inflammation and spinal arteriovenous malformations (Kerr, 2002; Campi, 1995; NINDS, 2007),
Transverse myelitis may be the first clinical manifestation of demyelinating disease. (Jacob, 2008; Beh, 2013; Sellner 2009)
Posterolateral spinal cord lesions were reported to be associated with increased risk of developing Multiple Sclerosis. (Sellner, 2009)
Patients with APTM may have a greater risk of developing MS than those presenting with ACTM. (Scott, 2011)
Neuromyelitis Optica Immune globulin-G (NMO-IgG) was found in 50% of those with longitudinally extensive transverse myelitis. (Jacob, 2008)
Others
Linked to radiation therapy, various invasive therapies (Ullrich, 2006), spinal surgery (Banit, 2003), and epidural anesthesia (Jha, 2006)
++
To help achieve accurate diagnosis, the patient's clinical history, examination, and MRI findings should all be utilized (Jacob, 2008).
Signs and Symptoms:
Dependent on lesion level-
Predominately at thoracic level
Upper cervical lesion can affect CN XI making it difficult to open the upper airway.
Lesions above C5 can weaken the diaphragm making it difficult to breathe.
Lesion above T6 can cause autonomic dysreflexia. (NINDS, 2007; Wolf, 2012)
Classic features (NINDS, 2007)
Paresthesia and dysesthesia at and below lesion level (Pidcock, 2007)
Weakness (especially of legs) below the lesion level; if severe can lead to paralysis; degree of paralysis depends on the severity of inflammation and the spinal cord level. (Calis, 2011)
Spasticity below the level of the lesion
Autonomic symptoms including bowel and bladder dysfunction (incontinence, or difficulty voiding and constipation)
PNS involvement: frequently acute; helps discriminate from MS (Harzheim, 2004)
Symptoms are bilateral (not necessarily symmetrical) (Krishnan, 2004)
Respiratory problems (if upper cervical cord is affected)
Neurological dysfunction, weakness, and flaccidity (Krishnan, 2004)
Most patients present with flu-like symptoms prior to the myelopathy (Awad, 2011).
Symptoms of Acute Transverse Myelitis
Sudden back pain, numbness, muscle weakness that starts from the feet and moves upward, loss of other sensations, and loss of bowel and bladder control (Awad, 2011).
The most frequent presenting symptoms
Sensory (100%), motor (47.6%), and autonomic (19%) disturbances. (n=63) (Calis, 2011; Sellner, 2009)
Among pediatric ATM in the initial phase of ATM (2-7days) 91% of patients described sensory loss predominately in the thoracic and cervical distribution, 89% reported weakness, 85% reported urinary dysfunction, and 75% reported pain. 89% of patients were bed/wheelchair bound or on assisted ventilation. (Pidcock, 2007)
Pain is the primary presenting symptom of transverse myelitis in approximately one-third to one-half of all patients.
The pain may be localized in the lower back or may consist of sharp, shooting sensations that radiate down the legs or arms or around the torso (NINDS, Jan 25 2012). 60% of children report pain as the most common initial symptom (Wolf, 2012).
Many patients also experience muscle spasms, a general feeling of discomfort, headache, fever, and loss of appetite.
Infantile acute transverse myelitis is associated with reduced movement, primarily affecting the lower extremities and urinary retention. (Wolf, 2012)
Symptoms can be acute (progressing over several hrs.) or progress initially over days or wks.
Medical testing
Imaging
Emergency MRI of spinal cord to exclude acute cord compression
Contrast enhanced MRI of cord and brain
Neuroimaging should exclude extra-axial compressive causes.
Transcranial magnetic stimulation (TMS) has also been used as a diagnostic tool to assess children who may have acute transverse myelitis (Voitenkov, 2015)
Lumbar puncture to rule-out the presence of infection in the serum and CSF (Goh, 2014)
Cerebrospinal fluid shows elevated protein levels and pleocytosis, elevated IgG index (Agmon-Levin, 2009; Awad, 2011).
Blood Tests
Differential blood cell counts, c-reactive proteins, rheumatoid factors, antinuclear antibodies (Harzheim, 2004)
Inflammatory markers such as interleukin-6 (IL-6) and other signaling proteins are elevated and correlate with disease severity (Agmon-Levin, 2009).
More definitive work needs to be done for specific CSF, MRI features, and pathological findings (Sellner, 2009)
Diagnostic criteria
For idiopathic transverse myelitis (Agmon-Levin, 2009):
"Bilateral sensory, motor, or autonomic dysfunction attributable to the spinal cord
A clearly defined sensory level
Peaking of symptoms within 4 hours and 21 days
The inclusion criteria for idiopathic transverse myelitis and disease-associated transverse myelitis includes (Transverse Myelitis Consortium Working Group, 2002):
Development of sensory, motor or autonomic dysfunction attributable to the spinal cord
Bilateral symptoms
Clearly defined sensory level
Exclusion of compressive etiology by MRI or CT myelography
Spinal cord inflammation demonstrated by CSF pleocytosis or elevated IgG or gadolinium enhancement
Progression to clinical nadir (maximal disability) between 4 hours and 21 days from onset of symptoms
The exclusion criteria for idiopathic transverse myelitis and disease-associated transverse myelitis includes:
History of radiation to the spine within 10 years
Clear arterial distribution clinical defect consistent with anterior spinal artery occlusion
Abnormal flow voids on the surface of the cord consistent with Arteriovenous malformation (AVM)
The exclusion criteria for idiopathic transverse myelitis includes:
Serologic or clinical evidence of connective tissue disease (sarcoidosis, Behcet's disease, Sjogren's syndrome, SLE, mixed connective tissue disorder)
CNS manifestations of syphilis, Lyme disease, HIV, HTLV-1, Mycoplasma, other viral infection.
Brain abnormalities suggestive of MS History of clinically apparent optic neuritis
Differential Diagnoses
TM is often misdiagnosed with other syndromes that present with rapidly progressive sensory and motor loss primarily affecting the lower extremities such as Gullain-Barre syndrome (Krishman,2004)
TM can be a presenting factor of MS and patients diagnosed with TM are at high risk for developing MS (Jacob, 2008, Krishnan, 2004).
++
Adults:
Usually non-progressive after initial development
Risk factors for relapse:
Poor prognosis is suggested by:
Rapid onset of severe symptoms (suggesting spinal shock) (de Seze, 2005)
Absence of central conduction and presence of protein 14-3-3 (marker of neuronal injury) in the CSF (Barnes, 2002)
Back pain, rapid progression, cervical spine involvement (Awad, 2011)
Denervation on EMG (Misra, 1998).
Progression
Symptoms stop progressing after 2-3 wks.
Recovery rapid during months 3-6 and may continue slowly for up to 2 yrs. (Krishnan, 2004)
If no improvement within the first 3 to 6 months, significant recovery is unlikely
1/3 have good recovery (no sequelae), 1/3 with moderate degree of permanent disability, 1/3 have no recovery (NINDS, 2007)
Children:
Predictors of poor outcome in children with acute idiopathic transverse myelitis (Deiva, 2015)
ASIA of A, B or C at onset
Spinal lesion with gadolinium enhancement
Female sex
CSF leukocytes <10
Absence of cervical or cervico-thoracic lesion
30% of children had a poor outcome associated with onset of maximal disability in less than 24 hours and sphincter dysfunction (Deiva, 2015)
Onset of ATM below age of three is associated with decreased functional outcomes. (Pidcock, 2007)
Predictors of good outcome:
Plateau of symptoms within 8 days of onset
Presence of supraspinal symptoms
Less than 1 month to independent walking
Increased deep tendon reflexes and Babinski reflex (Chen, 2013).
Patients who have had rehabilitation early in the recovery period have shown significant improvement in their recovery to perform activities of daily living (Calis, 2011).
Patients with idiopathic ATM who participated in a rehabilitation program had improved scores on the Ashworth Scale, Functional Independent Measure (FIM), Barthel Index, and Functional Ambulatory Scale after 4 weeks. (Calis, 2011)
Long-term outcome depends on remyelination, axonal regeneration, sprouting of collateral nerves, and neuronal plasticity (Kalita, 2001; Gupta, 2009).
Recovery can be limited secondary to immobility complications such as:
Relapse occurs in 20-30% of patients (Awad, 2011).
Risk of Multiple Sclerosis
1 in 10 individuals meeting diagnostic criteria may display an initial episode of MS (Bruna, 2006).
Acute partial transverse myelitis (APTM) is associated with increased risk of developing MS.
21% chance of progression to MS at 20 year follow up. (Fisniku, 2008)
If there are white matter lesions in the brain, this risk increases to 88% (Goh, 2014).
Longitudinally extensive transverse myelitis (LETM) has a very low conversion risk to MS (< 2%).
However, LETM is noted for a 60% risk of developing into neuromyelitis optica. (Chang, 2013; Goh, 2014)
Children with an initial diagnosis of APTM are 5-times more likely to have a subsequent diagnosis of MS.
If normal brain MRI results were present, 20% of children had subsequent diagnosis, but if abnormal brain MRI results were present, the subsequent diagnosis of MS increased to 66% (Meyer, 2014).
Confirmed independent risk factors of conversion of APTM to MS include abnormal brain MRI results with white matter lesions and presence of oligoclonal bands in the CSF. (Bourre, 2012).
Less than 10% of patients with idiopathic myelitis will develop Devic's syndrome (Scott, 2007).
Normal WBC count in CSF was associated with better mobility in children with Acute Transverse Myelitis. (Pidcock, 2007)
The majority of patients diagnosed with ATM reached independence in all skill areas (on the FIM) with the exception of sphincter control at mean 8 year follow up. (Pidcock, 2007)
Following non-traumatic SCI, although walking abilities may improve, gait speed often remains insufficient to safely negotiate community environments. (Beh, 2013)
+++
IV. Medical Management
++
Currently there is no effective cure for TM
Treatments are meant to reduce spinal cord inflammation and manage and alleviate symptoms (NINDS, Jan 25 2012).
Pharmacology
Corticosteroids (high dose intravenous methylprednisolone)
Used acutely work via immunosuppression to reduce disease activity, speed up recovery, and restore neurologic function (Krishnan, 2004; Awad, 2011; Scott, 2011; Kerr, 2002)
Immunoglobulin, plasmapheresis, cyclophosphamide, azathioprine
Used for autoimmune or infectious underlying disease (de Seze, 2005; Greenberg, 2007)
Plasma exchange (Plex) is often initiated if the patient exhibits little clinical improvement within 5-7 days of intravenous steroids in order to remove humoral factors inciting TM. (Krishnan, 2004; Beh, 2013)
Combination: Pulse methylprednisolone, intravenous immunoglobulin and plasmapharesis were used in the first reported case of longitudinal extensive transverse myelitis related to dengue infection in a pediatric patient. Using these immunomodulatory medications, the patient had a "near complete clinical recovery" from flaccid quadriplegia (Fong, 2016)
NSAIDs
Antidepressants (SSRIs, TCAs)
Anticholinergics (oxybutynin, hyoscyamine/ atropine, tolterodine) to treat spastic bladder (Wolf, 2012)
Rituximab may help decrease the number of relapses (Scott, 2011).
Muscle relaxants (Baclofen) treat spasticity (Krishnan, 2004)
Others:
Self-catheterization and indwelling catheters to treat urinary incontinence (Wolf, 2012)
CSF filtration is a new therapy in which spinal fluid is filtered for inflammatory factors prior to being reinfused into the patient (Krishnan, 2004).
Selective dorsal rhizotomy may be effective in treating spasticity (Mazarakis, 2015).
+++
V. Preview of Therapeutic management
++
Physical Therapy Preferred Practice Pattern (APTA, 2017)
5D: Impaired Motor Function and Sensory Integrity Associated With Nonprogressive Disorders of the Central Nervous System—Acquired in Adolescence or Adulthood
4F:Impaired Joint Mobility, Motor Function, Muscle Performance, Range of Motion, and Reflex Integrity Associated With Spinal Disorders
Transverse myelitis is a complex diagnosis that requires a multidisciplinary team in order to manage the treatment of the associated symptoms (Sadowsky, 2011)
Role of Physical Therapy
Address pain management, decreased ROM, weakness, spasticity, coordination, endurance,
Initial passive movement of limbs is important, followed by active range of motion exercises once the patient regains limb control to minimize spasticity (Sadowsky, 2011).
Begin PT early during the course of recovery for prevention of secondary complications due to immobility (e.g. contractures and skin breakdown) (Krishnan, 2004)
For the patient with transvers myelitis, pain had a greater impact than disability or depression on quality of life; thus treating pain may be more beneficial in some patients than treating the disability as related to improving quality of life of the patient (Kong, 2016)
Improve functional skills (mobility, transfers) (Grieve, 2006)
Gait assistance and retraining including assisted device training and prescription and fitting of orthotics to make up for body system declines (Sadowsky, 2011).
Functional electrical stimulation can help patients with weakness, spasticity, and gait assistance (Sadowsky, 2011).
Using the concepts of neural restoration as an essential treatment for TM could allow for greater functional gains and repair of the nervous system (Sadowsky, 2011).
Treatment for sensory deficits may include sensory stimulation to help activate afferent pathways and whole-body vibration (Sadowsky, 2011).
Improving mobility in an energy-efficient manner may improve health-related quality of life following TM. Exercise also imparts anti-inflammatory benefits. (Beh, 2013)
Education on appropriate assistive devices and pressure relief (Calis, 2011)
Challenges during therapy include but are not limited to:
Depression, Spasticity, bowel and bladder functions, weakness, and decreased range of motion (Krishnan, 2004)
+++
VI. Consumer and professional resources
++
Christopher and Dana Reeve Foundation: Paralysis Resource Center: Transverse Myelitis
National institute of Neurological Disorders and Stroke: Transverse Myelitis fact Sheet
Transverse Myelitis Association
The Mayo Clinic: Transverse Myelitis
Transverse Myelitis
Johns Hopkins Medicine: The Transverse Myelitis Center
Transverse Myelitis Online Community
+
Agmon-Levin
N, Kivity
S, Szyper-Kravitz
M, Shoenfeld
Y. Transverse myelitis and vaccines: a multi-analysis. Lupus. November 2009;18(13):1198–1204.
+
Awad
A, Stuve
O. Idiopathic Transverse Myelitis and Neuromyelitis Optica: Clinical Profiles, Pathophysiology and Therapeutic Choices. Current Neuropharmacology. September 2011;9(3):417–428
+
Banit
D, Wheeler
A, Anthony
H, Darden
B. Recurrent Transverse Myelitis After Lumbar Spine Surgery: A Case Report. Spine. 2003;28:165–168
+
Barnes
G, Benjamin
S, Bowen
JD, Cutter
N, De Lateur
BJ, Dietrich
WD, Dowling
MM, Griffin
JW, Hummers
L, Irani
D, Jorens
PG. Proposed diagnostic criteria and nosology of acute transverse myelitis. Neurology. 2002 Aug 27;59(4):499–505.
+
Beh
S, Greenberg
B, Frohman
T, Frohman
E. Transverse Myelitis. Neurologic Clinics. February 2013;31(1):79–138.
+
Bourre
B, Zéphir
H, Ongagna
J-C, Cordonnier
C, Collongues
N, Debette
S, Fleury
M-C, Outteryck
O, Hannequin
D, Vermersch
P. Long-term follow-up of acute partial transverse myelitis.
Arch Neurol[Archives of Neurology Full Text]. 2012;69(3):357–362.
[PubMed: 22410443]
+
Brinar
V, Habek
M, Brinar
M, Jalojcic
B, Boban
M. The Differential Diagnosis of Acute Transverse Myelitis.
Clinical Neurology and Neurosurgery. 2006;108:278–283.
[PubMed: 16376014]
+
Bruna
J, Martinez-Yelamos
S, Martinez-Yelamos
A, Rubio
F, Arbizu
T. Idiopathic Acute Transverse Myelitis: A Clinical Case Study and Prognostic Markers in 45 Cases.
Multiple Sclerosis. 2006;12:169–173.
[PubMed: 16629419]
+
Calis
M, Kirnap
M, Calis
H, Mistik
S, Demir
H. Rehabilitation results of patients with acute transverse myelitis.
Bratisl Lek Listy. 2011;112(3):154–156.
[PubMed: 21452769]
+
Campi
A, Filippi
M, Comi
G, Martinelli
V, Baratti
C, Rovaris
M, Scotti
G. Acute transverse myelopathy: spinal and cranial MR study with clinical follow-up. Am J Neuroradiol. 1995;16:115–123.
+
Chan
KH, Tsang
KL, Fong
GCY, Ho
SL, Cheung
RTF, Mak
W. Idiopathic inflammatory demyelinating disorders after acute transverse myelitis.
European Journal of Neurology. 2006;13:862–868.
