Unlabeled Uses of Tremor Treatments
Unlabeled Uses of Tremor Treatments
888 AU G U S T 2 0 1 9
C O N T I N U U M J O U R N A L .C O M 889
890 AU G U S T 2 0 1 9
C O N T I N U U M J O U R N A L .C O M 891
892 AU G U S T 2 0 1 9
The issue begins with articles on the primarily Dr Pichet Termsarasab next reviews the disorders
hypokinetic movement disorders, starting with one that present with chorea, including Huntington
by Dr Theresa A. Zesiewicz, who informs us about disease and various other choreic disorders, including
the most current approach to the diagnosis those that may resemble Huntington disease.
and optimal management of our patients with Dr Sheng-Han Kuo then provides us with a practical
idiopathic Parkinson disease. Next, Dr Paul Greene approach to the diagnosis and management of the
discusses the pathologic underpinnings, clinical ataxias. In the article that follows, Dr John N.
features, and diagnosis of progressive supranuclear Caviness provides us with an approach to the
palsy, corticobasal degeneration, and multiple categorization, diagnosis, and management of the
system atrophy. Although these three various forms of myoclonus we can encounter in
neurodegenerative disorders have been our practices.
historically—and still occasionally—referred to as Dr Joseph H. Friedman then reviews the diagnosis
Parkinson plus syndromes (a term that seems to have and current approaches to management of the
devolved from the somewhat more apt term tardive syndromes, potentially debilitating
parkinsonism plus1), this arguably archaic moniker syndromes that unfortunately remain a significant
has been “subtracted” from Continuum since these public health problem due to exposure to any of the
(not uncommon) disorders are not related to currently available dopamine receptor–blocking
Parkinson disease and are distinct diseases that are agents. Drs Toni S. Pearson and Roser Pons next
diagnosable by their classic and unique clinical and provide a thorough review of the diagnosis and
pathologic features. management of the many movement disorders that
The issue then turns to the hyperkinetic may occur in childhood. In the final review article of
movement disorders, starting with the article by the issue, Drs Mary Ann Thenganatt and Joseph
Dr Harvey S. Singer, who reviews the diagnosis and Jankovic share their expertise in the diagnosis and
management of patients with tics and Tourette management of functional (ie, psychogenic)
syndrome. This is followed by the article by Dr Louis, movement disorders, including identification of
who discusses the diagnosis and management of those positive clinical features that should suggest
our patients with tremor, whether predominantly the diagnosis.
action or resting. Dr H. A. Jinnah then reviews the In this issue’s Medicolegal Issues article, Dr Joseph
diagnosis and management of the many disorders S. Kass and Ms Rachel V. Rose present a hypothetical
characterized by the presence of dystonia. case of a new patient requesting a refill for a
CONTINUUMJOURNAL.COM 895
Parkinson Disease
C O N T I N U U M AUDIO By Theresa A. Zesiewicz, MD, FAAN
INTERVIEW AVAILABLE
ONLINE
VIDEO CONTENT ABSTRACT
A VA I L A B L E O N L I N E PURPOSE OF REVIEW:Parkinson disease is a common neurodegenerative
disorder that affects millions of people worldwide. Important advances in
CITE AS:
CONTINUUM (MINNEAP MINN) the treatment, etiology, and the pathogenesis of Parkinson disease have
2019;25(4, MOVEMENT DISORDERS): been made in the past 50 years. This article provides a review of the
896–918.
current understanding of Parkinson disease, including the epidemiology,
Address correspondence to
phenomenology, and treatment options of the disease.
Downloaded from http://journals.lww.com/continuum by BhDMf5ePHKbH4TTImqenVCDV3BItTVWTLH8JPlxaxC5EkKx9p9Ad0yc3ePNxUjt5P+IqNDHuKFk= on 08/03/2019
Dr Theresa A. Zesiewicz,
University of South Florida, 12901 RECENT FINDINGS: Parkinson disease is now recognized to be a heterogeneous
Bruce B. Downs Blvd, MDC Box
55, Tampa, FL 33612,
condition marked by both motor and nonmotor symptoms. It is composed
[email protected]. of preclinical, prodromal, and clinical phases. New medications with
improved ease of administration have been approved for its treatment.
RELATIONSHIP DISCLOSURE:
Dr Zesiewicz has received Innovative surgical therapies for Parkinson disease may be used when
personal compensation for motor symptoms persist despite optimal medical management.
serving on the advisory boards of
Boston Scientific; Reata
Pharmaceuticals, Inc; and SUMMARY: Parkinson disease is a complex, heterogeneous neurodegenerative
Steminent Biotherapeutics. Dr disorder. Considerable progress has been made in its treatment modalities,
Zesiewicz has received personal
compensation as senior editor for
both pharmacologic and surgical. While its cure remains elusive, exciting
Neurodegenerative Disease new research advances are on the horizon.
Management and as a consultant
for Steminent Biotherapeutics. Dr
Zesiewicz has received royalty
payments as co-inventor of
INTRODUCTION
varenicline for treating imbalance
P
(patent number 9,463,190) and arkinson disease is a chronic, progressive, and disabling disorder that
nonataxic imbalance (patent is characterized by both motor and nonmotor symptoms. The disease
number 9,782,404). Dr Zesiewicz
has received research/grant
affects millions of people worldwide and is the second most prevalent
support as principal investigator/ neurodegenerative condition next to Alzheimer disease.1 Patients
investigator for studies from experience progressive extrapyramidal motor symptoms, including
AbbVie Inc; Biogen; Biohaven
Pharmaceutics; Boston Scientific; tremor, bradykinesia, rigidity, imbalance, and a variety of nonmotor symptoms
Bukwang Pharmaceuticals Co, Ltd; such as sleep and mood disorders. Despite its progressive nature, it remains one
Cala Health, Inc; Cavion; of the few neurodegenerative diseases whose symptoms can be readily treated
Friedreich’s Ataxia Research
Alliance; Houston Methodist with dopamine replacement therapy.
Research Institute; National
Institutes of Health (READISCA
U01); Retrotope Inc; and Takeda BRIEF HISTORY OF PARKINSON DISEASE
Development Center
Americas, Inc.
The English physician, James Parkinson, first characterized Parkinson disease in
his 1817 monograph “An Essay on the Shaking Palsy.”2 Parkinson described
UNLABELED USE OF several people who presented with resting tremor, shuffling gait, stooped
PRODUCTS/INVESTIGATIONAL
USE DISCLOSURE:
posture, sleep problems, and constipation. He noted the progressive nature of
Dr Zesiewicz reports no the disease and the great disability it incurred and called it paralysis agitans.
disclosure. Charcot3 later expounded on the disease, adding bradykinesia and rigidity to the
constellation of symptoms, and renamed the condition Parkinson disease.
© 2019 American Academy In the 1950s Carlsson4 found that levodopa reversed reserpine-induced
of Neurology. akinesia, paving the way for its use as a treatment for Parkinson disease. Cotzias
Motor Symptoms
The four cardinal motor symptoms of Parkinson disease are tremor, rigidity,
bradykinesia, and postural instability, as identified by the acronym TRAP
(tremor, rigidity, akinesia (or bradykinesia), and postural instability)
(TABLE 1-1).13 The secondary motor symptoms include diminished arm swing,
decreased blink rate, masked facies (hypomimia), decreased voice volume
(hypophonia), and difficulty turning over in bed.