[PubMed: 16879297]
+
Chen
L, Li
J, Guo
Z, Liao
S, Jiang
L. Prognostic Indicators of Acute Transverse Myelitis in 39 Children.
Pediatr Neurol. 2013;49(6):397–400.
[PubMed: 24112847]
+
Choi
KH, Lee
KS, Chung
SO,
et al. Idiopathic transverse myelitis: MR characteristics.
American Journal of Neuroradiology. 1996;17(6):1151–1160.
[PubMed: 8791931]
+
De Seze
J, Lanctin
C, Lebrun
C,
et al. Idiopathic acute transverse myelitis: Application of the recent diagnostic criteria.
Neurology. 2005;65:1950–1953.
[PubMed: 16380618]
+
Deiva
K, Absoud
M, Hemingway
C, Hernandez
Y, Husson
B, Maurey
H, Niotakis
G, Wassmer
E, Lim
M, Tardieu
M. Acute idiopathic transverse myelitis in children: Early predictors of relapse and disability.
Neurology. 2015;84(4):341–349.
[PubMed: 25540303]
+
De Sousa
AM, Alvarenga
MP, Alvarenga
RMP. A Cluster of Transverse Myelitis Following Dengue Virus Infection in the Brazilian Amazon Region.
Trop Med Health. 2014;42(3):115–120.
[PubMed: 25324689]
+
Fisniku
L, Brex
P, Altmann
D, Miszkiel
K, Benton
C, Lanyon
R, Thompson
A, Miller
D. Disability and T-2 MRI lesions: a 20-year follow-up of patients with relapse onset of multiple sclerosis.
Brain. 2008;131(Part 3):808–817. doi: 10.1093/brain/awm329. Epub 2008 Jan 29.
[PubMed: 18234696]
+
Fong
CY, Hlaing
CS, Tay
CG, Kadir
KAA, Goh
KJ, Ong
LC. Longitudinal extensive transverse myelitis with cervical epidural haematoma following dengue virus infection.
Eur J Paediatr Neurol. 2016;20(3):449–453.
[PubMed: 26900103]
+
Frohman
EM, Wingerchuk
DM. Transverse Myelitis.
New England Journal of Medicine. 2010;363(6):564–572.
[PubMed: 20818891]
+
Goh
C, Desmond
P, Phal
P. MRI in transverse myelitis. J Magn Reson Imaging. December 2014;40(6):1267–1279.
+
Greenberg
GM, Thomas
KP, Krishnan
C, Kaplin
AI, Calabresi
PA, Kerr
DA. Idiopathic transverse myelitis: Corticosteroids, plasma exchange, or cyclophosphamide.
Neurology. 2007;68:1614–1617.
[PubMed: 17485649]
+
Grieve
J, Hale
L. Gait rehabilitation in Acute Transverse Myelitis: a case report. New Zealand Journal of Physiotherapy. 2006;35(1):17–23.
+
Gupta
A, Taly
AB, Srivastava
A, Murali
T. Non-traumatic spinal cord lesions: epidemiology, complications, neurological and functional outcome of rehabilitation.
Spinal Cord .2009: 47(4): 307–11.
[PubMed: 18936767]
+
Hammerstedt
H, Edlow
J, Seric
C. Emergency Department Presentations of Transverse Myelitits: Two Case Reports.
Annals of Emergency Medicine. 2005; 46: 256–259.
[PubMed: 16126136]
+
Harzheim
M, Schlegel
U, Urbach
H, Klockgether
T, Schmidt
S. Discriminatory features of acute transverse myelitis: a retrospective analysis of 45 patients.
Journal of the Neurological Sciences. 2004;217:217–223.
[PubMed: 14706227]
+
Jacob
A, Weinshenker
B. An Approach to the Diagnosis of Acute Transverse Myelitis.
Seminars in Neurology. 2008;28(1):105–120.
[PubMed: 18256991]
+
Jha
S, Kumar
R. Transverse Myelitis Following Spinal Anesthesia.
Neurology India. 2006;54:425–427
[PubMed: 17114858]
+
Kalita
J, Misra
UK. Is methyl prednisolone useful in acute transverse myelitis?
Spinal Cord. 2001;39:471–476.
[PubMed: 11571658]
+
Kerr
D, Ayetey
H. Immunopathogenesis of Acute Transverse Myelitits.
Current Opinion in Neurology. 2002;15:339–347.
[PubMed: 12045735]
+
Kong
Y, Okoruwa
H, Revis
J,
et al. Pain in patients with transverse myelitis and its relationship to aquaporin 4 antibody status.
J Neurol Sci. 2016;368:84–88.
[PubMed: 27538606]
+
Krishnan
C, Kaplin
A, Deshpande
D, Pardo
C, Kerr
D. Transverse Myelitis: Pathogenesis, Diagnosis, and Treatment.
Frontiers in Bioscience. 2004;9:1483–1499.
[PubMed: 14977560]
+
Lahat
E, Pillar
G, Ravid
S, Barzilai
A, Etzioni
A, Shahar
E. Rapid Recovery From Transverse Myelopathy in Children Treated With Methylprednisolone.
Pediatr Neurol. 1998;19:279–282.
[PubMed: 9830998]
+
Larik
A, Chiong
Y, Lee
L, Ng
Y. Longitudinally extensive transverse myelitis associated with dengue fever. BMJ Case Rep. May 11, 2012;2012: bcr1220115378. doi: 10.1136/bcr.12.2011.5378.
+
Lee
JE, Lee
S, Kim
K-H,
et al. A Case of Transverse Myelitis Caused by Varicella Zoster Virus in an Immunocompetent Older Patient.
Infect Chemother. 2016;48(4):334–337.
[PubMed: 27883372]
+
Lim
S, Park
S, Choi
H,
et al. Transverse Myelitis after Measles and Rubella Vaccination.
J Paediatr Child Health. 2004; 40: 583–584.
[PubMed: 15367159]
+
Mazarakis
NK, Ughratdar
I, Vloeberghs
MH. Excellent functional outcome following selective dorsal rhizotomy in a child with spasticity secondary to transverse myelitis. Child's Nerv Syst. 2015;31(11):2189–2191.
+
Moon
S, Lee
J, Kim
T, Kim
S. Idiopathic transverse myelitis presenting as the Brown-Sequard syndrome.
Spinal Cord. 2009;47(2):176–8.
[PubMed: 18332885]
+
Merrell
C, McKinley
W. Infection-related spinal cord injury: etiologies and outcomes. Top Spinal Cord Inj Rehabil. 2008;14(2):31–41.
+
Meyer
P, Leboucq
N, Molinari
N, Roubertie
A, Carneiro
M, Walther-Louvier
U, Cuntz-Shadfar
D, Leydet
J, Cheminal
R, Cambonie
G, Echenne
B, Rondouin
G, Deiva
K, Mikaeloff
Y, Rivier
F. Partial acute transverse myelitis is a predictor of multiple sclerosis in children. Mult Scler. 2014:1352458514526943
+
Misra
U, Kalita
J. Can electromyography predict the prognosis of transverse myelitis. J Neurol. November 1998;245(11):741–744.
+
Pidcock
FS, Krishnan
C, Crawford
TO, Salorio
CF, Trovato,
M, Kerr
DA. Acute transverse myelitis in childhood: Center-based analysis of 47 cases.
Neurology. 2007;68:1474–1480.
[PubMed: 17470749]
+
Pourhassan
A. Acute transverse myelitis secondary to Salmonella paratyphi B infection.
Infection. 2008;36(2):170–3.
[PubMed: 17906841]
+
Perumal
J. Relevance of Aquaporin 4 Antibodies in Longitudinally Extensive Transverse Myelitis. Neurology Alert. October 2013;32(10):11–14.
+
Sadowsky
CL, Becker
DM, Bosques
GM, Dean
JM, McDonald
JW, Recio
A, Frohman
EM. Rehabilitation in Transverse Myelitis. Continuum Lifelong Learning Neurology. 2011;17:816–830.
+
Scott
T. Nosology of idiopathic transverse myelitis syndromes. Acta Neurologica Scandinavica. June 2007;115(6):371–376.
+
Scott
TF, Frohman
EM, De Seze
J, Gronseth
GS, Weinshenker
BG. Evidence-based guideline: Clinical evaluation and treatment of transverse myelitis.
Neurology. 2011;77(24):2128–2134.
[PubMed: 22156988]
+
Sellner
J, Luthi
N, Schupach
WMM, Gebhardt
A, Findling
O, Schroth
G, Mattle
HP, Nedeltchev
K. Diagnostic workup of patients with acute transverse myelitis: spectrum of clinical presentation, neuroimaging and laboratory findings.
SPINAL CORD, 2009: 47(4): 312–7
[PubMed: 19015666]
+
Tolunay
O, Çelik
T, Çelik
Ü, Kömür
M, Tanyeli
Z, Sönmezler
A. Concurrency of Guillain-Barre syndrome and acute transverse myelitis: a case report and review of literature. Korean J Pediatr. 2016;59(Suppl 1):161–164.
+
Transverse Myelitis Consortium Working Group. Proposed diagnostic criteria and nosology of acute transverse myelitis. Neurology. August 27, 2002;59(4):499–505.
+
Ullrich
NJ, Marcus
K, Pomeroy
SL,
et al.. Transverse Myelitis After Therapy For Primitive Neuroectodermal Tumors.
Pediatric Neurology. 2006;35:122–125
[PubMed: 16876009]
+
Vaghela
SA, Donnellan
CP. Acupuncture for back pain, knee pain and insomnia in transverse myelitis -- a case report. ACUPUNCTURE MED. 2008 Sep;26(3):188–92.
+
Voitenkov
VB, Klimkin
AV, Skripchenko
NV, Pulman
NF, Ivanova
MV. Diagnostic transcranial magnetic stimulation as a prognostic tool in children with acute transverse myelitis.
Spinal Cord. 2015;54(3):226–228.
[PubMed: 26238314]
+
Waldrup
BC. Identifying transverse myelitis in the emergency care arena.
ADV EMERG NURS J. 2009;31(2):131–9.
[PubMed: 20118863]
+
Wolf
V, Lupo
P, Lotze
T. Pediatric Acute Transverse Myelitis Overview and Differential Diagnosis.
J Child Neurol. 2012;27(11):1426–1436.
[PubMed: 22914370]
+++
Traumatic Brain Injury / Diffuse Axonal Injury
++
++
Signs and Symptoms
Depend on severity and location
General:
Diplopia, Headaches, Dizziness, Drainage of CSF from eyes, nose (rhinorrhea), and ears (otorrhea), indicates skull fracture – or bleeding from same locations, Fatigue, Photophobia, Phonophobia, Sleeping more or less than usual, Seizures, Tinnitus.
Related to Motor Function:
Impaired motor control (abnormal synergies) -Hemiplegia or bilateral asymmetrical distribution, Decreased flexibility, Paresis/hemiparesis, Paralysis, Incoordination, Weakness, Ataxia, Dysarthria, Impaired balance.
Related to Autonomic Dysfunction:
Respiratory irregularity, Abnormal pulse, Abnormal blood pressure, Excessive sweating, Dilated pupils
Related to Cognition/Mental Status:
Altered levels or loss of consciousness (lethargy, obtundation, stupor, delirium, coma), "persistent vegetative state" - >1 yr, memory problems, disorientation/confusion, inappropriate sexual behavior, increased impulsivity, deceased perseverance (Rebetez, 2015), mood changes/mood swings, apathy, increased risk taking.
Key Findings on Neurologic Exam
Cranial Nerve Exam: Unresponsive or asymmetric light reflex; Dysconjugate eye movements, Facial muscle paralysis, Loss of facial sensation, Impaired sense of smell
Reflexes: Abnormal reflex responses to noxious stimuli
Muscle Tone: Often spasticity in affected muscles; Decorticate or decerebrate rigidity (most severe form of spasticity)
Scales / Outcome Measures
Glasgow coma scale
Helps determine levels of consciousness and severity of injury
Scale ranges from 3 (deep coma/death) to 15 (full awake)
High inter-rater reliability and good for prognosis
Galveston Orientation and Amnesia Test (GOAT)
Rancho Los Amigos scale
Dynamic visual acuity testing and the Dizziness Handicap Index
According to Veteran's Administration Clinical Practice Guidelines, diagnosis includes onset or worsening of one of the following (Jeter, 2012)
Initial Glasgow Coma Scale of 13-15
Loss of memory for events lasting less than 24 hours.
Loss of consciousness (LOC) lasting under 30 minutes.
Alterations in mental status including confusion, disorientation, or slowed thinking.
Corticosteroid Randomization after Significant Head Injury (CRASH) trial and the International Mission on Prognosis and Analysis of Clinical Trials in TBI (IMPACT) databases create prediction with or without CT data, and have been cross-validated (Menon, 2009).
Disability Rating Scale (for TBI) can be used to track recovery from coma. Used mainly with moderate to severe TBI, and best suited for inpatient and outpatient rehabilitation.
High-level Mobility Assessment Tool (HiMAT)
Free 13-item tool that measures higher-level activities (running, jumping, etc.) not included in other mobility outcome measures. Minimal equipment is required for the HiMAT, and requires 5-15 minutes to complete (Ward, 2014).
Functional Assessment Measure (FIM-FAM)
30 item scale that combines the functional independence measure and functional assessment measure
FIM: 13 motor and 5 cognitive items
FAM: 12 items on cognitive, behavioral, communication and community function
Used to assess activity function, cognitive impairment and need for assistance (Kahn, 2016).
Cognitive Log (COGLOG)
10 item scale, each item scored from 0-3 for a possible total of 30
Used to assess cognitive function
Diagnostic Imaging to identify Cerebral lacerations, contusions, and hematomas
Computed tomography (CT) scans to identify hematomas
Magnetic resonance imaging (MRI) clearly identifies CNS lesions
Positron emission tomography (PET): Cerebral blood flow mapping tracks metabolism and energy use of the brain
Biomarker Tests
Biomarkers are indicators of biological disease/injury that can diagnose mTBI and predict symptoms (Jeter, 2012).
S-100B biomarkers in severe traumatic brain injury predict injury and outcome in adults. Pediatrics biomarkers need more study (Kovedski, 2009).
Serum levels of TBI-related biomarkers such as s100B in the acute period can discriminate favorable versus unfavorable outcomes and survival versus death in moderate to severe TBI, as explained in the next section (Di Battista, 2015)
++
General Information
Mild TBI usually improves from partial to near complete recovery over time.
Within the first few hours to days most patients with mild TBI improve, but post-concussive symptoms may continue for extended periods in 5-20% of people. (Mott, 2012)
Most rapid recovery in first 6 months, with lesser changes afterward.
Expected outcome and degree of deficit varies and depends on location/severity of injury and age.
Recovery varies among people who appear to have identical injuries.
After Severe TBI (Sinha, 2013)
Around 60% will still be unemployed 1 year post injury
Approximately half of individuals with severe TBI will have impairments in simple memory
The most impaired cognitive function is the ability to learn novel things.
~ 37% of individuals will have severe deficits in personality characteristics, while ~ 45% will have satisfactory restoration
40% will have emotional instability
Neurorehabilitation therapy that focuses on function significantly improves function in those with TBI and allows most of these individuals return home (Orient-Lopez, 2004)
Factors that influence recovery time
Length of coma and post -traumatic amnesia (PTA) (Increased length worsens prognosis and functional return)
Age (Worse with increasing age and more severe, persistent deficits)
Pre-injury status: Intellectual capacity/intelligence, Motivation, Personality, General physical and mental health, Family or significant other support
Factors significantly associated with unfavorable outcomes
Lower sociodemographic factors
Adults ≥ 65 years of age (Roe, 2013) more likely to suffer severe TBI due to falls with increased mortality rates
Male gender
Lower level of education
Clinical factors (Kim, 2011): Lower Glasgow Coma Scale Score, No pupil reaction, Increased biomarker (such as s100B) levels (Di Battista, 2015)
Predictors of disability
Initial severity of injury as measured on GCS
Duration of coma: Moderate disability/good recovery is indicated for those who are in a coma less than 1-week duration or had PTA lasting 4 weeks or less. (Katz, 1992)
Length of posttraumatic amnesia (Katz, 1994; Ono, 2001)
Factors affecting long-term survival (Brooks, 2013)
Factors that predispose to persistent symptoms
Women, Lower education level, Older adults, Previous mental health diagnosis (Mott, 2012)
Long-Term Deficits
+++
IV. Medical and Surgical
++
Acute stage/ICU
Maintain: Open airway, adequate oxygen and fluid levels, adequate cerebral perfusion pressure.