Tremor refers to a rhythmic oscillation around a fixed point in the “rest” or
nonpostural position. Tremor is often the first motor symptom of Parkinson
disease and affects approximately 90% of patients at some point in their lives
(VIDEO 1-1, links.lww.com/CONT/A348).14 While the tremor of Parkinson disease
is typically a resting tremor, 50% of patients may also present with a tremor than
may reoccur with arms stretched outward.14 Tremors start asymmetrically and
are characterized by supination and pronation, or pill-rolling, eventually affecting
CONTINUUMJOURNAL.COM 897
the opposite side of the body. Tremors may be less responsive to pharmacologic
treatment, including levodopa.
Rigidity refers to stiffness or resistance of a limb when it is flexed passively,
activating both agonist and antagonist muscles, and may also be referred to as
cogwheeling.15 Bradykinesia refers to slowness of movement (akinesia refers to
lack of movement) and may occur during both initiation and continuation of
movement.16
Postural instability, or balance dysfunction, is experienced later in the
course of the disease, about a decade after initial diagnosis. Postural instability
Premotor Symptoms
◆ Constipation
◆ Anosmia
◆ Rapid eye movement (REM) sleep behavior disorder
◆ Depression
Nonmotor Symptoms: Neuropsychiatric
◆ Depression
◆ Anxiety (mood disorders)
◆ Apathy
◆ Impulse control disorder
◆ Psychosis
◆ Anhedonia
◆ Hallucinations
◆ Abulia
◆ Attention deficit disorder
◆ Panic attacks
Nonmotor Symptoms: Cognitive
◆ Executive dysfunction
◆ Memory loss
◆ Dementia
Nonmotor Symptoms: Autonomic
◆ Orthostatic hypotension
◆ Constipation
◆ Fecal incontinence
◆ Nausea
◆ Vomiting
◆ Drooling
CONTINUUMJOURNAL.COM 899
Nonmotor Symptoms
In the last 20 years, there has been increasing recognition of the importance of
nonmotor symptoms (ie, symptoms other than those involved in movement,
such as tremor, rigidity, and bradykinesia) in diagnosing and treating Parkinson
disease. It is estimated that nearly all patients with Parkinson disease will
experience several concurrent nonmotor symptoms throughout the course of
the disease. The impact from nonmotor symptoms is often greater than that
of motor symptoms; unfortunately, nonmotor symptoms are often underrecognized.
The nonmotor symptoms of Parkinson disease are listed below and are included
in TABLE 1-1.
CONTINUUMJOURNAL.COM 901
CONTINUUMJOURNAL.COM 903
TABLE 1-2 UK Parkinson’s Disease Society Brain Bank Clinical Diagnostic Criteriaa
CONTINUUMJOURNAL.COM 905
CONTINUUMJOURNAL.COM 907
Levodopa Levodopa 84 mg, 168 mg, 252 mg, 336 mg, Oral inhalation Intermittent treatment
inhalation powder 420 mg of off episodes in
patients with PD using
carbidopa/levodopa
Carbidopa/ Levodopa plus 12.5 mg/50 mg/200 mg; 18.75 mg/ Oral Indicated for end-of-
levodopa/ catechol-O- 75 mg/200 mg; 25 mg/100 mg/ dose wearing off
entacapone methyltransferase 200 mg; 31.25 mg/125 mg/200 mg;
(COMT) inhibitor 37.5 mg/150 mg/200 mg; 50 mg/
200 mg/200 mg
Pramipexole Dopamine agonist 0.125 mg, 0.25 mg, 0.5 mg, Oral Treatment of PD
immediate release 0.75 mg, 1 mg, 1.5 mg symptoms
Pramipexole Dopamine agonist 0.375 mg, 0.75 mg, 1.5 mg, 2.25 mg, Oral Treatment of PD
extended release 3 mg, 3.75 mg, 4.5 mg symptoms
Ropinirole Dopamine agonist 0.25 mg, 0.5 mg, 1 mg, 2 mg, 3 mg, Oral Treatment of PD
immediate release 4 mg, 5 mg symptoms
Selegiline oral MAO-B inhibitor 1.25 mg, 2.5 mg Sublingual Adjunct to levodopa
disintegrating therapy in patients with
PD who experience a
deterioration in positive
response to the
levodopa therapy
Amantadine NMDA antagonist 129 mg, 193 mg, 258 mg Oral Treatment of dyskinesia
extended-release in patients with PD
tablet receiving levodopa
therapy
CONTINUUMJOURNAL.COM 909
CASE 1-3 A 75-year-old woman presented for evaluation of an arm tremor. The
patient’s primary care physician observed a right-sided resting tremor
during a routine well care visit earlier that year and referred her to a
movement disorders center for evaluation. She dragged her right leg
when walking and reported stiffness and pain in her right arm and leg. She
reported daytime somnolence due to a long history of insomnia, and
described mild forgetfulness.
On examination, the patient was mildly hypertensive, but her vital
signs and physical examination were otherwise normal. Her Mini-Mental
State Examination was 27 out of 30. The patient had moderate
bradykinesia, moderate resting tremor in her right hand, and mild resting
tremor in her left hand, with rigidity noted in all limbs. She had
moderately stooped axial posture, and she had a narrow-based gait with
shortened strides. The patient took several steps back on the pull test
but recovered unaided.
The patient was diagnosed with Parkinson disease. She was placed
on carbidopa/levodopa 25 mg/100 mg, 1 tablet 4 times daily, with tablets
taken before meals. She returned to clinic after 6 weeks with marked
improvement in motor symptoms.
COMMENT The patient had Parkinson disease and exhibited the cardinal symptoms of
bradykinesia, resting tremor, and rigidity. She demonstrated supportive
criteria for the disease as well as the absence of exclusionary criteria and
red flags. The symptoms were bothersome to the patient, so treatment
was appropriate.
Levodopa remains the gold standard of treating Parkinson disease
and is the single most effective agent to treat all stages of the disease.70
The patient was in her midseventies, had mild cognitive impairment and
excessive daytime sleepiness, and was clearly in need of dopaminergic
therapy.
Dopamine agonists were not used in this case as they can worsen
excessive daytime sleepiness and exacerbate neuropsychiatric
symptoms. Anticholinergic medications might improve her tremor, but
will do little for the bradykinesia and rigidity she experiences, and
patients who are 70 years of age and older may also be more prone to
experiencing side effects from their use.
review regarding the controversy over initiating levodopa treatment in younger ● Patients with Parkinson
patients. Levodopa is a safe and efficacious medication for practically reversing disease should be offered
disease symptoms for a period of time. Motor fluctuations and dyskinesias may dopaminergic treatment
be more closely associated with longer disease duration and higher levodopa when their symptoms are
bothersome. Patients with
daily dose rather than the duration of levodopa therapy.67 Pulsatile delivery of Parkinson disease should be
levodopa also contributes to dyskinesia.68 As the disease progresses, higher and encouraged to exercise, as
more frequent doses of levodopa are required. While no definitive evidence long as it is performed
indicates that levodopa induces cell death, symptomatic therapy should be safely.
initiated while also considering both short-term and long-term potential
side effects.66
Dopamine agonists, MAO-B inhibitors, or anticholinergic medications may be
initiated in patients with Parkinson disease who are younger than 70 years of
age. However, non–levodopa medications eventually will be insufficient to
effectively ameliorate motor symptoms, and patients will need to be treated with
levodopa (levodopa rescue).