Avoid secondary brain insults (SBI)
Prevent intracranial hypertension (increased intracranial pressure) (Haddad, 2012) which will decrease brain perfusion
Brain tissue oxygen-directed critical care protocol (Narotam, 2009)
Surgery (if necessary)
Repairs, Removal of hematomas, Insertion of shunts, Insertion of ICP monitors
Surgery is necessary to: Relieve extremely high ICP, Repair bleeding vessels, Repair skull fractures and Remove blood clots
Common types of surgeries
Pharmaceuticals
Swelling (Diuretics)
Spasticity (Diazepam, Clorazepate, BoTox)
Aggression (Clonazepam, Lorazepam)
Depression (Antidepressants)
Sedation (Triazolam, Temazepam)
Memory Dysfunction (Donepezil)
Antihypertensive drugs
Anticonvulsants
Antibiotics for surgery (Grzankowski, 1997;Berkow, 1997)
Notes
Persons on antithrombotics prior to injury are at increased risk for intracranial bleed (McMillan, 2009).
Emerging area: anesthesiologist --role in age-dependent responses following pediatric traumatic brain injury (Huh, 2009).
+++
V. Implications for Therapeutic Management
++
Preferred Practice Pattern (APTA, 2017):
5C Impaired Motor function and Sensory Integrity Associated with Nonprogressive Disorders of the Central nervous System--Congenital Origin or Acquired in Infancy or Childhood.
5D Impaired Motor Function and Sensory Integrity Associated with Nonprogressive Disorders of the Central Nervous System---Acquired in Adolescence or Adulthood.
5I: Impaired Arousal, Range of Motion, and Motor Control Associated With Coma, Near Coma, or Vegetative State
Therapy: Focus on prevention (Fayol, 2009) and to help patients resume meaningful participation in their communities and social environments, regardless of whether specific impairments can be eliminated (Kahn, 2016).
Initial treatment:
PROM/stretching/flexibility to prevent contractures
Position changes to prevent pressure sores
Sensory stimulation
Ventilation and circulatory needs
Direct intervention:
Neuroprotective role of normobaric hyperoxia, an emerging treatment (Kumaria, 2009).
May require multiple episodes of care over time
Treatment of abnormal tone, primitive postures and spasticity
Early ROM to maintain joint flexibility and splinting to maintain muscle length.
Management should focus on therapeutic stretching and strengthening exercises as well as modalities and functional retraining. Positioning, such as maintaining neutral position of the head and neck, can help with increased tone and the effects of primitive posture. To decrease spasticity, PROM and strengthening of the antagonist muscles are important.
Prevention of secondary impairments
Treatment of impaired balance and motor function
Balance training should focus on the ability to balance safely while sitting, standing and walking. Gaze stability exercises, endurance training and general strengthening exercises can help.
Gait training to help with the impairments in coordination, timing and sequencing of movements.
Cognitive rehabilitation in post-acute TBI
Known to improve community functioning
Retrain functional skills or facilitate compensatory mechanisms that can aid in IADL's
Includes cognitive training for:
Executive function, verbal memory, working memory, attention, processing speed, non-verbal memory, visuospatial, language, IADL's and dysexecutive functions (Hallock, 2016).
Special concerns
Patients need direction: break tasks down into small, simple steps
Safety and cognitive issues especially in the early stages.
Initially avoid overstimulating environments
Make sure there is adequate lighting but be aware patients might be light sensitive and require a slightly dim environment
Health care team members include
Neurosurgeon, anesthesiologist, physiatrist, nurse, respiratory therapist, neuropsychologist, speech/language pathologist, occupational therapist, recreational therapist, case manager, physical therapist, and pharmacist.
Interdisciplinary team includes the patient, family, medical social worker, and all the above in the health care team.
Health promotion and prevention measures
Education should be provided for the patient's family so they know what to expect as well as reassurance to the patient about the typical time course for resolution or improvement of symptoms. (Weightman, 2010)
Early education related to diagnosis, prognosis and symptoms could help create realistic expectations and reduce anxiety. (Mott, 2012)
Patient education on how the patient can compensate for impairments as well as the possibility of support groups. This should also include education on wellness and prevention.
+++
VI. Consumer and Professional Resources
++
Information
National Institute of Neurological Disorders and Stroke
National Disability Rights Network
Family Caregiver Alliance
Directory of Independent Living Centers
Brainline
National Rehabilitation Information Center
Information in Spanish
Support Groups
Organizations
Brain Injury Association of America
North American Brain Injury Society
United States Brain Injury Alliance
Traumatic Brain Injury: The Journey Home
Brain Trauma Foundation
+
Abdul-Muneer
PM, Chandra
N, Haorah
J. Interactions of Oxidative Stress and Neurovascular Inflammation in the Pathogenesis of Traumatic Brain Injury. Mol Neurobiol. 2015 Jun 1;51(3):966–79.
+
Bains
M, Hall
ED. Antioxidant therapies in traumatic brain and spinal cord injury. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease. 2012;1822(5):675–684.
+
Bay
E. Current treatment options for depression after mild traumatic brain injury. Curr Treat Options Neurol. 2009 Sep;11(5):377–82.
+
Berker
E. Diagnosis, Physiology, Pathology, and Rehabilitation of Traumatic Brain Injuries. Int J Neurosci. 1996 Jan 1;85(3-4):195–220.
+
Berkow
R, Fletcher
A. Merck Manual of Diagnosis and Therapy. Sixteenth Edition. Rahway: Merck Research Laboratories; 1992: 1460–1464.
+
Bonds
B, Hu
P, Li
Y, Yang
S, Colton
K, Gonchigar
A, Cheriyan
J, Grissom
T, Fang
R, Stein
D. Predictive value of hyperthermia and intracranial hypertension on neurological outcomes in patients with severe traumatic brain injury. Brain Injury. December 2015;29(13/14):1642–1647.
+
Bor-Seng-Shu
E, Figueiredo
EG, Amorim
RLO,
et al. Decompressivecraniectomy: a meta-analysis of influences on intracranial pressure and cerebral perfusion pressure in the treatment of traumatic brain injury.
J Neurosurg. 2012;117(3):589–596.
[PubMed: 22794321]
+
Bouma
G J, Muizelaar
J P, Choi
S C, Newlon
P G, Young
H F. Cerebral circulation and metabolism after severe traumatic brain injury: the elusive role of ischemia.
J Neurosurg. 1991;75:685–693.
[PubMed: 1919689]
+
Brooks
JC, Strauss
DJ, Shavelle
RM, Paculdo
DR, Hammond
FM, Harrison-Felix
CL. Long-term disability and survival in traumatic brain injury: results from the National Institute on Disability and Rehabilitation Research Model Systems.
Arch Phys Med Rehabil. 2013;94(11):2203–2209.
[PubMed: 23872079]
+
Burke
WH. Head Injury Rehabilitation: An Overview. Houston, TX; HDI Publishers. 1998.
+
Carney
N, Chesnut
R, Maynard
H, Mann
C, Patterson
P, Helfand
M. Effect of Cognitive Rehabilitation on Outcomes for Persons with Traumatic Brain Injury: A Systematic Review. J Head Trauma Rehabil. 1999 Jun 1;14(3):277–307.
+
Coelho
C, DeRuyter
F, Stein
M. Treatment efficacy: cognitive-communicative disorders resulting from traumatic brain injury in adults. J Speech Hear Res. 1996 Oct;39:S5–S17.
+
Classen
S, Levy
C, McCarthy
D, Mann
WC, Lanford
D, Waid-Ebbs
JK. Traumatic brain injury and driving assessment: an evidence-based literature review.
Am J Occup Ther. 2009;63(5):580–91.
[PubMed: 19785257]
+
Di Battista
A, Buonora
J, Rhind
S, Hutchinson
M, Baker
A, Rizoli
S, Diaz-Arrastia
R, Mueller
G. Blood biomarkers in moderate-to-severe traumatic brain injury: potential utility of a multi-marker approach in characterizing outcome.
Frontiers In Neurology. May 2015;6:1–9.
[PubMed: 25699006]
+
Elliott
M, Parente
F. Efficacy of memory rehabilitation therapy: a meta-analysis of TBI and stroke cognitive rehabilitation literature.
Brain Inj. 2014;28(12):1610–1616.
[PubMed: 25058353]
+
Fann
JR, Hart
T, Schomer
KG. Treatment for Depression Following Traumatic Brain Injury: A Systematic Review. J Neurotrauma. 2009 Dec 1;26(12):2383–402.
+
Fayol
P, Carrière,
H, Habonimana
D, Dumond
JJ. Preliminary questions before studying mild traumatic brain injury outcome.
Ann Phys Rehabil Med. 2009;52(6):497–509.
[PubMed: 19760806]
+
Gottshall
K, Drake
A, Gray
N, McDonald
E, Hoffer
ME. Objective vestibular tests as outcome measures in head injury patients.
Laryngoscope. 2003;113(10):1746–1750.
[PubMed: 14520100]
+
Grzankowski
J. Altered Thought Processes Related to Traumatic Brain Injury and Their Nursing Implications.
Rehabil Nurs. 1997; 22: 24–31.
[PubMed: 9110840]
+
Haddad
SH, Arabi
YM. Critical care management of severe traumatic brain injury in adults. Scand J Trauma ResuscEmerg Med. 2012;20:12.
+
Hallock
H, Collins
D, Lampit
A, Deol
K, Fleming
J, Valenzuela
M. Cognitive Training for Post-Acute Traumatic Brain Injury: A Systematic Review and Meta-Analysis.
Front Hum Neurosci. 2016;10:537.
[PubMed: 27833541]
+
Huh
JW, Raghupathi
R. New concepts in treatment of pediatric traumatic brain injury.
Anesthesiol Clin. 2009; 27(2):213–40.
[PubMed: 19703674]
+
Jang
SH. Review of motor recovery in patients with traumatic brain injury.
NeuroRehabilitation. 2009;24(4):349–53.
[PubMed: 19597273]
+
Jeter
CB, Hergenroeder
GW, Hylin
MJ, Redell
JB, Moore
AN, Dash
PK. Biomarkers for the diagnosis and prognosis of mild traumatic brain injury/concussion. J Neurotrauma. 2012; Oct 12. doi:10.1089/neu.2012.2439
+
Kahn
F, Amatya
B, Judson
R, Chung
P, Truesdale
M, Elmalik
A, Galea
MP. Factors Associated with Long-Term Functional and Psychological Outcomes in Persons with Moderate to Severe Traumatic Brain Injury.
J Rehabil Med. 2016; 48(5): 442–448
[PubMed: 27058885]
+
Katz
DI. Neuropathology and neurobehavioral recovery from closed head injury. J Head Trauma Rehabil. 1992;7(2):1–15.
+
Kim
Y-J. A systematic review of factors contributing to outcomes in patients with traumatic brain injury.
J Clin Nurs. 2011;20(11/12):1518–1532.
[PubMed: 21453293]
+
Kokshoorn
N, Wassenaar
M, Biermasz
N, Roelfsema
F, Smit
J, Romijn
J, Pereira
A. Hypopituitarism following traumatic brain injury: the prevalence is affectedby the use of different dynamic tests and different normal values. Eur J Endocrinol. 2009.
+
Kövesdi
E, Lückl
J, Bukovics
P, Farkas
O, Pál
J, Czeiter
E, Szellár
D, Dóczi
T, Komoly
S, Büki
A. Update on protein biomarkers in traumatic brain injury with emphasis on clinical use in adults and pediatrics. ActaNeurochir (Wien). 2010;152(1):1–17.
+
Kumaria
A, Tolias
CM. Normobarichyperoxia therapy for traumatic brain injury and stroke: a review. Br J Neurosurg. 2009;(22):1–9.
+
Levin
HS, O'Donnell
VM, Grossman
RG. The Galveston Orientation and Amnesia Test. A practical scale to assess cognition after head injury.
J. Nerv. Ment. Dis. 1979;167(11):675–684.
[PubMed: 501342]
+
Lund
SB, Gjeilo
KH, Moen
KG, Mikalsen
K, Skandsen
T, Vik
A. Moderate Traumatic Brain Injury, Acute phase Course and Deviations in Physiological Variables: An Observational Study. Scand J Trauma Resusc Emerg Med. 2016;24:772–778
+
Lynch
W. Computer-Assisted Cognitive Retraining. J Head Trauma Rehabil.1986; 6: 77–78.
+
Mena
J, Sanchez
A, Rubiano
A, Peitzman
A, Sperry
J, Gutierrez
I, Puyana
J Effect of the modified Glasgow Coma Scale score criteria for mild traumatic brain injury on mortality prediction: comparing classic and modified Glasgow Coma Scale score model scores of 13.
Journal of Trauma. 2011;71(5):1185–1193.
[PubMed: 22071923]
+
Menon
DK, Zahed
C. Prediction of outcome in severe traumatic brain injury.
Curr Opin Crit Care. 2009.15(5):437–41.
[PubMed: 19713837]
+
McMillian
WD, Rogers
FB. Management of prehospital antiplatelet and anticoagulant therapy in traumatic head injury: a review. J Trauma, 2009. Vol. 66 (3), pp. 942–50.
+
Mott
TF, McConnon
ML, Rieger
BP. Subacute to chronic mild traumatic brain injury.
American Family Physician. 2012;86(11):1045–1051.
[PubMed: 23198672]
+
Myburgh
JA, Cooper
DJ, Finfer
SR,
et al. Epidemiology and 12-month outcomes from traumatic brain injury in australia and new zealand.
J Trauma. 2008;64(4):854–862.
[PubMed: 18404048]
+
Narotam
PK, Morrison
JF, Nathoo
N. Brain tissue oxygen monitoring in traumatic brain injury and major trauma: outcome analysis of a brain tissue oxygen-directed therapy.
J Neurosurg. 2009;111(4):672–682.
[PubMed: 19463048]
+
Ono
J, Yamaura
A, Kubota
M, Okimura
Y, Isobe
K. Outcome prediction in severe head injury: analyses of clinical prognostic factors.
Journal of Clinical Neuroscience. 2001;8(2):120–123.
[PubMed: 11484659]
+
Orient-López
F, Sevilla-Hernández
E, Guevara-Espinosa
D, Terré-Boliart
R, Ramón-Rona
S, Bernabeu-Guitart
M. [Functional outcome at discharge of patients with severe traumatic brain injury admitted to a brain damage unit].
Rev Neurol. 2004;39(10):901–906.
[PubMed: 15573302]
+
Rebetez
M, Rochat
L, Ghisletta
P, Walder
B, Van der Linden
M. Association between impulsivity, emotional/behavioural hyperactivation and functional outcome one year after severe traumatic brain injury. Brain Injury. September 2015;29(10):1175–1181.
+
Roe
C, Skandsen
T, Anke
A,
et al. Severe traumatic brain injury in Norway: impact of age on outcome.
J Rehabil Med. 2013;45(8):734–740. doi:10.2340/16501977-1198.
[PubMed: 24002308]
+
Sinha
S, Gunawat
P, Nehra
A, Sharma
BS. Cognitive, functional, and psychosocial outcome after severe traumatic brain injury: a cross-sectional study at a tertiary care trauma center.
Neurol India. 2013;61(5):501–506. doi:10.4103/0028-3886.121920.
[PubMed: 24262453]
+
Vaishnavi
S, Rao
V, Fann
JR. Neuropsychiatric problems after traumatic brain injury: unraveling the silent epidemic. Psychosomatics. 2009. May-Jun;50(3):198–205.
+
Vogenthaler
D. An Overview of Head Injury; Its Consequences and Rehabilitation. Brain Injury. Vol 1,1. 1987: 113–127.
+
Ward
I, Roth
HR, Kahn
JH, Tseng
E, Tappan
R. Measurement characteristics and clinical utility of the high-level mobility assessment tool among individuals with traumatic brain injury.
Arch Phys Med Rehabil. 2014;95(11):2229–2230.
[PubMed: 25587598]
+
Walker
PA, Harting
MT, Baumgartner
JE, Fletcher
S, Strobel
N, Cox
CS. Modern approaches to pediatric brain injury therapy. J Trauma. 2009 Aug;67(2Suppl):S120–7.
+
Weightman
MM, Bolgla
R, McCulloch
KL, Peterson
MD. Physical Therapy Recommendations for Service Members With Mild Traumatic Brain Injury.
J Head Trauma Rehabil. 2010;25(3):206–218.
[PubMed: 20473094]
+
White
J, Rosenthal
M. Rehabilitation of the Patient With Traumatic Brain Injury. In: DeLisa
JA. Rehabilitation Medicine: Principles and Practice. Second Edition. Philadelphia, PA: JB Lippincott Co; 1993: 825–853.
+++
Traumatic Spinal Cord Injury
++
SCI is a traumatic event that results in disturbances to normal sensory, motor, or autonomic function and ultimately impacts a patient's physical, psychological, and social well-being. (Singh, 2014)
The majority of SCI's result from trauma to the vertebral column. This trauma typically injures both vessels and neurons at the level of the injury (Sekhon, 2001).
Spinal cord injury can cause both temporary and permanent changes in strength, sensation, and body organ functions below the affected spinal level. (Mayo Clinic, 2014)
Primary injury to these structures results in fractured and displaced bone fragments, bleeding and swelling which compresses the spinal canal damaging blood vessels, axons and neural cell membranes of the spinal central and peripheral nervous system. This mechanical trauma initiates a cascade of events that rapidly enlarge the cross-sectional area from inflammation and swelling, and ultimately create a fluid filled cyst. Compression can lead to local spinal cord ischemia and irreversible damage. (McDonald, 2004)
Most injured cells die within 12 hours, affecting the center of the cord first.
Unless the cord is completely severed, the damaged area may retain a donut-like rim of surviving tissues.
The most susceptible areas to traumatic injuries are the lower cervical (C4-C6) vertebrae and the area of the thoracolumbar junction (T12-L1). (Sarhan, 2012) Thoracic injuries only make up 3% of traumatic SCI due to the fact that the thoracic spine is supported by the stiffness of the rib cage and ligamentous structures surrounding it.
Secondary injury also occurs from expanding hypoperfusion throughout the spinal cord. Hypoperfusion leads to spinal shock as it disturbs action potentials along axons. Excitotoxicity also occurs as result of secondary injury and this process produces free radicals that can kill healthy neurons. (McDonald, 2002)
Distribution of dysfunction is dependent on extent of cord damaged. This damage will result in partial or complete loss of sensation and motor function at and below the area of insult. (Singh, 2014)
The risk factors for TSCI include being young, male, engaging in risky behavior, such as diving into shallow water or driving under the influence. A pre-existing bone or joint disorder, such as osteoporosis, cervical spondylosis, atlantoaxial instability, ankylosing spondylitis or rheumatoid arthritis, also make some patients more susceptible to TSCI (Sekhon, 2001).
++
++
Extreme pain or pressure in the area of injury, numbness or anesthesia of the extremities in related regions or intense stinging sensation, weakness, incoordination, and paralysis of related region of the body
++
Loss of bladder or bowel control, impaired breathing, oddly positioned neck or back, spasticity in affected muscle groups with exaggerated tendon reflexes, muscle spasms, loss of sensation, altered consciousness, trouble with balance or ambulation, and flaccidity in muscles innervated from within the region of injury
Clinical signs include:
Associated back pain, midline tenderness, palpable midline step, and abnormal neurological signs. The patient may present with distension of the bladder and evidence of shock. The patient may also present with widening of paraspinal lines, mediastinal widening, and loss of vertebral body height.
+++
C. Diagnostic Testing
++
Asking questions about the accident and performing a full neurologic exam can help to determine the level and completeness of the SCI (Sarhan, 2012), particularly using the American Spinal Injury Association (ASIA) system and forms (also known as International Standards for Neurological Classification)
In diagnosing the level of injury there are a few rules to follow. In the upper thoracic spine, sensory loss will be one segment below the injured vertebral body, in the middle third, sensory loss will be two segments below the injured vertebral body, and in the lower third, sensory loss will be approximately three segments below the injured vertebral body.
Identifying the lesion level:
Locate the most distal uninvolved nerve root segment in the spinal cord with normal motor and sensory function bilaterally to determine neurological level.
Definition of normal function: motor-manual muscle test of at least 3/5, sensory- 2 on the 3 point ordinal scale
ASIA Impairment Scale (AIS): developed so healthcare professionals could better communicate the degree of impairment in individuals with spinal cord injuries. (ASIA, 2013)
A= Complete: No motor or sensory function is preserved in the sacral segments S4 to S5.
B= Sensory Incomplete: Sensory but not motor function is preserved below the neurological level and includes the sacral segments S4 to S5.
C= Motor Incomplete: Motor function is preserved below the neurological level, and more than half of key muscles below the neurological level have a muscle grade less than 3.
D= Motor Incomplete: Motor function is preserved below the neurological level, and at least half of key muscles below the neurological level have a muscle grade of ≥3.
E= Normal: Motor and sensory function has returned to normal.
Key Motor Levels: (ASIA, 2008a)
Key Sensory Levels: (ASIA, 2008b)
Categorization of Spinal Cord Injury Level:
Tetraplegia/Quadriplegia: injury to upper cord spinal level (C1 through T1) results in complete paralysis of the trunk, all four extremities (at least some involvement in the hands) and the respiratory muscles
Paraplegia: injury below spinal level T1 (thoracic cord, lumbar cord, sacral cord, or cauda equina) results in paralysis of at least part of lower extremities and trunk
Description of extent of lesion:
Complete lesions are characterized by complete loss of all sensory or motor function below the level of the lesion, and result from complete severing of the cord, severe compression of the cord or extensive vascular injury to the cord.
Incomplete lesions occur when some sensory or motor function below the level of injury is preserved or spared, and result from contusions produced by pressure on the cord or swelling from within the spinal canal. They may also occur when there is a partial severing of the cord, including hemisection of one side of the cord.
Imaging:
++
The chance of full recovery in patients who have a complete spinal cord injury is less than 5%. This percentage drops to nearly zero if complete paralysis is still present 72 hours after the injury. (Chin, 2012)
Within the first 72 hours post insult, the clinical neurological assessment should be used as described by the International Standards for Neurological Classification of SCI (ASIA) in order to determine a preliminary prognosis for neurological recovery; MRI findings may also be used to help determine prognosis. (Consortium, 2008).
"The most important predictor of improved outcome is retention of sacral sensation (S4-S5), especially pinprick, 72 h to 1 week after injury," i.e., incomplete SCI. Most patients with SCI's experience improvements in motor function and the most significant improvement normally takes place in the first 6 months. (McDonald, 2002)
The chances that a person will be able to walk again is greater than 50% if some sensory function is preserved. (Chin, 2012)
Factors that significantly predict resilience in patients with SCI at discharge from an inpatient facility include self-efficacy, low levels of negative mood and lower functional independence. (Guest, 2015)
Younger patients have greater prognosis for ambulation than those >50-60 years old with similar injury severity. (Whiteneck, 2000)
Some patients who initially have complete injury may progress to incomplete lesions following resolution of the edema and spinal cord contusion. (Sarhan, 2012)
80%-90% of patients will maintain complete injury, while 50% of patients with incomplete sensory injury may become ambulatory. Patients with motor incomplete injury have 75-95% chance of ambulation depending on level of injury and intact sensory function. (Whiteneck, 2000)
Average length of stay in the hospital acute care unit for a patient who has experienced a spinal cord injury is 11 days, and the number of days that the individual ends up staying in a rehabilitation facility following discharge from the hospital is around 36 days. (NSCISC, 2015)
Less than 1% of persons experienced complete neurologic recovery by hospital discharge following initial hospitalization. (NSCISC, 2015)
Life expectancies for individuals with SCI are significantly below average vs. those without SCI, and mortality rates are significantly higher during the first year after injury. (NSCISC, 2015).
Normally no effect on women's ability to engage in sexual intercourse, and complete pregnancy, labor or have a normal delivery. (Vodusek, 2014)
Secondary complications:
Respiratory complications associated with SCI are the most important cause of morbidity and mortality in both acute and chronic stages. These complications include insufficiency of respiratory muscles, reduction in vital capacity, ineffective cough, reduction in lung and chest wall compliance, and excess oxygen cost of breathing. (Sezer, 2015)
Decreased bladder control can lead to urinary tract infections, decreased bowel control can contribute to constipation, circulation changes can contribute to orthostatic hypotension, blood clot formation, deep vein thrombosis and pulmonary embolism, and chronic kidney disease. (Vodusek, 2014)
Neurogenic bowel (NB) is a major problem for individuals with traumatic SCI, affecting nearly half of those with SCI (46.9%). NB occurs when there is a dysfunction of the colon due to lack of nervous system control. (Sezer, 2015)
Pressure sores can develop within hours in immobilized patients. These are most common on the buttocks and heels. (Jia, 2013)
About 30% of persons with SCI experience one or more hospitalizations during a 12-month period, with each stay averaging 22 days. Diseases of the genitourinary system are the leading cause of rehospitalization, followed by diseases of the skin. (NSCISC, 2015)
At least one organ fails in 75% of individuals with SCI and multiple organ failure in 55% by Multiple Organ Dysfunction Scale. (Stein, 2010)
Benign paroxysmal positional vertigo is 2.87 times more likely to affect individuals suffering from cervical spinal cord injuries when compared to those with lumbar or thoracic spinal cord injuries. (Lee, 2012)
Risk of mortality following SCI is increased in individuals with a history of pneumonia or kidney calculus, poor general health, a decline in health over the past year or a grade-3 or -4 pressure ulcer. (Cao, 2013)
Patients with tetraplegia had a higher prevalence of orthostatic hypotension and a greater fall in blood pressure than patients with paraplegia irrespective of whether the lesion is complete or incomplete. (Illman, 2000) The persistent of orthostatic hypotension during the first month was prevalent in 74% of cervical and only 20% of upper thoracic motor complete spinal cord injury patients. (Sidorov, 2007)
Emotional adjustment following a traumatic SCI can be extremely difficult for a patient. Depression, drug addiction, and divorce are common following a traumatic SCI. It is estimated that 20% to 45% of individuals who have suffered a traumatic SCI will experience some type of depressive disorder within the first month following their injury. (Kirshblum, 2007)
The suicide rate is 2 to 6 times greater in individuals who have suffered a traumatic spinal cord injury than in individuals who have not suffered from a traumatic spinal cord injury. (Kirshblum, 2007)
Only 12% of individuals with spinal cord injury are employed one year after initial injury, and by 20 years post injury, only one third are employed. (NSCISC, 2015)
++
Spinal Cord injuries are considered medical emergencies that require immediate attention in order to reduce long-term effects. The amount of time between the injury and treatment will affect the ultimate prognosis.
It has been shown in studies that shorter surgical delay following traumatic SCI has resulted in reduction of complications and optimal neurological recovery during the acute care hospital stay. The study suggests that surgery should occur before or up to 24 hours post injury. If there are reasons to delay beyond 24 hours, the study recommends surgery before 72 hours. (Bourassa-moreau, 2013).
Conservative management should be preferred over acute surgical management in cases of Brown-Séquard syndrome. (Amendola, 2014)
In the emergency room, medical treatment is focused on maintaining ability to breathe, preventing shock, immobilization of the neck to prevent further spinal cord injury, and avoiding possible complications (blood clots, cardiovascular issues, etc.). (Mayo Clinic, 2014).
Pharmacological Interventions
High dose methylprednisolone (anti-inflammatory) has been shown to improve motor function at 1 year if given within the first 8 hours following injury. (McDonald, 2002) It is thought that methylprednisolone "reduces swelling, inflammation, glutamate release and free-radical accumulation." However, methylprednisolone may be associated with higher incidence of gastric bleeding and wound infection. (McDonald, 2002)
Focal microinjection of 2,3-dihydro-6-nitro-7sulfamolybenzo(f)quinoxaline (NBQX) has also been shown to reduce tissue loss and neurological deficits after spinal cord injury (Rosenberg, 1999)
Osmotic diuretics are also used to battle the effects of edema.
Proteolytic enzymes are used to reduce inflammation and scar tissue formation, to speed the process of recovery.
Other appropriate medications include: Acetaminophen, minor tranquilizers or muscle relaxants, narcotics, nonsteroidal anti-inflammatory drugs, oral steroids, psychotropic/Anti-anxiety drugs and tramadol
Surgery may be required for:
Relief from fluid/pressure that presses on the spinal cord
Removal of bone fragments, disc fragments, and foreign objects
Stabilization of fractured vertebrae by fusing bones or inserting hardware
Spinal traction may be used to reduce dislocation and/or to immobilize the spine. The skull may also need to be immobilized through the use of tongs.
Experimental treatments are being performed to develop more efficient ways to control inflammation, stop cell death and promote nerve regeneration. (Mayo Clinic, 2014)
+++
V. Implications for Therapeutic Management
++
Preferred Practice Pattern from Guide to Physical Therapy Practice (APTA, 2017):
Other Practice Patterns (APTA, 2017):
Pattern 4F: Impaired Joint Mobility, Motor Function, Muscle Performance, Range of Motion, and Reflex Integrity Associated with Spinal Disorders
Pattern 4G: Impaired Joint Mobility, Muscle Performance, and Range of Motion Associated with Fracture
Pattern 6B: Impaired Aerobic Capacity/Endurance Associated With Deconditioning 4. Pattern 6F: Impaired Ventilation and Respiration/Gas Exchange Associated With Respiratory Failure
Pattern 7A: Primary Prevention/Risk Reduction for Integumentary Disorders
++
A. Role of Physical Therapy
++
Prevention of complications. Treatment for respiratory issues. Management of contractures, spasticity, skeletal deformities, postural abnormalities and skin integrity. Functional training in mobility, vocational and recreation activity and use of assistive and orthotic devices. Bladder and bowel program. Interventions including toileting, seating, positioning and transferring. Counseling to address psychosocial issues and possible suicidal risk. (McDonald, 2002)
Active Therapy: Requires internal effort by the individual and supervision from a therapist; helps to restore flexibility, strength, endurance, function and range of motion.
Training in compensatory mechanisms for functional ability when deficits are permanent.
Activities of daily living-designed to improve a person's ability to perform daily activities such as self-care, housekeeping and driving.
Functional activities-designed to improve mobility, body mechanics, employability, coordination and sensory motor integration.
Neuromuscular re-education-the use of manual, mechanical or electrical facilitation to apply exercises designed to improve strength, posture, balance, coordination, kinesthetic sense, proprioception, neuromuscular response and movement patterns.
Functional electrical stimulation-used in patients with muscle atrophy, weakness and poor muscle contraction; utilizes electrical current to produce involuntary or assisted contractions.
Cervical Lumbar stabilization-designed to strengthen the spine in its neural and anatomic position; gives the patient the ability to move and function through postures and activities normally without creating undue stress on the vertebrae.
Therapeutic Exercises:
With or without mechanical assistance or resistance
Includes isoinertial, isotonic, isometric and/or isokinetic exercises
Designed to promote natural movement patterns
Potential Risks of Therapeutic Exercise in Individuals with SCI: (Jacobs, 2004)
Autonomic Dysreflexia: Individuals with injury at or above the T6 spinal cord level may develop autonomic dysreflexia after being exposed to noxious stimuli.
Musculoskeletal Injury: Individuals with SCI are at a greater risk for fracture and dislocation due to abnormal muscle synergies.
Orthostatic Hypotension
Thermal Dysregulation: Individuals with SCI may have decreased ability to maintain body temperature.
Passive Therapy: Does not require the patient to expend energy; effective during early phases of treatment; designed to control symptoms such as pain, inflammation and swelling; improves the rate of healing of soft tissue injuries; should be used in conjunction with active therapies. Passive therapy includes:
Electrical Stimulation
Infrared Therapy
Massage
Mobilization
Superficial heat and cold therapy
Short-wave diathermy
Traction
TENS
Ultrasound
Laser Therapy
Develop an appropriate out of bed sitting program
Wheelchair Evaluation and Mobility Training:
Many patients, depending on their level of injury and completeness of injury, may require long-term use of a wheelchair in order to maximize their independence at home and in the community.
Roles of a Physical Therapist's during WC Evaluation/Prescription: (Natale, 2009)
Measurement and Prescription of the Proper WC
Patient/Caregiver Education on the Advantages and Disadvantages of Certain WC Types
Proper Position in the Chair in order to prevent skin breakdown and postural deficits while also optimizing patient function
WC Propulsion Training
Management of doors, curbs, and elevators in order to maximize mobility in the community
Education:
Patients should understand that they can live a productive life in spite of a spinal injury, even if that injury is permanent.
The patient should grasp an understanding about what has happened and expected outcomes in the near and distant future from the health care team.
Many patients with a spinal injury understandably battle bouts of depression and will require emotional support from their family and health team members.
It is the role of the physical therapist to preserve muscle function and to teach techniques to overcome lost functions.
Physical therapists can also educate the patient and caregiver/family on certain modifications that may need to be made at home in order to accommodate the patient's level and severity of injury. (Natale, 2009)
Work as health care team with speech and language therapist, occupational therapists rehabilitation nurse, social worker, dietitian, psychologist and a specialized doctor.
+++
VI. Consumer and Professional Resources
++
+
Amendola
L, Corghi
A, Cappuccio
M, De Iure
F. Two cases of brown-séquard syndrome in penetrating spinal cord injuries. European Review for Medical Pharmacoogical Sciences. 2014;18(1 Suppl):2–7.
+
Bourassa-moreau
É, Mac-thiong
JM, Ehrmann
feldman D, Thompson
C, Parent
S. Complications in acute phase hospitalization of traumatic spinal cord injury: does surgical timing matter?.
J Trauma Acute Care Surg. 2013;74(3):849–54.
[PubMed: 23425747]
+
Cao
Y, Krause
JS, Dipiro
N. Risk factors for mortality after spinal cord injury in the USA. Spinal Cord. 2013; doi: 10.1038/sc.2013.2.
+
Guest
R, Craig
A, Middleton
J. Factors predicting resilience in people with spinal cord injury during transition from inpatient rehabilitation to the community. Spinal Cord. 2015; doi: 10.1038/sc.2015.32.