Treatment of younger patients with levodopa should be considered if
symptoms are bothersome enough to cause suffering or interfere with qualify
of life. Younger patients may need more effective control of their symptoms
with levodopa if they need to remain employed or have other responsibilities
including childcare or eldercare. Patients may also be unable to receive optimal
treatment with non–levodopa agents because of untoward side effects. Several
studies suggest that starting treatment with levodopa leads to better long-term
motor outcomes and better functioning long-term.20 In older patients with
evidence of cognitive decline, excessive daytime sleepiness, or other comorbid
conditions, it is more appropriate to initiate treatment for Parkinson disease
with levodopa. Dopamine agonists, MAO-B inhibitors, and anticholinergic
medications are more likely than levodopa to cause cognitive side effects in
the elderly.
Exercise should be encouraged for all patients with Parkinson disease as
long as it is performed safely. Some evidence suggests that long-term aerobic
exercise may slow Parkinson disease progression.69 Studies to confirm this
hypothesis are ongoing.19 Exercise modalities include core strength training
exercises, tai chi, yoga, boxing, and dance and music therapy. Cognitive
training with puzzles and computer games should also be encouraged
(CASE 1-3).
CONTINUUMJOURNAL.COM 911
OFFICE VISIT
Patients with Parkinson disease will most likely visit a neurology or movement
disorder specialist for treatment. Health care providers must pay attention to
both motor and nonmotor symptoms of the disease. Information about any
recent falls, swallowing issues, comorbid conditions, hospitalizations, or a
change in living arrangements should be obtained. Nonmotor symptoms
including constipation, pain, and mood disorders should not be neglected (refer
to the section on nonmotor symptoms). Patients should be encouraged to visit
their primary care physicians and other specialists for general health and
psychiatric care.
CONTINUUMJOURNAL.COM 913
the table according to when the patient reports they are taken. Attention should
be paid to whether the medications have been given in immediate-release or
controlled-release forms.
Questions regarding motor response may include the following:
u How much time does it take for your Parkinson disease medications to take effect
after each dose?
u How long does the effect of each medication last? (Record this information for each
dose interval.)
u Do you develop dyskinesia during the mid-dose period? Does dyskinesia occur toward
the beginning or end of the dose interval?
u Do you experience wearing off toward the end of the dose interval? Can you estimate
how long this occurs before the next dose begins?
u Do you experience early morning dystonia or pain or curling in your limbs? Does dystonia
occur at other times during the day?
u Do you have periods of the day when your medications do not seem to work? Do
these periods occur around meal times?
CONCLUSION
Parkinson disease is a complex neurodegenerative disease that appears to be a
heterogeneous disorder. Exciting research continues to take place regarding the
etiology and pathogenesis of the disease. Novel neuroimaging techniques such
as SPECT scans may now assist with disease diagnosis. New formulations of
Parkinson disease medications are available for easier administration and
improved clinical efficacy.
Treatment of Parkinson disease continues to be symptomatic, however,
and the disease cannot yet be cured. The realization that Parkinson disease
has a preclinical phase has prompted the need for early biomarkers.
Research continues to focus on neuroprotective and disease-modifying
strategies, including therapies targeting α-synuclein. Future research in the
etiology, pathophysiology, and ultimately a cure for Parkinson disease
remains hopeful, considering the remarkable progress made in the last
half century.
ACKNOWLEDGMENTS
The author would like to thank Shaila Ghanekar and Yarema Bezchlibnyk, MD,
PhD, FRCSC, for their help and guidance.
REFERENCES
1 Hirtz D, Thurman DJ, Gwinn-Hardy K, et al. 10 de Lau LM, Breteler MM. Epidemiology of
How common are the “common” neurologic Parkinson's disease. Lancet Neurol 2006;5(6):
disorders? Neurology 2007:68(5):326–327. 525–535. doi:10.1016/S1474-4422(06)70471-9.
doi:10.1212/01.wnl.0000252807.38124.a3.
11 Elbaz A, Bower JH, Maraganore DM, et al. Risk
2 Parkinson J. An essay on the shaking palsy. tables for parkinsonism and Parkinson’s disease.
London: Whittingham and Rowland Sherwood, J Clin Epidemiol 2002;55(1):25–31. doi:10.1016/
Neely and Jones, 1817. S0895-4356(01)00425-5.
3 Charcot JM. Lectures on the diseases of the 12 Taylor KS, Cook JA, Counsell CE. (2007)
nervous system, delivered at La Salpetriere. Heterogeneity in male to female risk for
London: The New Sydenham Society, 1877. Parkinson’s disease. J Neurol Neurosurg
Psychiatry 78(8):905–906. doi:10.1136/jnnp.
4 Carlsson A, Lindqvist M, Magnusson T.
2006.104695.
3,4-Dihydroxyphenylalanine and
5-hydroxytryptophan as reserpine 13 Jankovic J. Parkinson’s disease: clinical features
antagonists. Nature 1957;180(4596):1200. and diagnosis. J Neurol Neurosurg Psychiatry
doi:10.1038/1801200a0. 2008;79(4):368–376. doi:10.1136/jnnp.2007.
131045.
5 Cotzias GC, Van Woert MH, Schiffer LM.
Aromatic amino acids and modification of 14 Obeso J, Stamelou M, Geotz CG, et al. Past,
Parkinsonism. N Engl J Med 1967;282(7):31–33. present, and future of Parkinson’s disease: a
doi:10.1056/NEJM196702162760703. doi:10.1056/ special essay on the 200th anniversary of the
NEJM196702162760703. shaking palsy. Mov Disord 2017;32(9):1264–1310.
doi:10.1002/mds.27115.
6 Olanow CW, Watts RL, Koller WC. An algorithm
(decision tree) for the management of 15 Samii A, Nutt JG, Ransom BR. Parkinson's disease.
Parkinson’s disease (2001): treatment guidelines. Lancet 2004;363(9423):1783–1793. doi:10.1016/
Neurology 2001;56(11 suppl 5):S1–S88. doi:10.1212/ S0140-6736(04)16305-8.
WNL.56.suppl_5.S1.
16 Berardelli A, Rothwell JC, Thompson PD, Hallett M.
7 Ahlskog JE, Muenter MD. Frequency of Pathophysiology of bradykinesia in Parkinson's
levodopa-related dyskinesias and motor disease. Brain 2001;124(pt 11):2131–2146. doi:
fluctuations as estimated from the cumulative 10.1093/brain/124.11.2131.
literature. Mov Disord 2001;16(3):448–458.
17 Luquin MR, Scipioni O, Vaamonde J, et al.
doi:10.1002/mds.1090.
Levodopa-induced dyskinesias in Parkinson’s
8 Cell Signaling Technology. Cell Signaling disease: clinical and pharmacological
Technology web site. Dopamine signaling in classification. Mov Disord 1992:7(2):117–124.
Parkinson's disease interactive pathway. doi:10.1002/mds.870070204.
cellsignal.com/reference/pathway/parkinsons_
18 Tolosa E, Compta Y. Dystonia in Parkinson’s
disease.html. Accessed June 9, 2019.
disease. J Neurol 2006;253(suppl 7):vii7–vii13.
9 Nussbaum RL, Ellis CE. Alzheimer’s disease and doi:10.1007/s00415-006-7003-6.
Parkinson’s disease. N Engl J Med 2003;348:
19 Aquino CC, Fox SH. Clinical spectrum of
1356–1364.
levodopa-induced complications. Mov Disord
2015;30(1):80–89. doi:10.1002/mds.26125.
CONTINUUMJOURNAL.COM 915
20 Morgan JC, Fox SH. Treating the motor symptoms 34 Hirsch E, Graybiel AM, Agid YA. Melanized
of Parkinson disease. Continuum (Minneap Minn) dopaminergic neurons are differentially
2016;22(4):1064–1085. doi:10.1212/CON. susceptible to degeneration in Parkinson’s
0000000000000355. disease. Nature 1988;334(6180):345–348.
doi:10.1038/334345a0.
21 Giladi N, Nieuwboer A. Understanding and
treating freezing of gait in parkinsonism, 35 Lang AE, Obeso JA. Time to move beyond
proposed working definition, and setting the nigrostriatal dopamine deficiency on Parkinson’s
stage. Mov Disord 2008;23(suppl 2):S423–S425. disease. Ann Neurol 2004;55(6):761–765.
doi:10.1002/mds.21027. doi:10.1002/ana.20102.
22 Chaudhuri KR, Healy DG, Schapira AH; National 36 Nutt JG. Motor subtype in Parkinson's disease:
Institute for Clinical Excellence. Non-motor different disorders or different stages of
symptoms of Parkinson's disease: diagnosis and disease? Mov Disord 2016;31(7):957–961.
management. Lancet Neurol 2006;5(3):235–245. doi:10.1002/mds.26657.
doi:10.1016/S1474-4422(06)70373-8.
37 Gan-Or Z, Liong C, Alcalay RN. GBA-associated
23 Storch A, Schneider CB, Wolz M, et al. Nonmotor Parkinson's disease and other synucleinopathies.
fluctuations in Parkinson disease: severity and Curr Neurol Neurosci Rep 2018;18(8):44.
correlation with motor complications. Neurology doi:10.1007/s11910-018-0860-4.
2013;80(9):800–809. doi:10.1212/WNL.
38 Mazzulli JR, Xu YH, Sun Y, et al. Gaucher
0b013e318285c0ed.
disease glucocerebrosidase and α-synuclein
24 Pfeiffer RF. Management of autonomic form a bidirectional pathogenic loop in
dysfunction in Parkinson’s disease. Semin Neurol synucleinopathies. Cell 2011;146:37–52.
2017;37(2):176–185. doi:10.1055/s-0037-1601568. doi:10.1016/j.cell.2011.06.001.
25 Goldman JG, Postuma R. Premotor and nonmotor 39 Hauser RA, Grosset DG. [123I]FP-CIT (DaTscan)
features of Parkinson's disease. Curr Opin SPECT brain imaging in patients with suspected
Neurol 2014;27(4):434–441. doi:10.1097/WCO. parkinsonian syndromes. J Neuroimaging 2012;
0000000000000112. 22(3):225–230. doi:10.1111/j.1552-6569.2011.
00583.x.
26 Postuma RB, Berg D, Stern M, et al. MDS clinical
diagnostic criteria for Parkinson's disease. 40 Benamer TS, Patterson J, Grosset DG, et al.
Mov Disord 2015;30(12):1591–1601. doi:10.1002/ Accurate differentiation of parkinsonism and
mds.26424. essential tremor using visual assessment of
[123I]-FP-CIT SPECT imaging: the [123I]-FP-CIT
27 Gelb DJ, Oliver E, Gilman S. Diagnostic criteria for
study group. Mov Disord 2000;15(3):503–510.
Parkinson’s disease. Arch Neurol 1999;56(1):
doi:10.1002/1531-8257(200005)15:3<503::AID-
33–39. doi:10.1001/archneur.56.1.33.
MDS1013>3.0.CO;2-V.
28 Hughes AJ, Daniel SE, Kilford L, Lees AJ. Accuracy
41 Benamer HT, Oertel WH, Patterson J, et al.
of clinical diagnosis of idiopathic Parkinson’s
Prospective study of presynaptic dopaminergic
disease: a clinico-pathological study of 100
imaging in patients with mild parkinsonism and
cases. J Neurol Neurosurg Psychiatry 1992;55(3):
tremor disorders: part 1. Baseline and 3-month
181–184. doi:10.1136/jnnp.55.3.181.
observations. Mov Disord 2003;18(9):977–984.
29 Gibb WR, Lees AJ. The relevance of the Lewy doi:10.1002/mds.10482.
body to the pathogenesis of idiopathic Parkinson’s
42 Asenbaum S, Pirker W, Angelberger P, et al. [123I]
disease. J Neurol Neurosurg Psychiatry 1988;51(6):
beta-CIT and SPECT in essential tremor and
745–752. doi:10.1136/jnnp.51.6.745.
Parkinson’s disease. J Neural Transm (Vienna)
30 Berg D, Lang AE, Postuma RB, et al. Changing the 1998;105(10–12):1213–1228. doi:10.1007/
research criteria for the diagnosis of Parkinson’s s007020050124.
disease: obstacles and opportunities. Lancet
43 Jennings DL, Seibyl JP, Oakes D, et al. (123I) beta-
Neurol 2013;12(5):514–524. doi:10.1016/S1474-
CIT and single-photon emission computed
4422(13)70047-4.
tomographic imaging vs clinical evaluation in
31 Stern MB, Lang A, Poewe W. Toward a redefinition Parkinsonian syndrome: unmasking an early
of Parkinson’s disease. Mov Disord 2012;27(1): diagnosis. Arch Neurol 2004;61(8):1224–1229.
54–60. doi:10.1002/mds.24051. doi:10.1001/archneur.61.8.1224.
32 Kempster PA, Williams DR, Selikhova M, et al. 44 Pagano G. Niccolini F, Politis M. Imaging in
Patterns of levodopa response in Parkinson's Parkinson’s disease. Clin Med (Lond) 2016;16(4):
disease: a clinico-pathological study. Brain 371–375. doi:10.7861/clinmedicine.
2007;130(pt 8):2123–2128. doi:10.1093/brain/
45 Fahn S, Elton RL, UPDRS program members.
awm142.
Unified Parkinson’s disease rating scale. In: Fahn S,
33 McFarland NR, Hess CW. Recognizing atypical Marsden CD, Goldstein M, Calne DB, editors.
Parkinsonisms: “Red Flags” and therapeutic Recent developments in Parkinson’s disease,
approaches. Semin Neurol 2017;37(2):215–227. vol. 2. Florham Park, NJ: Macmillan Healthcare
doi:10.1055/s-0037-1602422. Information, 1987:153–163, 293–304.
CONTINUUMJOURNAL.COM 917
71 Benabid AL, Pollak P, Seigneuret E, et al. 73 Bari AA, Thum J, Babayan D, Lozano AM. Current
Chronic VIM thalamic stimulation in Parkinson’s and expected advances in deep brain stimulation
disease, essential tremor and extra-pyramidal for movement disorders. Prog Neurol Surg 2018;
dyskinesias. Acta Neurochir Suppl (Wien) 1993;58: 33:222–229. doi:10.1159/000481106.
39–44. doi:10.1007/978-3-7091-9297-9_8.
74 Schuepbach WM, Rau J, Knudsen K, et al.
72 DeLong MR, Huang KT, Gallis J, et al. Effect of Neurostimulation for Parkinson’s disease
advancing age on outcomes of deep brain with early motor complications. N Engl J Med
stimulation for Parkinson disease. JAMA Neurol 2013;368(7):610–622. doi:10.1056/
2014;71(10):1290–1295. doi:10.1001/jamaneurol. NEJMoa1205158.
2014.1272.