+
Illman
A, Stiller
K, William
M. The prevalence of orthostatic hypotension during physiotherapy treatment in patients with an acute spinal cord injury.
Spinal Cord. 2000;38(12):741–7.
[PubMed: 11175374]
+
International Standards for the Classification of Spinal Cord Injury: Motor Exam Guide. American Spinal Injury Association. http://www.asia-spinalinjury.org/elearning/Motor_Exam_Guide.pdf. Published June 2008a. Accessed February 26, 201.
+
Jacobs
P, Nash
M. Exercise Recommendations for Individuals with Spinal Cord Injury. Sports Medicine. August 2004;34(11):727–751.
+
Jia
X, Kowalski
RG, Sciubba
DM, Geocadin
RG. Critical care of traumatic spinal cord injury. J Intensive Care. 2013; 28:12–23.
+
Kirshblum
SC, Priebe
MM, Ho
CH, Scelza
WM, Chiodo
AE, Wuermser
L-A. Spinal Cord Injury Medicine. 3. Rehabilitation Phase After Acute Spinal Cord Injury.
Arch Phys Med Rehabil. 2007;88(3, Supplement 1):S62–S70.
[PubMed: 17321851]
+
Lee
WK, Koh
SW, Wee
SK. Benign paroxysmal positional vertigo in people with traumatic spinal cord injury: incidence, treatment efficacy and implications.
AM J Otolaryngol. 2012;33(6):723–730.
[PubMed: 22884482]
+
Mcdonald
JW, Becker
D, Holekamp
TF, Howard
M, Liu
S, Lu
A, Lu
J, Platik
MM, Qu
Y, Stewart
T, Vadivelu
S. Repair of the injured spinal cord and the potential of embryonic stem cell transplantation.
J Neurotrauma. 2004;21(4):383–93.
[PubMed: 15115588]
+
McDonald
JW, Sadowsky
C. Spinal-cord injury. Lancet. 2002;359:17–425.
+
Natale
A, Taylor
S, LaBarbera
J, Bensimon
L, McDowell
S, Mumma
SL, Backus
D, Zanca
JM, Gassaway
J. SCIRehab Project Series: The Physical Therapy Taxonomy.
J Spinal Cord Med. 2009;32(3):270–282.
[PubMed: 19810629]
+
Dreisbach
J, Dungan
D, Falci
S, Flanders
A, Lammertse
D, Marino
R, Schwartz
E. Neuroimaging in Traumatic Spinal Cord Injury: An Evidence-based Review for Clinical Practice and Research: Report of the National Institute on Disability and Rehabilitation Research Spinal Cord Injury Measures Meeting.
J Spinal Cord Med. 2007;30(3):205–214.
[PubMed: 17684886]
+
Rosenberg
LJ, Teng
YD, Wrathall
JR. 2,3-Dihydroxy-6-Nitro-7-Sulfamoyl-Benzo(f)Quinoxaline Reduces Glial Loss and Acute White Matter Pathology after Experimental Spinal Cord Contusion.
J Neurosci. 1999;19(1):464–475.
[PubMed: 9870974]
+
Sarhan
F, Saif
D, Saif
A. An overview of traumatic spinal cord injury: part 1. Aetiology and pathophysiology. Br Journal of Neurosci Nurs. 2012;8(6):319–325.
+
Sekhon
L, Fehlings
M. Epidemiology, Demographics, and Pathophysiology of Acute Spinal Cord Injury.
Spine. 2001; 26:S2–S12.
[PubMed: 11805601]
+
Sezer
N, Akkus
S, Ugurlu
FG. Chronic complications of spinal cord injury.
World Journal of Orthopedics. 2015;6(1):24–33.
[PubMed: 25621208]
+
Sidorov
EV, Townson
AF, Dvorak
MF, Kwon
BK, Steeves
J, Krassioukov
A. Orthostatic hypotension in the first month following acute spinal cord injury.
Spinal Cord. 2008;46(1):65–9.
[PubMed: 17420772]
+
Singh
A, Tetreault
L, Kalsi-Ryan
S, Nouri
A, Fehlings
MG. Global prevalence and incidence of traumatic spinal cord injury.
Clinical Epidemiology. 2014;6:309–331.
[PubMed: 25278785]
+
Stein
DM, Menaker
J, McQuillan
K, Handley
C, Aarabi
B, Scalea
TM. Risk factors for organ dysfunction and failure in patients with acute traumatic cervical spinal cord injury.
Neurocrit Care. 2010;13(1):29–39.
[PubMed: 20407933]
+
Vodusek
DB. Lower urinary tract and sexual dysfunction in neurological patients.
European Neurology. 2014;72:109.
[PubMed: 24993182]
+
Whiteneck
G. Outcomes following traumatic spinal cord injury: clinical practice guidelines for health-care professionals.
J Spinal Cord Med. 2000;23(4):289–316.
[PubMed: 17536300]
++
Trigeminal Neuralgia (TN) is related to hyperactive/abnormal discharges of the trigeminal nerve, with sensory distribution to the skin of the face as well as mucosa of orbital, nasal, and oral cavities (Iro, 2005).
Results from demyelinization of trigeminal sensory fibers which can be caused by a variety of pathologies either within the nerve root or the brain stem (Larsen, 2011).
Central cells - hyperactivity of the trigeminal nerve nucleus (Nurmikko, 2001; Wall, 2000).
Peripheral nerves (Bagheri, 2004; Love, 2001; Nurmikko, 2001)
Classical TN (idiopathic)
TN occurs in about 2% of the patients with MS, which is 20% higher than the incidence rate found in the general population (Zorro, 2009), MS is strongly associated with TN (Garg, 2012).
Patients with multiple sclerosis can experience plaque build-up at the trigeminal nucleus, the junction of the trigeminal nerve with the pons, or in the trigeminal nerve's descending tracts causing pain and dysfunction in the distribution of the Trigeminal nerve.
Impaired electrophysiological control in patients with diabetes mellitus can also trigger the onset of TN.
Herpes simplex virus and histamine release in the case of an allergic reaction are also possible causes of TN (Toda, 2007).
Vascular contact along the trigeminal nerve can be present on the symptomatic and asymptomatic side, and can occur at multiple locations along the nerve (Lorenzoni, 2012).
Onset of trigeminal neuralgia following surgery unrelated to the fifth cranial nerve may indicate that nerve contact with a vascular structure due to postoperative pressure and changes in the flow of cerebrospinal fluid may be the trigger of TN in patients with increased susceptibility (Duransoy, 2013).
25% of normal, asymptomatic individuals demonstrate vascular compression of the trigeminal nerve. This indicates that genetic tendency could be a factor in whether neurovascular compression leads to the clinical signs and symptoms of TN (Yang, 2009; Jia, 2010).
Classification
Idiopathic TN: Etiology remains unclear in 11% of cases (Cruccu, 2016).
Classical TN
Episodic pain resulting from vascular compression with morphologic changes of the trigeminal nerve root (Cruccu, 2016; IHS, 2013).
Most frequent form of TN with known etiology (Cruccu, 2016).
Symptomatic (secondary) TN:
Typical: only paroxysmal pain present (Screenivasan, 2014; Zakrzewska, 2014).
Atypical: both paroxysmal and constant pain present (Screenivasan, 2014).
Not related to a particular etiology, but can occur in idiopathic, classical, or secondary TN (Cruccu, 2016).
Painful trigeminal neuropathy is due to a lesion unrelated to vascular compression including: post-herpetic neuralgia, TN due to herpes zoster, posttraumatic TN, TN due to multiple sclerosis plaque, TN due to space occupying lesion, or TN due to any other disorder (IHS, 2013).
Demographics
Females affected twice as often as males (Garg, 2012, Van Kleef, 2009).
No racial differences reported (eMed, 2009).
Large range of age onset but most cases over 50 years old (Garg, 2012).
Incidence of TN increases in the elderly with highest incidence occurring between 50 and 70 years (Van Kleef, 2009; Iro, 2005)
Patients had an average age of onset of 52.9 years (Maarbjerg, 2014).
New research suggests patients with hypertension have an increased risk for developing TN (Pan, 2011).
TN diagnosis is more common in people with multiple sclerosis or stroke (Zakrzewkska, 2014).
Right side is affected more often than the left (Garg, 2012).
Autosomal dominant inheritance is seen in 1-2% of all cases (Wang, 2011; Riederer, 2010).
++
Characterized by brief and recurrent episodes of unilateral shock-like pain that affects one or more distributions of Trigeminal Nerve (CN V) initiated by a non-painful stimulus (IHS, 2013).
Past medical history of cranial trauma may be related as a mechanism of injury to the trigeminal nerve (NINDS, 2012).
Signs/symptoms:
Facial muscle spasm/wince on affected side as secondary effect/response to pain (TN originally tic douloureux --resemblance of spasm to a tic)
Rarely occurs bilaterally (if bilateral, primary cause may be something like MS)
No sensory deficit
May experience dull ache after severe attack but otherwise refractory periods between episodes; rarely occur during sleep.
Diagnostic Criteria for Classical TN according to The International Classification of Headache Disorders (ICHD), 3rd Edition (IHS, 2013)
A) Minimum of three attacks of unilateral facial pain that also fulfill parts B and C.
B) Pain occurs in one or more of the divisions of the trigeminal nerve (CN V) and does not radiate past the nerve's distribution.
C) Pain has minimum of three of the four characteristics below:
Paroxysmal attacks lasting from less than a second to two minutes.
Severe intensity.
Pain that is shock-like, shooting, sharp, or stabbing in quality.
Minimum of three attacks brought about by non-painful stimuli on the affected side of the face.
D) No neurological deficit.
E) Condition can not be explained by another ICHD-3 diagnosis.
Diagnostic Criteria for Classical TN (Van Kleef, 2009; Bennetto, 2007; Olesen, 2004).
A) Paroxysmal pain attacks that last from a fraction of a second to 2 minutes and affect one or more divisions of CN V
B) Pain can be characterized by at least one of the following:
Sharp, intense, superficial, or stabbing pain
Trigger areas (located in chin or nasolabial fold) or trigger factors (nonpainful sensory stimuli [a breeze, air conditioning, tooth brushing] or facial movements [talking, chewing, kissing, yawning])
C) Pain attacks "stereotyped" in the individual
D) No neurological deficit.
E) Pain not caused by another disorder.
Diagnostic Criteria for Symptomatic TN (Bennetto, 2007; Olesen, 2004)
Criteria 1, 2, and 3 are same as Classical TN
Discriminatory keys for symptomatic TN--trigeminal sensory deficits, bilateral involvement, and abnormal trigeminal reflexes (Cruccu, 2008).
Signs/symptoms: No refractory period between attacks of pain in Symptomatic TN; may have sensory deficit.
4. Lesion other than vascular compression identified (tumor, cyst, meningioma).
5. Abnormal Trigeminal reflexes are strongly associated with Symptomatic TN; Sensitivity = 96%, Specificity = 93% (Cruccu, 2006).
Diagnostic tests for both Classical and Symptomatic TN
MRI can help identify structural causes (Cruccu, 2008)
CAT scan (Meaney, 1995; Woolfall, 2001)
The 3D time-of-flight magnetic resonance angiography (3D-TOF-MRA) is a valid diagnostic tool for demonstrating neurovascular compression in TN with high sensitivity and specificity (Cai, 2015).
Neuro physiological tests may help in differentially diagnosing the symptomatic from classical TN. Symptomatic TN may present with (Zakrzewska, 2011):
"Abnormal trigeminal reflexes" (specificity 94%, sensitivity 87%).
Abnormal trigeminal nerve-evoked potentials.
Trigeminal sensory deficits and or bilateral involvement
Younger age of onset, involvement of first division, unresponsiveness to treatment and abnormal trigeminal evoked potentials not useful in distinguishing symptomatic from classic TN. (Cruccu, 2008).
++
Cycles of attack (Bagheri, 2004; Nurmikko, 2001)
Exacerbations and Remissions: up to 100 bouts/day, may last weeks to months, remissions lasting weeks to years
Attacks worsen over time: fewer and shorter pain-free periods
No impact on life expectancy but many report a significant decrease in their quality of life due to symptoms (Elaimy, 2012)
Sequelae: Possible depression and self-isolation
TN referred to as the "suicide disease" due to severity of pain (TNA, 2008).
Anxiety and depression disappear following relief of pain in patients with TN (Xia, 2016).
Symptoms become less responsive to treatment, even though medications and additional agent therapy increased (Zakrzewska, 2014).
Trigeminal neuralgia is short-lived in susceptible patients following intracranial operation (Duransoy, 2013).
Duration of TN symptoms is average of 6.4 years (Maarbjerg, 2014).
+++
IV. Medical/Surgical Management
++
+++
V. Preview of therapeutic management
++
Guide to Physical Therapy Preferred Practice Pattern (APTA, 2017):
Treatment for pain:
TENS (Niv, 1989).
Adjunctive therapeutic procedures to treat pain from TN like massage, meditation, relaxation are not well supported by the evidence (NHS, 2012).
"Repetitive transcranial electromagnetic stimulation at 10 Hz, 50 mA, and 20 minutes duration is more effective than low level laser therapy at reducing trigeminal pain, increasing maximum oral mouth opening, masseter and temporalis muscle tension in multiple sclerosis patients." (Seada, 2013)
Patient education/planning:
Be aware of medications the patient is taking as many anticonvulsants used to treat TN can cause drowsiness, ataxia, and diplopia (Zakrzewska, 2011) while also being aware of any other comorbidities (such as MS).
Electrical stimulation can be used to treat wounds due to neurotropic ulcers and habitual biting of cheeks, lips, and tongue that occur due to lack of sensation (Iro, 2005).
+++
VI. Consumer and Professional Resources
++
Traditionally, patients with TN have been managed through a biomedical model with no emphasis on psychological therapies. However, the prevalence of support groups has become more and more common with this disease (Zakrzewska, 2011).
Many resource groups exist online for patients to communication and gain information about their condition (Facial Neuralgia Resources).
Other agencies such as the National Institute of Neurological Disorders and Stroke offer information in addition to news regarding current research, clinical trials, and a database of support groups for TN patients (NINDS, 2012).
Facial Neuralgia.org
This is a patient-to-patient resource with links to access support groups, information about medications, surgery and other ways to treat TN. It provides a vector for communication with other people who are dealing with pain associated with TN
http://facial-neuralgia.org/
This page provides information regarding all types of medications that are prescribed to treat pain associated with TN. Information about side effects, dosing and many other helpful facts can be located here:
This webpage is a great resource for a variety of components. Patients can join a support group or talk with their peers who are also suffering from their diagnosis. There is also a link where a patient can find answers to questions they may have or purchase/read books and articles about their diagnosis. The "Find a Doctor" link may also be helpful for some people who need to find a doctor with a certain specialty.
UpToDate
Offers two patient education resources known as "The Basics" and "Beyond the Basics".
"The Basics" are patient education materials written at a 5th to 6th grade reading level and provides answers to questions the patient might have regarding their condition such as patients with trigeminal neuralgia.
"Beyond the Basics" is a more in-depth patient education resource that is written at a 10th-12th grade reading level and provides broader and more detailed information. These resources can be printed by the healthcare provider and given to the patient or be found at
http://www.uptodate.com /contents/trigeminal-neuralgia-the-basics?source=see_link)
+
Attal
N, Cruccu
G, Haanpää
M, Hansson
P, Jensen
TS, Nurmikko
T, Sampaio
C, Sindrup
S, Wiffen
P. EFNS guidelines on pharmacological treatment of neuropathic pain.
European Journal of Neurology. 2006;13:1153–1169.
[PubMed: 17038030]
+
Bagheri
SC, Farhidvash
F, Perciaccante
VJ. Diagnosis and treatment of patients with trigeminal neuralgia. J American Dental Association. 2004;135:1713–1717.
+
Barker
FG, Jannetta
PJ, Bissonette
DJ, Larkins
MV, Jho
HD. The Long-term outcome of microvascular decompression for trigeminal neuralgia.
The New England Journal of Medicine. 1996;334:1077–1083.
[PubMed: 8598865]
+
Bennetto
L, Patel
N, Fuller
G. Trigeminal Neuralgia and its management.
BMJ. 2007;334:201–205.
[PubMed: 17255614]
+
Bonaventura
R, Cioni
B, Meglio
M, Montano
N, Papacci
F. What is the best treatment of drug-resistant trigeminal neuralgia in patients affected by multiple sclerosis? A literature analysis of surgical procedures.
Clin Neurol and Neurosurg. July 2012.
http://dx.doi.org/10.1016/j.bbr.2011.03.031 +
Brisman
R. Trigeminal neuralgia: diagnosis and treatment. World Neurosurg. 2011 Dec; 76(6): 533–4.
+
Broggi
G, Ferroli
P, Franzini
A, Servello
D, Done
I. Microvascular decompression for trigeminal neuralgia: comments on a series of 250 cases including 10 patients with multiple sclerosis. J Neurology Neurosurgery Psychiatry. 2000;65:59–64.
+
Bünyamin
M, Burhanettin
U, Hüseyin
S, Muhammet
G. Successful Treatment of a Resistance Trigeminal Neuralgia Patient By Acupuncture. Clinics (Sao Paulo). 2009 December;64(12):1225–1226.
+
Cai
J, Xin
ZX, Zhang
YQ, Sun
J, Lu
JL, Xie
F. Diagnostic value of 3D time-of-flight MRA in trigeminal neuralgia.
J Clin Neurosci. 2015;22(8):1343–8.
[PubMed: 26077938]
+
Chandan
S, Halli
R, Sane
VD. Peripheral neurectomy: minimally invasive surgical modality for trigeminal neuralgia in Indian population: a retrospective analysis of 20 cases.
J Maxillofac Oral Surg. 2014;13(3):295–9.
[PubMed: 25018603]
+
Cruccu
G, Biasiotta
A, Galeotti
F, Iannetti
G, Truini
A, Gronseth
G. Diagnostic accuracy of trigeminal reflex testing in trigeminal neuralgia.
Neurology. 2006;66:139–141.
[PubMed: 16401867]
+
Cruccu
G
et al.. AAN-EFNS guidelines on trigeminal neuralgia management.
Eur J Neurol. 2008;15(10):1013–28.
[PubMed: 18721143]
+
Cruccu
G, Finnerup
N, Jensen
T,
et al. Trigeminal neuralgia: new classification and diagnostic grading for practice and research.
Neurology. 2016; 87: 220–28.
[PubMed: 27306631]
+
Duransoy
YK, Mete
M, Akçay
E, Selçuki
M. Differences in individual susceptibility affect the development of trigeminal neuralgia.
Neural Regen Res. 2013;8(14):1337–42.
[PubMed: 25206428]
+
Edwards
RJ, Clarke
Y, Renowden
SA, Coakham
HB. Trigeminal neuralgia caused by microarteriovenous malformations of the trigeminal nerve root entry zone: symptomatic relief following complete excision of the lesion with nerve root preservation.
Journal of Neurosurgery. 2002;97(4):874–80.
[PubMed: 12405376]
+
Elaimy
AL, Hanson
PW, Lamoreaux
WT,
et al. Clinical outcomes of gamma knife radiosurgery in the treatment of patients with trigeminal neuralgia. Int J Otolaryngol. 2012; 2012: 1–13. Epub 2011 Oct 25.
+
eMedicine. Trigeminal Neuralgia page. Available at: http://emedicine.medscape.com/article/1215194-overview. Accessed February 18, 2009.
+
Garg
A, Guez
G. The unnerving dilemma: understanding trigeminal neuralgia.
Dental Implantology Update. 2012;23(1):1–4.
[PubMed: 22308573]
+
Gronseth
G, Cruccu
G, Alskne
J,
et al. Practice parameter: the diagnostic evaluation and treatment of trigeminal neuralgia (an evidence based review): report of the Quality Standards Subcommittee of the American Academy of Neurology and the European Federation of Neurological Societies.
Neurology. 2008;71:1183–1190.
[PubMed: 18716236]
+
Headache Classification Committee of the International Headache Society (IHS). The International Classification of Headache Disorders, 3rd edition (beta version).
Cephalalgia. 2013;33(9):629–808.
[PubMed: 23771276]
+
Iro
H, Bumm
K, Waldfahrer
F. Rehabilitation of the trigeminal nerve. GMS Current Topics in Otorhinolaryngology Head and Neck Surgery. 2005;4:Doc12.
+
Jia
DZ, Li
G. Bioresonance hypothesis: a new mechanism on the pathogenesis of trigeminal neuralgia.
Med Hypotheses. 2010;74(3):505–507.
[PubMed: 19900765]
+
Kanpolat
Y, Savas
A, Bekar
A, Berk
C. Percutaneous controlled radiofrequency trigeminal rhizotomy for the treatment of idiopathic trigeminal neuralgia: 25-year experience with 1,600 patients.
Neurosurgery. 2001;48:524–32.
[PubMed: 11270542]
+
Kasat
V, Gupta
A, Ladda
R, Kathariya
M, Saluja
H, Farooqui
A-A. Transcutaneous electric nerve stimulation (TENS) in dentistry- A review.
J Clin Exp Dent. 2014;6(5):e562–568.
[PubMed: 25674327]
+
Larsen
A, Piepgras
D, Chyatte
D, Rizzolo
D. Trigeminal neuralgia: diagnosis and medical and surgical management. JAAPA. 2011 Jul; 24(7): 20–5.
+
Lorenzoni
J, David
P, Levivier
M. Patterns of neurovascular compression in patients with classic trigeminal neuralgia: A high-resolution MRI-based study. European Journal Of Radiology. August 2012;81(8):1851–1857.
+
Love
S, Coakham,
HB. Trigeminal neuralgia: Pathology and pathogenesis.
Brain. 2001;124:2347–2360.
[PubMed: 11701590]
+
Maarbjerg
S, Gozalov
A, Olesen
J, Bendtsen
L. Trigeminal neuralgia--a prospective systematic study of clinical characteristics in 158 patients.
Headache. 2014;54(10):1574–82.
[PubMed: 25231219]
+
Meaney
JF, Eldridge
PR, Dunn
LT, Nixon
TE, Whitehouse
G, Miles
JB. Demonstration of neurovascular compression in trigeminal neuralgia with magnetic resonance imaging: Comparison with surgical findings in 52 consecutive operative cases. J Neurosurgery. 1995;83:799–805.
+
Niv
D, Ben-Ari
S, Rappaport
A,
et al. Postherpetic neuralgia: clinical experience with a conservative treatment.
Clin J Pain. 1989;5:295–300.
[PubMed: 2562461]
+
Nurmikko
TJ, Eldridge
PR. Trigeminal neuralgia – pathophysiology, diagnosis and current treatment. British Journal of Anesthesia. 2001;87:117–32.
+
Obermann
M, Yoon
MS, Ese
D, Maschke
M, Kaube
H, Diener
HC, Katsarava
Z. Impaired trigeminal nociceptive processing in patients with trigeminal neuralgia.
Neurology. 2007;69(9):835–41.
[PubMed: 17724285]
+
Obermann
M, Yoon
MS, Sensen
K, Maschke
M, Diener
HC, Katsarava
Z. Efficacy of pregabalin in the treatment of trigeminal neuralgia.
Cephalalgia. 2008;28:174–181.
[PubMed: 18039340]
+
Olesen
J. The International Classification of Headache Disorders. Cephalalgia. 2004;24:1–152.
+
Pan
SL, Yen
MF, Chiu
YH, Chen
LS, Chen
HH. Increased risk of trigeminal neuralgia after hypertension: a population-based study.
Neurology. 2011;77(17):1605–10.
[PubMed: 21998318]
+
Riederer
F, Sandor
PS, Linnebank
M, Ettlin
DA. Familial occipital and nervus intermedius neuralgia in a Swiss family.
J Headache Pain. 2010;11(4):335–338.
[PubMed: 20349261]
+
Seada
YI, Nofel
R, Sayed
HM. Comparison between Trans-Cranial Electromagnetic Stimulation and Low-Level Laser on Modulation of Trigeminal Neuralgia.
J Phys Ther Sci. 2013;25(8):911–914.
[PubMed: 24259883]
+
Shah
SA, Khan
MN, Shah
SF, Ghafoor
A, Khattak
A. Is peripheral alcohol injection of value in the treatment of trigeminal neuralgia? An analysis of 100 cases.
Int J Oral Maxillofac Surg. 2011;40(4):388–92.
[PubMed: 21168309]
+
Shimohata
K, Shimohata
T, Motegi
R, Miyashita
K. Nasal sumatriptan as adjunctive therapy for idiopathic trigeminal neuralgia: report of three cases. Headache: The Journal of Head & Face Pain, 2009:49(5):768–70.
+
Smith
ZA, Gorgulho
AA, Bezrukiy
N, McArthur
D, Agazaryan
N, Selch
MT, De Salles
AA. Dedicated linear accelerator radiosurgery for trigeminal neuralgia: a single-center experience in 179 patients with varied dose prescriptions and treatment plans.
Int J Radiat Oncol Biol Phys. 2011;81(1):225–31.
[PubMed: 21236592]
+
Sreenivasan
P, Raj
SV, Ovallath
S. Treatment Options in Trigeminal Neuralgia an Update. Eur J Gen Med. 2014;11(3):209–216.
+
Toda
K. Etiology of Trigeminal Neuralgia. Int J Oral Sci. May 2007;4:10–18.
+
Wall,
P. Pain: The Science of Suffering. New York: Columbia University Press; 2000.
+
Wang
Y, Yu
CY, Huang
L, Riederer
F, Ettlin
D. Familial neuralgia of occipital and intermedius nerves in a Chinese family.
J Headache Pain. 2011;12(4):497–500.
[PubMed: 21607730]
+
Watson
CP. Management issues of neuropathic trigeminal pain from a medical perspective.
J Orofac Pain. 2004;18:366–373.
[PubMed: 15636022]
+
Woolfall
P, Coulthard
A. Trigeminal nerve: anatomy and pathology.
British Journal of Radiology. 2001;74:458–467.
[PubMed: 11388997]
+
Van Kleef
M, Van Genderen
WE, Narouze
S, Nurmikko
TJ, Van Zundert
J, Geurts
JW, Mekhail
N.. Trigeminal neuralgia. Pain practice. 2009;9(4):252–9.
+
Xia
JH, He
CH, Zhang
HF, Lian
YJ, Chen
Y, Wu
CJ, Ma
YQ. Botulinum toxin A in the treatment of trigeminal neuralgia.
Int J Neurosci. 2016;126(4):348–53.
[PubMed: 26000810]
+
Yang
KH, Na
JH, Kong
DS, Park
K. Combined hyperactive dysfunction syndrome of the cranial nerves.
J Korean Neurosurg Soc. 2009; 46(4):351–354.
[PubMed: 19893725]
+
Zakrzewska
JM, Linskey
ME. Trigeminal neuralgia.
BMJ Clinical Evidence. 2014; 2014:1207.
[PubMed: 25299564]
+
Zakrzewska
JM, McMillan
R. Trigeminal neuralgia: the diagnosis and management of this excruciating and poorly understood facial pain. Postgrad Med J. 2011 Jun; 87(1028): 410–6.
+
Zakrzewska
JM, Patsalos
PN. Long-term cohort study comparing medical (oxcarbazepine) and surgical management of intractable trigeminal neuralgia.
Pain. 2002;95:259–266
[PubMed: 11839425]
+
Zorro
O, Lobato-Polo
J, Kano
H, Flickinger
J, Lunsford
L, Kondziolka
D. Gamma knife radiosurgery for multiple sclerosis-related trigeminal neuralgia. Neurology. October 6, 2009;73(14):1149–1154.
+++
Vestibular Hypofunction (Unilateral or Bilateral)
++
Unilateral Vestibular Hypofunction (UVH): With unilateral damage, neuronal activity reaching the ipsilateral nuclei is reduced compared to the contralateral side. The brain translates the asymmetry in firing rates as head rotation. (Schubert, 2004)
Common cause: vestibular neuritis, secondary to viral infections (herpes simplex) (Schubert, 2004)
Other causes: Meniere's disease, vestibular schwannoma, vascular lesions or unilateral brain injury. (Schubert, 2004)
Iatrogenic causes: complication of ablative surgery (Weber, 2008)
Bilateral(BVH): abnormal firing in both ears that can cause issues such as vertigo (Schubert, 2010)
Causes: Aminoglycoside antibiotics (gentamicin, streptomycin), which are known to damage vestibular hair cells and cause ototoxicity (Schubert, 2010)
Damage is selective to the hair cells so audition is spared (Schubert, 2010)
Other causes:
Meningitis, head trauma, tumors on bilateral eighth cranial nerves, bilateral vestibular schwannoma, transient ischemic episodes of vessels perfusing the vestibular system, unilateral vestibular neuronitis (Schubert, 2010; Hodge, 2012)
Degeneration of the cristae ampullaris of the semicircular canals, which sense angular head rotations.
Displays the most profound degeneration in the vestibular end organ, 40% decrease in hair cells for all canals by age 80 (Chau, 2015)
++
Subjective findings:
Complaints occur during head movement in any direction but may be present at rest
Vertigo, disequilibrium, gait ataxia, postural instability, oscillopsia, nystagmus, and decrements in dynamic visual acuity (DVA). (Schubert, 2004; Herdman, 2003)
Objective findings:
Subjects with uncompensated UVH posturally sway to moving visual environments (Sparto, 2006), with impaired balance (increased Timed Up and Go time).
Common associated musculoskeletal findings include protracted head and shoulders, reduced trunk flexibility and ability to relax, tense muscles of the upper trunk and legs, and restricted abdominal breathing. (Wilhelmsen, 2014)
Increased upper extremity movement time and decreased head velocity during voluntary reaching to offset gaze stability impairments. (Borello-France, 2002)
Spontaneous nystagmus: slow components directed toward side of lesion, with fast components directed toward contralateral side of lesion. (Hain, 1987)
Absence of responses with ice-cold caloric stimulations is indicative of bilateral loss of vestibular function (Albernaz, 2016)
Self report questionnaires
Activity and participation restrictions (Alghwiri, 2011):
Vestibular Disorders Activity Daily Living (VADL) scale, Activities of Daily Living Questionnaire (ADLQ), Activities Balance Confidence (ABC) scale, Dizziness Handicap Inventory (DHI), and UCLA Dizziness questionnaire (UCLA-DQ)
Functional impairments (Alghwiri, 2011):
Diagnostic Testing:
Head Thrust for semicircular canal function:
The Head Shaking Induced Nystagmus (HSN) Test
Bedside Head Impulse Test (bHIT)
The video-head-impulse test (vHIT)
Assesses all 6 semicircular canals (Albernaz, 2016)
Positive when a correcting saccade is observed because the eye cannot compensate for head movement., and more sensitive and specific than the (bHIT) (Mahringer, 2014)
vHIT is largely independent of age and should be performed over caloric to evaluate semicircular function (Maheu, 2015)
Head Impulse Test (HIT) with high head acceleration revealed VOR deficits better than low acceleration (Weber, 2008)
High Speed Video-assisted Head Impulse Test (HSVA-HIT)- will reveal catch-up saccade in different planes after rotation to the healthy side. (D'Onofrio, 2013)
Positional Testing to rule out benign paroxysmal positional vertigo (BPPV).
Electronystagmography (ENG):vestibular function and organization with a series of battery tests. (Markley, 2007)
DVA test: ability to read a screen while head is shaking. (Schubert, 2006)
Measurement of visual acuity during horizontal motion of the head.
Identify the functional significance of the vestibular hypofunction.
Head velocities need to be greater than 100°/s in order for the vestibular afferents on the contralateral side to be driven into inhibition. (Schubert, 2010)
Sensory Organization Test (SOT) of dynamic posturography with head shaking, sensitive for UVH but not specific (Mishra, 2009).
The Sideways Stepping Test (SST) is useful in supporting a diagnosis of unilateral vestibular hypofunction (UVH) and is positive when there is involuntary leaning or hip swaying to one side or when subject lifts hand or takes steps in order to compensate(Al Saif, 2014)
Unterberger-Fukuda Test provides signs of unilateral vestibular dysfunction and is positive when there is greater than 1 m displacement and/or rotation higher than 30° (Rosha Junior, 2014)
++
++
Vestibular Ocular Reflex: measured by monitoring eye motion during stimulation of the peripheral vestibular system and is expressed as eye velocity/head velocity.
Eyes not adducting: VOR gain is –1 = implying a compensatory eye velocity equal to the head velocity and in the opposite direction. (Schubert, 2010)
Semicircular Canal Function: caloric test, "gold standard," for identifying peripheral UVH.
Cold or warm stimulus in the external auditory canal causes a temperature gradient, change in temperature noted at the lateral temporal bone causing a convective flow generating a nystagmus. (Schubert, 2010)
Vestibular-evoked myogenic potentials (VEMP) test: series of loud (95 dB) clicks while assessing the SCM
Normal: inhibitory potential is followed by an excitatory potential
Vestibular hypofunction: the VEMPs are absent on the side of the lesion. (Schubert, 2010)
Cervical VEMP (cVEMP) – assesses saccular function, the inferior vestibular nerve and vestibular nuclei.
Ocular VEMP (oVEMP) – reflects utricular function and involves the medial longitudinal fasciculus, oculomotor nuclei, and nerves.