Palsy, Corticobasal C O N T I N U UM A U D I O
I NT E R V I E W A V A I L AB L E
ONLINE
Degeneration, and
VIDEO CONTENT
By Paul Greene, MD
ABSTRACT
PURPOSE OF REVIEW: Patients who have parkinsonian features, especially
Downloaded from http://journals.lww.com/continuum by BhDMf5ePHKbH4TTImqenVJ8WlwlLSkxgQL8l5tAGR0ztQKfE9dfXzUa0Fmli1DHP on 08/03/2019
without tremor, that are not responsive to levodopa, usually have one of
these three major neurodegenerative disorders rather than Parkinson
disease: progressive supranuclear palsy (PSP), multiple system atrophy
(MSA), or corticobasal degeneration (CBD). Each of these disorders
eventually develops signs and symptoms that distinguish it from idiopathic
Parkinson disease, but these may not be present at disease onset.
Although these conditions are not generally treatable, it is still important to
correctly diagnose the condition as soon as possible.
RECENT FINDINGS: In recent years, it has been increasingly recognized that the
symptoms of these diseases do not accurately predict the pathology, and the
pathology does not accurately predict the clinical syndrome. Despite this,
interest has grown in treating these diseases by targeting misfolded tau (in the
case of PSP and CBD) and misfolded α-synuclein (in the case of MSA). CITE AS:
CONTINUUM (MINNEAP MINN)
2019;25(4, MOVEMENT DISORDERS):
SUMMARY: Knowledge of the characteristic signs and symptoms of PSP, 919–935.
MSA, and CBD are essential in diagnosing and managing patients who have
atypical parkinsonian syndromes. Address correspondence to
Dr Paul Greene, Mt. Sinai School
of Medicine, 5 E 98th St,
New York, NY 10029,
[email protected].
INTRODUCTION RELATIONSHIP DISCLOSURE:
A
fter the introduction of levodopa to treat Parkinson disease (PD) in Dr Greene reports no disclosure.
the late 1960s, the pathologies and clinical pictures of dopamine
UNLABELED USE OF
deficiency syndromes broadened dramatically. Clinicians identified PRODUCTS/INVESTIGATIONAL
patients who had signs of dopamine deficiency (masked facies, USE DISCLOSURE:
Dr Greene discusses the
hypophonia, rigidity, slowness of movement and gait, loss of
unlabeled/investigational use of
postural reflexes, and sometimes resting tremor and other features) but, unlike medications for the treatment
patients with PD, had minimal or no improvement with levodopa. As a group, of atypical parkinsonism, none
of which have been approved
these patients are said to have atypical parkinsonism. Today, the most common by the US Food and Drug
of these disorders are progressive supranuclear palsy (PSP), followed by multiple Administration.
system atrophy (MSA) and corticobasal degeneration (CBD). The major problem,
especially for PSP and CBD, is that patients with typical clinical signs and © 2019 American Academy
symptoms of the disease may have very different pathologies, and patients of Neurology.
CONTINUUMJOURNAL.COM 919
with the same pathology may have different clinical signs and symptoms.
Unfortunately, no definitive diagnostic tests are easily available to distinguish
these diseases from each other during life. The gold standard diagnostic test is
autopsy, but autopsies are highly selected and may lead to conclusions that do
not represent the entire population of a disease.
The history of MSA extends back at least to 1900.1 In 1900, Dejerine and
Thomas2 described cases of late-onset ataxia and parkinsonism, later termed
olivopontocerebellar atrophy. In 1960, Shy and Drager3 presented two cases of
men aged 39 and 49 who developed impotence and urinary disturbance followed
by multiple autonomic deficits, parkinsonism, ataxia, and, in one case, lower
motor neuron disease, which was referred to as Shy-Drager syndrome.3 In 1960
and 1961, Adams, van Bogaert, and van der Eecken4,5 reported four cases of
patients with gliosis of the striatum and degeneration of the substantia nigra who
had rapidly progressive parkinsonism but also autonomic failure and cerebellar
deficits, which they referred to as striatonigral degeneration. Similar cases had
been reported before these three reports, but these reports were the first to
identify each of these conditions as a syndrome. In 1969, Graham and
Oppenheimer6 reviewed the published literature on patients with autonomic
failure and either clinical symptoms or pathology indicating cerebellar and
striatonigral involvement. They felt these syndromes overlapped so much that
they should all be labeled MSA.6 In 1998, Lantos1 described α-synuclein deposits
in glia (glial cytoplasmic inclusions) in all three conditions, making it even more
likely they were all part of the same spectrum.
In 1964, Steele, Richardson, and Olszewski7 coined the term progressive
supranuclear palsy (PSP) after reporting on patients with parkinsonism, loss of
postural reflexes, supranuclear gaze palsy, axial rigidity, pseudobulbar state, and
dementia who had midbrain and pontine degeneration as well as neuronal loss
in the substantia nigra.
In 1968, Rebeiz and colleagues8 reported on three patients with asymmetric
parkinsonism, apraxia, involuntary elevation of upper and lower extremities,
and posturing, which would probably now be called dystonia, and a unique
pathology involving the cortex as well as the basal ganglia and cerebellum and
with neuronal achromasia. They referred to this as corticodentatonigral
degeneration with neuronal achromasia, which was later shortened to
corticobasal degeneration (CBD).
Although PSP, CBD, and MSA are difficult to accurately clinically diagnose at
this time, this article presents the signs and symptoms of these conditions based
primarily on clinical experience. Although relatively large series of patients with
these conditions have been published based on autopsy, this article bases descriptions
primarily on clinical experience. The reason for this choice is that autopsy series have
two major shortcomings: (1) autopsies are rarely based on random or consecutive
selection, leading to referral bias that is impossible to quantify and (2) the history and
physical examinations in most autopsy series are not performed systematically
by movement disorder specialists and sometimes not even by neurologists.
For more information on clinical features of autopsy-documented cases of
PSP, CBD, and MSA, refer to Respondek and colleagues,9 Armstrong and
colleagues,10 and Wenning and colleagues,11 respectively.
CONTINUUMJOURNAL.COM 921
COMMENT This case shows a typical presentation and examination for the classic
presentation of progressive supranuclear palsy (PSP) (PSP–Richardson
syndrome). The supranuclear gaze palsy developed fairly early, making the
diagnosis easier, but some patients with PSP may not develop eye
movement abnormalities until late in the course. The early loss of postural
stability, growling voice, furrowed brow, and apraxia of eyelid opening
were important clues to the correct diagnosis.
CORTICOBASAL DEGENERATION
The classic presentation of CBD combines markedly asymmetric rigidity and
bradykinesia with focal or hemidystonia and cortical deficits. Most patients
develop symptoms in their fifties to seventies.
CONTINUUMJOURNAL.COM 923
The patient’s dystonia may make it difficult to assess apraxia. Speech is hesitant
and dysarthric, and aphasia and apraxia of speech occur. Other cortical signs
may include apraxia (difficulty performing motor activities despite normal
understanding and normal sensory/motor systems); cortical sensory loss
(eg, two-point discrimination, agraphesthesia [difficulty recognizing letters/numbers
written on the skin with eyes closed] and astereognosis [difficulty recognizing
objects by touch alone]); cortical myoclonus (spontaneous or reflex, or both),
which can be diagnosed accurately only with electrophysiology but usually
consists of multifocal, very rapid shocklike jerks; alien limb phenomenon
Pathology ● As in progressive
supranuclear palsy, the
The pathology of CBD consists of asymmetric cortical atrophy usually in the
pathology of corticobasal
frontoparietal region with relative sparing of the occipital lobes. Swollen degeneration also involves
vacuolated neurons are found in the atrophic cortical areas and to a lesser widespread deposition of
degree in the affected subcortical regions. These ballooned neurons contain 4-repeat tau but also
includes asymmetric
phosphorylated neurofilaments and sometimes tau and ubiquitin. As with
cortical atrophy and
PSP, widespread neuronal loss and gliosis is seen, not just in the affected neuronal, oligodendroglial,
cortex but also in the globus pallidus, putamen, red nucleus, thalamus, and astrocytic deposits
subthalamic nucleus, substantia nigra, locus coeruleus, and, to a lesser degree, distinct from the deposits in
in the dentate nucleus. Remaining neurons in affected areas contain progressive supranuclear
palsy.
inclusions: globose tangles called corticobasal bodies and tau fibrils around
nuclei of oligodendroglia called coiled bodies. Unlike the tufted astrocytes in ● As with progressive
PSP, the typical glial findings are tau-containing processes surrounding supranuclear palsy, multiple
astrocytes called astrocytic plaques. As in PSP, 4-repeat tau predominates, but pathologies may mimic the
signs and symptoms of
the insoluble tau fragments in CBD have a different ultrastructure from the corticobasal degeneration,
insoluble fragments of tau in PSP.17 including progressive
As with PSP, the pathology of CBD may also present as multiple other supranuclear palsy,
syndromes: a classic PSP syndrome, primary nonfluent aphasia, a frontal Alzheimer disease, Pick
disease, and Creutzfeldt-
dementia with executive dysfunction and behavioral change, pure Alzheimerlike
Jakob disease. When this
dementia or, rarely, idiopathic PD.10,17 Also, as in PSP, a variety of other happens, it is known as
pathologies can mimic the clinical features of CBD, known as corticobasal corticobasal syndrome.
syndrome (CBS). These include the pathologies of PSP, Alzheimer disease, Pick Similarly, the pathology of
corticobasal degeneration
disease, frontotemporal lobar degeneration with ubiquitin– and TDP-43–positive
may present as progressive
inclusions, dementia with Lewy bodies, frontotemporal lobar degeneration supranuclear palsy, primary
with fused-in-sarcoma–positive inclusions, and Creutzfeldt-Jakob disease.18 nonfluent aphasia,
Consensus criteria have been proposed for the clinical diagnosis of CBD, but Alzheimer disease, and
the criteria have proven neither sensitive nor specific (sensitivity was 68.4%, other conditions.
specificity was described as low but percentage was not given).10,19 VIDEO 2-1
(links.lww.com/CONT/A56) and VIDEO 2-2 (links.lww.com/CONT/A204) contain
examples of patients with CBS and CBD, respectively.20,21
Treatment
As with PSP, the current attempts at treating CBD itself as opposed to treating
the symptoms are focused on detoxifying misfolded tau.22
CONTINUUMJOURNAL.COM 925
COMMENT This patient has multiple system atrophy with predominant parkinsonism
(MSA-P), although his initial symptoms were autonomic dysfunction. His
orthostasis was severe, but this can be seen in dementia with Lewy bodies
as well. The stridor was a major clue that the diagnosis was MSA. In
addition, he also had some signs of cerebellar dysfunction: sustained
nystagmus, broad-based gait, and falling to the side as well as forward and
backward. Stridor in patients with MSA may respond to levodopa even
when other motor manifestations do not.
Pathology
Classically, the pathology of MSA consists of widespread neuronal loss and atrophy
including striatonigral, cerebellar, autonomic, and corticospinal pathways. These
can involve the substantia nigra, globus pallidus, parts of the cerebellum, middle
cerebellar peduncle, inferior olives, intermediolateral cell columns, corticospinal
tracts, anterior horn cells, and other structures. Unlike in PSP, the subthalamic
nucleus, dentate nucleus, and superior cerebellar peduncle are relatively uninvolved.
CONTINUUMJOURNAL.COM 927
Improvement with Very small subgroup, usually Rare Small subgroup (multiple system
levodopa modest and short lived atrophy with predominant
parkinsonism), usually modest
Resting tremor Very uncommon (may occur in Probably very uncommon Very uncommon but possible
progressive supranuclear palsy (hard to separate from
with predominant parkinsonism myoclonus)
or progressive supranuclear
palsy–Richardson syndrome)
Supranuclear gaze Common; early in many but not all Occurs in small subgroup Very uncommon but possible
palsy patients
Axial rigidity Marked axial rigidity, especially Common, often not Common, often not dramatic
in progressive supranuclear dramatic
palsy–Richardson syndrome
Cortical deficits Very small subgroup (progressive Very common Very uncommon
(eg, apraxia, agnosia, supranuclear palsy–corticobasal
astereognosis) syndrome)
Ataxia/cerebellar Gait ataxia is common, limb ataxia Rare Cerebellar signs sometime
deficits rarely reported during the course are common
Autonomic failure Some features common Same as progressive Multiple failures common
orthostasis (eg, impotence, constipation, supranuclear palsy (eg, urinary retention, sweating)
urinary urgency), but orthostasis
uncommon
Inspiratory stridor Very rare Very rare Small but significant minority
CONTINUUMJOURNAL.COM 929
affect the tau protein) and for MSA (in the gene COQ2) but the significance of
these findings is not clear.31
Progressive Widespread neuronal loss and atrophy of the Neurofibrillary tangles in neurons in the involved
supranuclear midbrain, substantia nigra, subthalamic nucleus, areas consisting of mainly abnormally
palsy globus pallidus, dentate nucleus, superior phosphorylated 4-repeat tau; tau inclusions in
cerebellar peduncle, and multiple areas of the astrocytes consisting of fibrils in a tuft
frontal cortex; the particular frontal areas involved configuration (tufted astrocytes); tau inclusions in
depend on the subtype of progressive oligodendroglia that form perinuclear fibers
supranuclear palsy (coiled bodies); tau-containing threadlike
structures in the white matter (neuropil threads)
Corticobasal Widespread neuronal loss and atrophy Swollen, vacuolated neurons containing
degeneration asymmetrically in the cortex, usually maximal in the phosphorylated neurofilaments and sometimes
frontoparietal areas and much less involvement of 4-repeat tau are found in affected areas, more
the occipital lobes; widespread neuronal loss and commonly in cortical areas, which are common in
gliosis in the globus pallidus, putamen, red nucleus, corticobasal degeneration but are not specific for
thalamus, subthalamic nucleus, substantia nigra, that disease; the 4-repeat tau has a different
locus coeruleus, and dentate nucleus structure from the 4-repeat tau typical of
progressive supranuclear palsy; tau inclusions in
astrocytes (astrocytic plaques) are more diffuse
than the tufted astrocytes of progressive
supranuclear palsy; tau-containing threads
(neuropil threads) in gray and white matter of the
cortex in corticobasal degeneration
Multiple system Widespread neuronal loss and atrophy in the Abnormal inclusions in multiple system atrophy
atrophy substantia nigra, globus pallidus, putamen, contain abnormal α-synuclein, not tau; the
thalamus, parts of the cerebellum, middle characteristic inclusion is the glial cytoplasmic
cerebellar peduncle, inferior olives, red nucleus, inclusion; there are less frequent glial nuclear
intermediolateral cell columns, corticospinal tracts, inclusions containing α-synuclein and α-synuclein-
anterior horn cells, and other structures; containing neuronal cytoplasmic inclusions (similar
subthalamic nucleus, dentate nucleus, and superior in structure to the glial cytoplasmic inclusions) and
cerebellar peduncle are relatively uninvolved neuronal nuclear inclusions (consisting of
networks of fibrils) as well as dystrophic neurites
in involved structures
PSP but does not distinguish PSP from PD, CBD, or MSA and may not be
sensitive for all subtypes.14
The most important clinical clues to the diagnosis of CBD/CBS are focal
cortical deficits such as myoclonus, apraxia, and aphasia. However, these can
occur in a minority of patients with PSP and other conditions.