Using oVEMP and cVEMP together allows for the evaluation of both ascending and descending vestibular pathways in the brainstem and identifies a higher percentage of abnormalities (Kim, 2015)
Subjective visual vertical (SVV) and subjective visual horizontal (SVH) tests: used to assess otolith function.
SVV test: align a bar with what they perceive as being vertical. (Schubert, 2010)
SVH test: align a bar with what they perceive as being horizontal.
Normal: align the bar within 1.5 degrees of true vertical or horizontal
UVH: align the bar more than 2 degrees of true vertical or horizontal with the bar tilted toward the lesioned side. (Schubert, 2010)
++
Short-term, individualized vestibular rehabilitation therapy (VRT) shown to improve "symptoms, handicaps, balance, and postural stability" (Giray, 2009) through compensation.
Improvements at 3 weeks using VRT including gaze stabilization techniques and habituation. (Schubert, 2006)
Recovery of dynamic visual acuity in less than 5 weeks of vestibular exercises. (Herdman, 2003)
Since vestibular cues contribute to gaze stabilization during optokinetic stimulation around line of sight, this may be part of long-term problems post unilateral loss of vestibular functions (Lopez, 2005).
Patients with BVH have shown improvements in DVA after treatment with vestibular exercises (Herdman, 2007)
After effective treatment, patient's with little to no vestibular function may return to driving (MacDougall, 2009)
++
Variables associated with prognosis:
++
Patients with a greater loss of vestibular function may improve, but aren't likely to return to normal DVA after VRT. (Herdman, 2012)
Increased risk for falls as predicted by the Dynamic Gait Index (Hall, 2004) and the Tetrax Balance System posturagraphy (Quitschal, 2014).
Poor rehabilitation results may be attributable to increased severity of vestibular insult, progressive peripheral or central vestibular dysfunction, and multiple medical problems. (Gillespie, 1999)
Recovery depends on initial severity of the disability, when treatment is received and their prior level of activity. Patients with co-morbidities have limited potential for full recovery. (Gillespie, 1999)
The sum effects of initial DVA, percentage of time symptoms interfere with life, and initial disability score are related to degree of disability at discharge. This allows the PT to evaluate the benefits and determine the need for assisted devices. (Herdman, 2012)
Occurrence of spontaneous nystagmus varies (Fetter, 1990; Cass, 1992)
+++
IV. Medical/Surgical Management
++
Pharmacological Treatment:
Medications suppress vestibular system and reduce nystagmus and other imbalance symptoms (Hain, 2003)
Promethazine, Meclizine, diazepam, and Lorazepam (Walker, 2009)
Antihistamines (Dramamine®), anticholinergics (Elavil®), antidopaminergics (Reglan®), and GABA enhancing agents (Lorazepam) reduce vertigo symptoms. (Underbrink, 2008; Hain, 2003)
If due to Vestibular neuritis, then medication decreases inflammation. (Underbrink, 2008)
If Vestibular neuritis is the cause, it can be treated with oral steroids, anti herpes agents or both (Walker, 2009)
If due to Meniere's disease, then diuretics with salt restrictive diet. (Underbrink, 2008)
H4 receptor antagonists modulate the activity of vestibular activity (Desmadryl, 2012)
Endogenous stimulation of GABAB receptors in vestibulo-oculomotor circuits reduces the vestibular asymmetry during early period after UVH. (Magnusson, 2002)
Surgical Treatment:
Labyrinthectomy creates a stable unilateral lesion needing VRT. (Shepard, 1995)
Vestibule neurectomies beneficial in those with underlying Meniere's disease. (Topuz, 2004)
Surgery to remove causal tumor (Hodge, 2012)
+++
V. Implications For Therapeutic Management
++
Preferred Practice Patterns: (APTA, 2017)
5A: Prevention/Risk Reduction for Loss of Balance and Falling
5D: Impaired Motor Function and Sensory Integrity Associated with Progressive Disorders of the Central Nervous System- Acquired in Adolescence or Adulthood
5E: Impaired Motor Function and Sensory Integrity Associated with Progressive Disorders of the Central Nervous System
5F: Impaired Peripheral Nerve Integrity and Muscle Performance Associated With Peripheral Nerve Injury
Physical Interventions
Treatment for vestibular deficits:
Habituation exercises as developed by Cawthorne-Cooksey are used to teach the patient to compensate for vestibular deficits (Underbrink, 2008).
By addressing musculoskeletal dysfunction and promoting body awareness, patients with UVH may inhibit a self-sustaining cycle of dizziness and musculoskeletal problems (Wilhelmsen, 2015).
Task oriented virtual reality (VR) interactive training methods provide a new functional approach for treatment of UVH (Chen, 2012)
Vestibular rehabilitation used in conjunction with Wii® therapy decreases perception of disability, improves dynamic gait index, and improves gaze stability in patients with unilateral vestibular hypofunction (Verdeechia, 2014).
Vertiguard vibrotactile stimulation improves the body's balance in patients who did not achieve good response to training by vestibular rehabilitation (Brugnera, 2015)
Treatment for decreased VOR:
Improved accuracy of vestibular-ocular reflex via VOR stimulation exercises (Schubert, 2006)
VOR adaption to position error signals such as the "imaginary target" paradigm improve VOR (Scherer, 2010)
Adaptation exercises develop compensatory saccadic eye movements, which provide substitution for an impaired VOR (Brodovsky, 2013)
Treatment for visual acuity:
Gaze stabilization exercises improve visual acuity. (Schubert, 2006; Giray, 2009)
iPod-based in-home application system is useful in monitoring the compliance and performance of gaze-stabilization exercises in individuals with vestibular hypofunction. (Huang, 2014)
Quality of Life:
Aquatic therapy improves quality of life, balance, and self-perception of dizziness intensity. (Gabilan, 2008)
Prevention of Falls:
Prevention of falls via dynamic and static balance and gait exercises improve postural control. (Schubert, 2006; Giray, 2009)
Real time vibrotactile feedback during locomotor activity decreases medial/lateral sway (Sienko, 2013)
Start VRT at slow speeds, in one position, and quiet environments; progress accordingly. (Underbrink, 2008)
Contraindications/Precautions
Fall risk
Triggered by position
Possible panic reactions due to nystagmus
Nausea/vomiting
+++
VI. Consumer and Professional Resources
++
+
Al Saif
AA, Alsenany
S. The Efficiency of the Sideways Stepping Test in Detecting Unilateral Vestibular Hypofunction.
J Phys Ther Sci. 2014;26(11):1719–1722.
[PubMed: 25435685]
+
Albernaz
PLM, Cusin
FS. The Video Head Impulse Test in a Case of Suspected Bilateral Loss of Vestibular Function.
Int Arch Otorhinolaryngol. 2016;20(1):84–86.
[PubMed: 26722351]
+
Alghwiri
AA, Marchetti
GF, Whitney
SL. Content Comparison of Self-Report Measures Used in Vestibular Rehabilitation Based on the International Classification of Functioning, Disability and Health.
Phys Ther. 2011;91(3):346–357. doi:10.2522/ptj.20100255.
[PubMed: 21252307]
+
Borello-France
DF, Gallagher
JD, Furman
JM, Redfern
MS, Carvell
GE. Voluntary upper-extremity movements in patients with unilateral peripheral vestibular hypofunction.
Phys Ther. 2002;82(3):216–227.
[PubMed: 11869150]
+
Brodovsky
JR, Vnenchak
MJ. Vestibular Rehabilitation for Unilateral Peripheral Vestibular Dysfunction.
Phys Ther. 2013;93(3):293–298.
[PubMed: 23329556]
+
Brown
KE, Whitney
SL, Wrisley
DM, Furman
JM. Physical therapy outcomes for persons with bilateral vestibular loss.
Laryngoscope. 2001;111(10): 1812–7.
[PubMed: 11801950]
+
Brugnera
C, Bittar
RSM, Greters
ME, Basta
D. Effects of vibrotactile vestibular substitution on vestibular rehabilitation – preliminary study.
Braz J Otorhinolaryngol 2015;81(6):616–621.
[PubMed: 26480904]
+
Cass
SP, Kartush
JM, Graham
MD. Patterns of vestibular function following vestibular nerve section.
Laryngoscope. 1992;102(4):388–94.
[PubMed: 1556887]
+
Chau
AT, Menant
JC, Hübner
PP, Lord
SR, Migliaccio
AA. Prevalence of Vestibular Disorder in Older People Who Experience Dizziness.
Front Neurol. 2015;6:268. doi:10.3389/fneur.2015.00268.
[PubMed: 26733940]
+
Chen
P-Y, Hsieh
W-L, Wei
S-H, Kao
C-L. Interactive wiimote gaze stabilization exercise training system for patients with vestibular hypofunction.
J Neuroeng Rehabil. 2012;9:77. doi:10.1186/1743-0003-9-77.
[PubMed: 23043886]
+
Desmadryl
G, Gaboyard-Niay
S, Brugeaud
A,
et al. Histamine H4 receptor antagonists as potent modulators of mammalian vestibular primary neuron excitability. Brit J Pharmacol. 2012;167(4):905–916.
+
D'Onofrio
F. Vertical eye movements during horizontal head impulse test: a new clinical sign of superior vestibular neuritis.
Acta Otorhinolaryngol Ital. 2013;33(6):418–424.
[PubMed: 24376299]
+
Fetter
M, Dichgans
J. Adaptive mechanisms of VOR compensation after unilateral peripheral vestibular lesions in humans.
J Vestib Res. 1990;1(1):9–22.
[PubMed: 1670141]
+
Gabilan
YP, Perracini
MR, Munhoz
MS, Gananc
FF. Aquatic physiotherapy for vestibular rehabilitation in patients with unilateral vestibular hypofunction: exploratory prospective study [abstract].
J Vestib Res. 2008;18(2-3):139–146.
[PubMed: 19126984]
+
Gillespie
MB, Minor
LB. Prognosis in bilateral vestibular hypofunction.
Laryngoscope. 1999;109(1):35–41.
[PubMed: 9917037]
+
Giray
M, Kirazli
Y, Karapolat
H, Celebisoy
N, Bilgen
C, Kirazli
T. Short-term effects of vestibular rehabilitation in patients with chronic unilateral vestibular dysfunction: a randomized controlled study.
Arch Phys Med Rehabil. 2009;90(8):1325–31.
[PubMed: 19651266]
+
Hain
TC, Fetter
M, Zee
DS. Head-shaking nystagmus in patients with unilateral peripheral vestibular lesions.
Am J Otolaryngol. 1987;8(1):36–47.
[PubMed: 3578675]
+
Hain
TC, Uddin
M. Pharmacological treatment of vertigo.
CNS Drugs. 2003;17(2):85–100.
[PubMed: 12521357]
+
Hall
C, Schubert
M, Herdman
S. Prediction of fall risk reduction as measured by dynamic gait index in individuals with unilateral vestibular hypofunction. Otology & Neurotology. 2004;25:746–751.
+
Herdman
SJ, Hall
CD, Delaune
W. Variables associated with outcome in patients with unilateral vestibular hypofunction.
Neurorehabil Neural Repair. 2012;26(2):151–62.
[PubMed: 21959673]
+
Huang
K, Sparto
PJ, Kiesler
S, Siewiorek
DP, Smailagic
A. iPod-based in-home system for monitoring gaze-stabilization exercise compliance of individuals with vestibular hypofunction.
J Neuroeng Rehabil. 2014;11:69–81.
[PubMed: 24746068]
+
Jorns-Haderli
M, Straumann
D, Palla
A. Accuracy of the bedside head impulse test in detecting vestibular hypofunction.
J Neurol Neurosurg Psychiatry. 2007;78:1113–1118.
[PubMed: 17220287]
+
Kim
CH, Jang
MU, Choi
HC, Sohn
JH. Subclinical vestibular dysfunction in migraine patients: a preliminary study of ocular and rectified cervical vestibular evoked myogenic potentials. J Headache Pain. 2015;16. doi:10.1186/s10194-015-0578-5.
+
Lopez
C, Borel1
L, Magnan
J, Lacour
M. Torsional optokinetic nystagmus after unilateral vestibular loss: asymmetry and compensation.
Brain. 2005;128(7):1511–1524.
[PubMed: 15917290]
+
MacDougall
HG, Moore
ST, Black
RA, Jolly
N, Curthoys
IS. On-road assessment of driving performance in bilateral vestibular-deficient patients. Ann N Y Acad Sci. 2009 May;1164:413–8.
+
Magnusson
A, Ulfendahl
M, Tham
R. Early compensation of vestibulo-oculomotor symptoms after unilateral vestibular loss in rats is related to GABAB receptor function.
Neuroscience. 2002;111(3):625–634.
[PubMed: 12031349]
+
Mahringer
A, Rambold
HA. Caloric test and video-head-impulse: a study of vertigo/dizziness patients in a community hospital.
Eur Arch Otorhinolaryngol. 2014;271(3):463–472.
[PubMed: 23494283]
+
Maheu
M, Houde
M-S, Landry
SP, Champoux
F. The Effects of Aging on Clinical Vestibular Evaluations. Front Neurol. 2015;6. doi:10.3389/fneur.2015.00205.
+
Markley
BA. Introduction to electronystagmography for END technologists.
Am J Electroneurodiagnostic Technol. 2007;47(3):178–89.
[PubMed: 17982846]
+
McDonnell
MN, Hillier
SL. Vestibular rehabilitation for unilateral peripheral vestibular dysfunction. The Cochrane Library. 2015 Jan 13. DOI: 10.1002/14651858.CD005397.pub4.
+
Mishra
A, Davis
S, Speers
R, Shepard
NT. Sensory Organization Test (SOT) of dynamic posturography. Am J Audiol. 2009 Jun;18(1):53–9.
+
Quitschal
RM, Fukunaga
JY, Ganança
MM, Caovilla
HH. Evaluation of postural control in unilateral vestibular hypofunction.
Braz J Otorhinolaryngol. 2014;80(4):339–345.
[PubMed: 25183185]
+
Rocha
Júnior PR, da Silva
Peres A, Garbi
FP, Frizzo
AC, Valenti
VE. Effects of physiotherapy on balance and unilateral vestibular hypofunction in vertiginous elderly.
Int Arch Med. 2014;7:8–11.
[PubMed: 24576350]
+
Scherer
M, Schubert
MC. High-Velocity Angular Vestibulo-Ocular Reflex Adaptation to Position Error Signals:
J Neurol Phys Ther. 2010;34(2):82–86.
[PubMed: 20588093]
+
Schubert
M, Migliaccio
A, Della
Santina C. Modification of compensatory saccades after aVOR gain recovery.
J Vestib Res. 2006;16:285–291.
[PubMed: 17726282]
+
Schubert
M, Minor
L. Vestibulo-ocular physiology underlying vestibular hypofunction.
Phys Ther. 2004;84:373–385.
[PubMed: 15049730]
+
Schubert
M, Tusa
R, Grine
L, Herdman
S. Optimizing the sensitivity of the head thrust test for identifying vestibular hypofunction.
Phys Ther. 2004;84:151–158.
[PubMed: 14744205]
+
Sienko
KH, Balkwill
MD, Oddsson
LIE, Wall
C. The effect of vibrotactile feedback on postural sway during locomotor activities.
J Neuroeng Rehabil. 2013;10:93. doi: 10.1186/1743-0003-10-93
[PubMed: 23938136]
+
Sparto
C, Furman
J, Redfern
M. Head sway response to optic flow: effect of age is more important than the presence of unilateral hypofunction.
J Vestib Res. 2006;16:137–145.
[PubMed: 17312341]
+
Topuz
O, Topuz
B, Ardic
N, Sarhus
M, Ogmen
G, Ardic
F. Efficacy of vestibular rehabilitation on chronic unilateral vestibular dysfunction.
Clin Rehabil. 2004;18:76–83.
[PubMed: 14763722]
+
Verdecchia
DH, Mendoza
M, Sanguineti
F, Binetti
AC. Outcomes after vestibular rehabilitation and Wii® therapy in patients with chronic unilateral vestibular hypofunction.
Acta Otorrinolaringol Esp. 2014;65(6):339–345.
[PubMed: 24997123]
+
Weber
KP,
et al. Head impulse test in unilateral vestibular loss. vestibulo-ocular reflex and catch-up saccades.
Neurology. 2008;70:454–463.
[PubMed: 18250290]
+
Whitney
S, Wrisley
D, Furman
J. Concurrent validity of the Berg Balance Scale and the Dynamic Gait Index in people with vestibular dysfunction.
Physiother Res Int. 2003;8(4):178–186.
[PubMed: 14730722]
+
Wilhelmsen
K, Kvåle
A. Examination and treatment of patients with unilateral vestibular damage, with focus on the musculoskeletal system: A case series.
Phys Ther. 2014;94(7):1024–1033.