It is currently very difficult to identify MSA in patients whose MSA begins
with isolated cerebellar signs or autonomic failure. Fluorodopa PET can
accurately identify patients with unsuspected dopaminergic deficiency,
but the test is currently not easily
available. Brain MRI in patients with
MSA may show hyperintensity in the
dorsolateral margin of the putamen
(putaminal slit sign) (FIGURE 2-234)
and cruciform increased signal in the
pons (hot cross bun sign) (FIGURE 2-3),
but the sensitivity of these tests, while
unknown, is probably low.26,35 If
orthostasis is an early feature in a patient
with parkinsonism, the diagnosis may be
either MSA or diffuse Lewy body disease.
In MSA, the central preganglionic
sympathetic neurons are mostly affected
(plasma epinephrine is normal when the
patient is supine but fails to rise when
FIGURE 2-2
the patient stands), and this may
Putaminal slit sign. Axial T2-weighted
differentiate it from diffuse Lewy MRI of a patient with autopsy-
body disease, where the autonomic confirmed multiple system atrophy
impairment affects mostly peripheral with predominant parkinsonism
postganglionic neurons. showing slitlike hyperintensity in the
dorsolateral putamen (arrows).
Moderate to marked cognitive impairment Reprinted with permission from Horimoto Y,
is common is PSP and CBD but uncommon et al, J Neurol.34 © 2002 Steinkopff Verlag.
CONTINUUMJOURNAL.COM 931
CONCLUSION
PSP, CBD, and MSA were first identified by pathology that involved the
substantia nigra but, unlike PD, also involve other cortical and subcortical
VIDEO LEGENDS
VIDEO 2-1 VIDEO 2-3
Corticobasal syndrome. Video shows a 75-year- Multiple system atrophy. Video shows a man with
old woman clinically diagnosed with corticobasal signs of a mild cerebellar syndrome. On initial
syndrome. Among other features, she illustrates an presentation (not shown), he had a hint of facial
asymmetric parkinsonism with a markedly dystonic masking, decreased arm swing, and some dystonic
right arm, myoclonus, ideomotor apraxia, and posturing with his right arm when walking. He
cortical sensory loss. developed bowel and bladder dysfunction, erectile
links.lww.com/CONT/A56 dysfunction, temperature dysregulation, and a
cerebellar syndrome (primarily scanning speech and
Reproduced with permission from Williams DR, ataxia). Video shows mild flattening of the left
Litvan I, Continuum (Minneap Minn).20 © 2013 nasolabial fold, depression of the left corner of the
American Academy of Neurology. mouth and, at times, a widened left palpebral fissure.
VIDEO 2-2 Video also shows his mildly broad-based stance and
Corticobasal degeneration. Video shows an gait and his difficulty standing with feet together and
80-year-old man demonstrating progressive eyes closed. When walking, occasionally either foot
asymmetric limb dysfunction, rigidity, will be placed either laterally or medially, especially
bradykinesia, dystonia, apraxia, and cortical when walking quickly. His right arm tends to be flexed,
sensory deficits consistent with probable and his stride and arm swing are reduced on the right
corticobasal degeneration. (most obviously when walking quickly). When pulled
links.lww.com/CONT/A204 backward, he moves his feet quickly to recover, which
may throw him off balance. The patient later
Reproduced with permission from McFarland NR, developed clear signs of parkinsonism (not shown) that
Continuum (Minneap Minn).21 © 2016 American were not responsive to levodopa, and he did not
Academy of Neurology. develop orthostasis until late in the disease course.
links.lww.com/CONT/A283
© 2019 American Academy of Neurology.
CONTINUUMJOURNAL.COM 933
REFERENCES
1 Lantos PL. The definition of multiple system 14 Armstrong MJ. Progressive supranuclear palsy:
atrophy: a review of recent developments. an update. Curr Neurol Neurosci Rep 2018;18(3):
J Neuropathol Exp Neurol 1998;57(12):1099–1111. 12. doi:10.1007/s11910-018-0819-5.
2 Dejerine J, Thomas AA. L’atrophie olivo-ponto- 15 Dickson DW, Ahmed Z, Algom AA, et al.
cerebelleuse. Nouv Iconog Salpetriere 1900;13: Neuropathology of variants of progressive
330–370. supranuclear palsy. Curr Opinion Neurol 2010;
23(4):394–400. doi:10.1097/WCO.0b013e32833be924.
3 Shy GM, Drager GA. A neurologic syndrome
associated with orthostatic hypotension. Arch 16 Ayers JI, Giasson BI, Borchelt DR. Prion-like
Neurol 1960;2:511–527. doi: 10.1001/archneur. spreading in tauopathies. Biol Psychiatry 2018;
1960.03840110025004. 83(4):337–346. doi:10.1016/j.biopsych.2017.04.003.
4 Van der Eecken H, Adams RD, van Bogaert L. 17 Kouri N, Whitwell JL, Josephs KA, et al.
Striopallidal-nigral degeneration. An hitherto Corticobasal degeneration: a pathologically
undescribed lesion in paralysis agitans. distinct 4R tauopathy. Nat Rev Neurol 2011;7(5):
J Neuropathol Exp Neurol 1960;19:159–161. 263–272. doi:10.1038/nrneurol.2011.43.
5 Adams RD, van Bogaert L, van der Eecken H. 18 Boeve BF. The multiple phenotypes of corticobasal
Nigro-striate and cerebello-nigro-striate syndrome and corticobasal degeneration:
degeneration. (Clinical uniqueness and pathological implications for further study. J Mol Neurosci
variability of presenile degeneration of the 2011;45(3):350–353. doi:10.1007/s12031-011-9624-1.
extrapyramidal rigidity type) [in French]. Psychiat
19 Alexander SK, Rittman T, Xuereb JH, et al. Validation
Neurol (Basel) 1961;142:219–259.
of the new consensus criteria for the diagnosis of
6 Graham JG, Oppenheimer DR. Orthostatic corticobasal degeneration. J Neurol Neurosurg
hypotension and nicotine sensitivity in a case Psychiatry 2014;85(8):923–927. doi:10.1136/
of multiple system atrophy. J Neurol Neurosurg jnnp-2013-307035.
Psychiatry 1969;32(1):28–34.
20 Williams DR, Litvan I. Parkinsonian syndromes.
7 Steele JC, Richardson JC, Olszewski J. Progressive Continuum (Minneap Minn) 2013;19(5, Movement
supranuclear palsy. A heterogeneous degeneration Disorders):1189–1212. doi:10.1212/01.CON.
involving the brain stem, basal ganglia and 0000436152.24038.e0.
cerebellum with vertical gaze and pseudobulbar
21 McFarland NR. Diagnostic approach to atypical
palsy, nuchal dystonia and dementia. Arch
parkinsonian syndromes. (Minneap Minn) 2016;
Neurol 1964;10:333–359. doi:10.1001/archneur.
22(4, Movement Disorders):1117–1142. doi: 10.1212/
1964.00460160003001.
CON.0000000000000348.
8 Rebeiz JJ, Kolodny EH, Richardson EP Jr.
22 Ali F, Josephs KA. Corticobasal degeneration: key
Corticodentatonigral degeneration with neuronal
emerging issues. J Neurol 2018;265(2):439–445.
achromasia. Arch Neurol 1968;18(1):20–33.
doi:10.1007/s00415-017-8644-3.
doi:10.1001/archneur.1968.00470310034003.