[PubMed: 24557651]
++
The Compendium authors acknowledge each of the following physical therapists graduates from University of South Alabama who contributed to the ongoing literature searches, collecting information, and progressive development of these Diagnosis Outlines and also to the general content of the Focus on Evidence tables for chapters throughout the textbook. Dr Fell expresses his sincere thanks and personal wishes for a productive and rewarding career to each of you:
++
Class of 1996: Kirk Allen, Andy Biggs, Jason Boatfield, Stacey Boris, Mitzie Burgess, Michael Clark, Tracey Coale, Wendy Davidson, Christi Davis, Kimberly Enlow Baxter, Amada Forward, Jeanie Garner, Tyler Hale, Shannan Halverson, Dody Herring, Karyn Lanier, Ron Lassiter, Samuel Lawrence, Jennie Lewis, Kristie Love, Amy Maurer, Marian Merritt, Paula Roeser, Michael Smith, Petra Stolt, Debra Taylor, Jeff Thomas, Mark Turner, Ramona Vowels, Day Wallace, Bob Wessel, Keri White, Leigh Wiggins, Louann Williams, Jennifer Wilson, Cindy Wilson, Deane Wilson, Heidi Yurcisin.
++
Class of 1997: Tanya Adams, Shawn Babcock, Jason Blackburn, Donovan Brooks, Michelle Bryan, Jennifer Burroughs, Tamara Campbell, Kristye Chastang, Jason Chisolm, Michael Courtenay, Barbara Dady, Tony DiRusso, Melissa Dryden, Amy Dunbar, Richard Dymond, Thuymai Fee-Vazquez, Julie Fleisher, Jeremy Gray, Brittney Hogue, Laura Jones, Rhonda Kirchoff, Brian Lackey, DeeDee Martin, Brian McReath, Miira Midgorden, Julie Norton, William Perkins, Daniel Peters, Candi Poiroux, Kerry Richardson, Patrick Roberts, Susan Seal, Jennifer Shiver, Heather Sizemore, Monica Smith, Stacy Smith, Mike Tucker, Christine Vegas, Teresa Wells.
++
Class of 1998: Jennifer Abbott, Edward Adams, Nicole Andino, Churan Bell, Stephanie Beveridge, Emily Brigance, Timothy Burnell, Stewart Campbell, Jennifer Davis, Deanna Drew, Amy Forsyth, Damon Garcia, Misty Gibson, Angella Greer, Jean Guy, Joshua Hamilton, Cary Helton, Drake Ibsen, Ashley Isbell, Laura Jackson, Olivia Jones, Jennifer Lincecum, Matthew Mandeville, Jay Megginson, Danielle Morrow, Lauren Muller, Amy Norman, Rita Parsley, Scott Pruitt, Carrie Rice, Stephen Schonk, Brian Seymore, Katy Shrauner, Linda Smith, Elizabeth Stubblefield, James Wallace, Brant Wilson, Gray Woodham, Ryan Yolitz, Aimee Youngblood.
++
Class of 1999: Andrew Bennett, Allison Blythe, Kim Boler, Julie Buscavage, Leigh Cannon, Brian Davis, Melissa Dowling, Steven Estrada, Amanda Fleener, Lisi Fox, Melanie Galla, Rita Gedney, Bill Gill, Heather Giroir, Dan Hartman, Andrew Hatch, Jennifer Erin Hicks, Tiffany Hill, Shawna House, Jolanta Hunter, Robert Keane, Tim Lutkins, Erin Mason, Lisa Munson, Robert Murphy, III, Jamey Odom, Lori Porter, Jennifer Prevost, Lisa Reddington, Susan Thomas, Monica Thompson, Jessica Tranchina, Alexandra Vaughn, Claudia Velez, Karl von Tiesenhausen, Jerrod Walker, Thomas Walters, Gail Willamson, Lance Willis, Mark Willis, Richard Wilson.
++
Class of 2000: Leigh Anne Alexander, J. David Bass, Jack Bentley, Michael Beuoy, Brent Boardman, Jessica Bostick, Regina Brown, Randolph Budd, Shannon Newman, Valerie Clenney, Emily Fedor, Rick Gamble, Monique Gandy, Mark Granger, William Hale, Tammy Hardy, Valerie Hare, Chris Hidgon, Anna Howard, Lori Hurley, Amy Langille, Joshua Lenox, Darius McAphee, Rashad Pearson, Jill Renegar, Jaime Rhodes, Kristina Scheblein, Karen Schoennagle, David Smith, Teresa Smith, David Sweatt, Hope Tapley, Shane White, Jamie Williams, Loretta Yoder.
++
Class of 2001: Amy Apker, Maria Camacho, Cameron Cannon, Douglas Cole, Lori Cowan, Sandy Eldridge, Rich Evans, Jennifer Farris, Melanie Fayard, Kellie Gallaway, Margaret Harper, Natasha Howell, Lea Hurlburt, Marc Hussey, Katrina Kirkland, Kallen Krause, Tressa Melancon, Jennifer Munch, LaDonna Pate, Gita Patel, Lorrie Peoples, Mark Pilkinton, April Rich, Jason Riley, Sheri Shultz, Cheryl Sellers, Sabrina Simmons, Christina Toland, Jill Van Camp, Jennifer Von Antz, MaryAnna Wavra, Catherine Welford, Ruthba Zaman.
++
Class of 2002: Maria Benvenutti-Payne, Polly Bhattacharia, Ashlie Blake, Atiba Callins, Emily Collins, Candace Crosby, Sarah DeWit, Christopher Drewes, Laura Estes, William Farnell, Shon Frachiseur, Jessica Hartley, Gabrielle Hornsby, Shasta Johnson, Kimberly Jones, Sarah Knopf, Shannon Lassiter, Amelia Lowe, Lisa Lowery, Joy Lyda, Jocelyn Mayfield, Jennifer Montgomery, Charlie Murphy, Melissa Reed, Monica Reese, Ashley Riggs, Christina Shelley, Amanda Smith, Kelly Smith, Michelle Smith, Seth Smothers, Digby Watt, Kacey Wehmeier.
++
Class of 2003: Jonathan Baird, Brandon Balenger, Lavon Beard, Richard Brasseaux, Joann Brown, Adrian Call, Katie Fletcher, Brian Guttmann, Rachel Hicks, Scott Higgs, Amy Jacobs, Lynn Johnson, Pamela Jones, John Kirk, III, Melissa Lewis, Jennifer McLaughlin, Elizabeth Monk, Nicole Neve, Shellie Parchman, Mason Reid, Dwan Shoemake, Stacy Simmons, Emily Smith, Desshondra Walker,
++
Class of 2004: Jennifer Andress, Jaclyn Berry, Jackeline Castro-Cooper, Amberly Cooper, Tyson Crisman, Mindy Forsyth, Wendy Forsyth, Sara-Kathryn Green, Trista Griffin, Crystal Gunn, Jamie Hall, Leigh Ann Kittrell, Matthew Madere, Jennifer Orleans-Sorrell, Megan Pockrus, Jeff Porter, Justin Rich, Kalyn Russell, Christopher Thompson, Amy Vining, Bonnie Vogel, Erin Williams, MollyAnna Wilson.
++
Class of 2005: Patrick Benefield, Ashley Wells, Robert Bolton, Barbara Borg, Curtis Borum, Diane Boyd, Roger Broome, Lauren Brown, Alexis Clarkson, Brandon Cloud, George Collins, Brandon Cox, Jennifer Daigrepont, Katherine Davidson, Lucinda Davis, Tara Eaton, Abby Ellenburg, Julie-Anne Foley, Mark Gagnon, Beate Gant, Angela Gattis, Anna Goynes, Trisch Grimes, Courtney Haynes, Lauren Held, Luke Hendrix, Jorel Hernani, Dana Hester, Heather Hillhouse, Karen Kelley, Angela Lambard, Thomas Lett, Whitney Livingston, Chandra Miles, Nick Nuchereno, Quin Sirmon, Jean Soileau, Brian Strawbridge, Lindy Vann, Hillary Willrodt, Amanda Yerbey, Jessica Young.
++
Class of 2007: Lydia Allen, Mary Bounds, Stephanie Boyer, Chad Caswell, Kelly Crager, Kyle Ford, Heather Higgins, Crystal Jones, Bethany Duncan, Summer Mathis, Megan Mayeux, Tiffany Mcginnis, Bethany Merkel, Katherine Moore, Anthony Nuchereno, Valerie Shankland, Jonathan Shaw, Amanda Smolinski, Heather Sollie, Sheena Fanton, Christopher Stewart, Kaidong Tang, Stephanie Thomas-Ware, Melanie Thomason, Austin Trippe, Janelle Wixwat, Catherine Wood.
++
Class of 2008: Chris Allison, Kimberly Bentley, Megan Boos, Brittany Boyte, Julie Brown Weathers, Lisa Brown, Nicholas Butler, Timothy Clemmons, Kimberly Cornell, Elizabeth Crosby, Justin Fetsko, Rachel Fountain, Laura Gilliam, Julie Henderson, Lisa Hundley, Julie Ingram, Alan Jensen, Christina Kilborn, Erin Kinnear, Charles Kirchem, Lauren Lacoste, Jacob McKenzie, Erin O'Rourke, Melanie Owens, Adam Ridgdell, Alicia Sappenfield, Daniel Singler, Timothy Sirmon, Jennifer Spann, Michael Stallworth, Amanda Thomas, James Warren, Jonathan Weathers, Kathleen Wimberly, Wendy Woods.
++
Class of 2009: Patrick Bonvillain, Tammi Brasfield, Melanie Buchanan, Erin Burke, Rebecca Butler, Natalie Day, Julie Gamble, Kevin Gasque, Karen Gettys, Ernest Green, Sara Greer, Cassie Harmon, Ryan Harrell, Heidi Horchen, Cari Hudson, Brad Hutto, Enoch Jones, Kimberly LeBlanc, Melvia Mims, Kasey Moore, Jonathan Moran, Tan Nguyen, Brittney Paisley, Katelyn Parkes, Elizabeth Pearson, Lorie Powell, Cherie Ricau, Joel Scherr, Lindsey Scott, Shelby St. John, Joanie Thomas, Ashley Vaccarella, Tricia Williams, Emily Windham.
++
Class of 2010: Kali Adams, Tara Andrews, Richard Cahanin, Jarrod Cain, Mallory Campbell, Mallory Crafton, Marci Dill, Jennifer Douglas, Jeremy Fletcher, Kelley Greathouse, Susan Howard, Joshua Hutto, Olga Jenus, Zachary Kirby, Jennifer Lanier, Tara Lindsey, Martha McDaniel, Kalyn McNair, Katherine Milner, Lindsay Nemanich, Alicia Ponsock, Julie Porche, Larry Rapp, Jackson Rawls, Kirstin Reggel, Robin Rouse, Brittany Shaw, Leslea Stands, Lauren Steiner, Preston Warren, Derek Winstead.
++
Class of 2011: Julie Brantley, Avery Bridgmon, Jamie Cain, Jamie Castjohn, Holly Cauthen, Patrick Childress, Patricia Cooper, Jon Dockery, Clint Gallo, Michael Giardina, Cassie Gibson, Sherre Gibson, Annie Gilliland, David Greer, Matthew Griggs, Neil Isom, Arthur Kremer, Jessica Kucala, Kristen Locicero, Anthony Marino, Allison Meyer, Baley Moseley, Jennifer Odom, Lauren Pritchard, Bradley Redmond, Cameron Riley, Matthew Salter, Stacey Schacherer, Jena Smith, Lori Smith, Jarred Watson, Sarah Weaver, Erin Wood.
++
Class of 2012: Ashley Baker, Laura Ballard, Lauren Bee, Jordan Brewer, Chelsie Butler, Clint Caballero, Katelin Caraway, Sarah Deshotel, Ashely Dodd, Samuel Goff, Deborah Griffin, Kristi Hatten, Mary Hoppis, Christopher Hornbuckle, Jenae Jackson, Christine Kelly, Lindsey Kreig, Matt Lopez, Jennifer Melancon, Alicia Miles, Steven Mistretta, John Murphy, Merri Murphy, Rebecca Oliver, Raleigh Reid, Kimberly Rudd, Camille Shipman, Courtney Turner, Brittney Tynes, Sara Walker.
++
Class of 2013: Amber Blocker, Johnathon Breland, Justin Broussard, Alexander Burkett, Traci Busker, Michelle Cadden, Jessica Clements, Caroline Coleman, Eric David, Meredith Dillon, Emily Foster, Louis Haar, Matthew Howell, Corey Irby, Morgan Jackson, Rachel Kozakiewicz, Christopher Lawson, Jillian Mars, Melanie McDonald, Kayla Mowdy, Caitlin Oliver, Katie Peavy, Megan Peterson, Elizabeth Phillips, Coral Quinn, Amanda Schermerhorn, Michelle Segretto, Katherine Strickland, J. Clay Thomley, Madison Walker, Tabitha Weed, Leah Wilson, Ryan York.
++
Class of 2014: Joni Barnes, Alex Bell, Michelle Bell, Amanda Brewer, Corey Brown, Christina Calametti, Amanda Caravella, Ryan Clayton, Jared Cole, Sydnie Cole, Tyler Covington, Andrew Crowley, John Fletcher, Cory Gibson, Yan Goh, Stephanie Jensen, Sophia LeBlanc, Kenna McDaniel, Dale Meitzler, Ryan Myers, William Parker, Amanda Peterson, Tiffany Pickett, Michelle Ray, Luke Rowe, Jeana Stanley, Madalyn Staub, Evan Stringfellow, Sarah Taylor, Ashley Theobald, Sharon Thompson, Jackson Walker, Zachary Willers, Darlene Williams, Johnitia Williams, Benjamin Wilt, Kacie Wilt.
++
Class of 2015: Robert Best, Alan Bigbee, Brooke Constant, Elizabeth Davis, Olivia Deese, Noah Dickens, Kristen Donnelly, Lauren Dorsett, Brianna Foster, Kasey Hamric, Rachel Harp, Jared Henson, Erin Higginbotham, Caroline Hollowell, Meridith Johnson, Megan Josey, Emily LaRue, Jessie Lazzari, Lucas Lindsey, Tim Lipe, Melissa Payne, Ty Peterson, Brittany Poist, Benjamin Roussel, Christina Shepherd, Alexa Sheridan, Ashley Smith, Sara Stringfellow, Byrnes Tatford, Amie Vette, Jason Wamsley, Nicholas Wheeler.
++
Class of 2016: Michael Anderson, Nadia Araiinejad, Michael Ard, Lauren Barrios, Lee Bland, Rachel Blavos, Joshua Cash, Erin Cersley, Mary Davis, Sharyn DeMoss, Emily DeVries, Aaron Estilette, Kayla Glenzer, Taylor Hallum, Kaitlyn Hambrick, Brittany Hanks, Charles Hubbard, Meagan Jackson, Jade Jones, Colin Kruse, Lauren Lewis, Bonetia Linder, Callie Lyon, Albert Mayhan, Allison MacKay, Bradley Murph, Dalton Newell, Benjamin Parrish, Christen Pontius, Laura Risse, Kathryn Roberts, Lauren Roberts, Sarah Roberts, Garris Strickland, Amber Sumrall, John Taylor, Hunter Todd, Lindsey Tubb, Matthew Wilt, Brandon Wyatt, Joseph Zebrowski.
++
Class of 2017: Janice Albert, Ashley Bannister, Megan Battles, Aimee Beets, Zachary Boyd, Lauren Breland, Lauren Cleavelin, Marissa De Los Santos, Morgan Duncan, Kristen Estes, Dillion Evans, Luke Ford, Alyssa Free, Christopher Glover, Bailey Harrelson, Ryan Haskins, Rush Hendricks, Sean Hiller, Cameron King, Mallory Kraft, Jaunitha Magwood, Sarah Megahee, Krista Melsheimer, Madeline Osbrink, Mary Philips, Charles Phillips, Taylor Price, Lucas Quebedeaux, Kayla Rogers, Christa Roscigno, Erik Schmich, Savannah Shumake, Jonathan Smart, Molly Stephan, Alexandra Straughn, Kyle Thibodeaux, Nicholas Tolstick, Tyler Vaughn, Shay Williams, Leah Winstead.
++
Class of 2018: Sarah Allen, Aubrey Bennett, Rachel Bostick, Kayla Cobbs, Vincent Cochran, Jonathan Corley, Michael Cowan, Charles Davis, Hannah Davis, Dana Dowell, Jared Dowling, Elizabeth Ehrhardt, Laura Fish, Abby Fitts, Marissa Gacek, Sophia Girardeau, Elizabeth Glover, Shannon Herold, Heather Jeziorski, Brock Kirby, Tiffany Latham, Melanie Lewis, Grace Nix, Hillary Papich, Kaitlyn Revette, Serena Roddam, Laura Rose, Christy Saia, Leslie Salisbury, Maggie Smith, Kyle Stringfellow, Lydia Stringfield, Aubrey Toole, Silas Tucker, Christina Tygielski, Morgan Wesson, Susan Whitted, Phillip Wyatt.