23 Palma JA, Norcliffe-Kaufmann L, Kaufmann H.
9 Respondek G, Stamelou M, Kurz C, et al.
Diagnosis of multiple system atrophy. Auton
The phenotypic spectrum of progressive
Neurosci 2018;211:15–25. doi:10.1016/j.autneu.
supranuclear palsy: a retrospective multicenter
2017.10.007.
study of 100 definite cases. Mov Disord 2014;
29(14):758–766. doi:10.1002/mds.26054. 24 Sima AA, Caplan M, D’Amato CJ, et al. Fulminant
multiple system atrophy in a young adult presenting
10 Armstrong MJ, Litvan I, Lang AE, et al. Criteria for
as motor neuron disease. Neurology 1993;43(10):
the diagnosis of corticobasal degeneration.
2031–2035. doi:10.1212/WNL.43.10.2031.
Neurology 2013;80(5):496–503. doi:10.1212/WNL.
0b013e31827f0fd1. 25 Koga A, Dickson DW. Recent advances in
neuropathology, biomarkers and therapeutic
11 Wenning GK, Ben Shlomo Y, Magalhães M. Clinical
approach of multiple system atrophy. J Neurol
features and natural history of multiple system
Neurosurg Psychiatry 2018;89(2):175–184. doi:10.1136/
atrophy. An analysis of 100 cases. Brain 1994;
jnnp-2017-315813.
117(pt 4):835–845. doi: 10.1093/brain/117.4.835.
26 Gilman S, Wenning GK, Low PA, et al. Second
12 Höglinger GU, Respondek G, Stamelou M, et al.
consensus statement on the diagnosis of
Clinical diagnosis of progressive supranuclear
multiple system atrophy. Neurology 2008;71(9):
palsy: the Movement Disorder Society criteria.
670–676. doi:10.1212/01.
Mov Disord 2017;32(6):853–864. doi:10.1002/
wnl.0000324625.00404.15.
mds.26987.
27 Erkkinen MG, Kim MO, Geschwind MD. Clinical
13 Esteban A, Giménez-Roldán S. Involuntary closure
neurology and epidemiology of the major
of eyelids in parkinsonism. Electrophysiological
neurodegenerative diseases. Cold Spring Harb
evidence for prolonged inhibition of the levator
Perspect Biol 2018;10(4):a033118. doi:10.1101/
palpebrae muscles. J Neurol Sci 1988;85(3):
cshperspect.a033118.
333–345. doi:10.1016/0022-510X(88)90191-8.
CONTINUUMJOURNAL.COM 935
Tics and Tourette
C O N T I N UU M A UD I O
I NT E R V I E W A V AI L A B L E
ONLINE
Syndrome
By Harvey S. Singer, MD, FAAN
VIDEO CONTENT
A VA I L A B L E O N L I N E
ABSTRACT
PURPOSE OF REVIEW: The purpose of this article is to present current
information on the phenomenology, epidemiology, comorbidities, and
pathophysiology of tic disorders and discuss therapy options. It is hoped
that a greater understanding of each of these components will provide
Downloaded from http://journals.lww.com/continuum by BhDMf5ePHKbH4TTImqenVLIlg0cBmue4T5mm7WKE1WkwapLJtCp3kAo1PIu4JaP0YSZXFg1EEvs= on 08/03/2019
CITE AS: RECENT FINDINGS: Recent advances include the finding that Tourette
CONTINUUM (MINNEAP MINN)
2019;25(4, MOVEMENT DISORDERS):
syndrome is likely due to a combination of several different genes, both
936–958. low-effect and larger-effect variants, plus environmental factors.
Pathophysiologically, increasing evidence supports involvement of the
Address correspondence to cortical–basal ganglia–thalamocortical circuit; however, the primary
Dr Harvey S. Singer, Professor of
Neurology and Pediatrics, Johns location and neurotransmitter remain controversial. Behavioral therapy
Hopkins Hospital, Rubenstein is first-line treatment, and pharmacotherapy is based on tic severity.
Child Health Building, 200 N
Several newer therapeutic agents are under investigation (eg, valbenazine,
Wolfe St, Ste 2141 Baltimore,
MD 21287, [email protected]. deutetrabenazine, cannabinoids), and deep brain stimulation is a
promising therapy.
RELATIONSHIP DISCLOSURE:
Dr Singer serves as a consultant
for Abide Therapeutics, Inc; Cello SUMMARY: Tics, defined as sudden, rapid, recurrent, nonrhythmic motor
Health BioConsulting; ClearView movements or vocalizations, are essential components of Tourette
Healthcare Partners; Teva
Pharmaceutical Industries Ltd; syndrome. Although some tics may be mild, others can cause significant
and Trinity Partners, LLC. psychosocial, physical, and functional difficulties that affect daily
Dr Singer receives publishing activities. In addition to tics, most affected individuals have coexisting
royalties from Elsevier and
research/grant support from the neuropsychological difficulties (attention deficit hyperactivity disorder,
Tourette Association of America. obsessive-compulsive disorder, anxiety, mood disorder, disruptive
UNLABELED USE OF
behaviors, schizotypal traits, suicidal behavior, personality disorder,
PRODUCTS/INVESTIGATIONAL antisocial activities, and sleep disorders) that can further impact social and
USE DISCLOSURE: academic activities or employment.
Dr Singer discusses the
unlabeled/investigational use of
baclofen, botulinum toxin,
cannabidiol, clonazepam,
clonidine, deutetrabenazine, INTRODUCTION
T
ecopipam, fluphenazine,
ourette syndrome, also known as Gilles de la Tourette syndrome or
guanfacine, nabiximols,
risperidone, sulpiride, Tourette disorder, is a complex, childhood-onset neuropsychiatric
tetrabenazine, Δ-9- condition that includes multiple phenotypic motor and vocal tics. The
tetrahydrocannibinol, tiapride,
topiramate, and valbenazine.
condition is named after the French physician Georges Gilles de la
Tourette, who in 1885 reported nine patients with the “maladie of
tics.”1 Before Tourette’s report, motor and vocal tics had been reported in the
© 2019 American Academy context of witchcraft, in well-known historical individuals, and in the medical
of Neurology. literature by Jean Itard in 1825.2
TIC PHENOMENOLOGY
Tics are sudden, rapid, recurrent, nonrhythmic motor movements or
vocalizations (phonic productions).
CONTINUUMJOURNAL.COM 937
Characteristics
Tics have several characteristics that are useful in identifying their presence,
including precipitating factors, a waxing and waning pattern, admixture of new
and old tics, a premonitory urge that resolves when the tic is done, reduction
when engrossed, and variable severity. They can range from infrequent and
unnoticed to very frequent, intense, intrusive, and even self-injurious. Tics are
exacerbated by stress, anxiety, excitement, anger, fatigue, elevated temperatures,
or infections.9,10 These conditions, however, do not account for more prolonged
changes in tic severity.11 Tics are reduced when the individual is concentrating on
a physical or mental task or sleeping. Although most parents and patients report
that tics do not occur while sleeping, polysomnograms have identified their
presence in all phases of sleep.12 About 90% of adults13 and 37% of children14
report a premonitory urge/sensation just before a motor or phonic tic. Vaguely
defined as an urge, mounting tension, pressure, itch, or feeling, it is generally
localized to the region of the tic and resolves when the tic is permitted to occur.15
Many individuals can briefly suppress their tics, although this effort may
trigger an exacerbation of premonitory sensations or a sense of increased
internal tension.
Tic disorders are more common in males than in females (ratio of 3:1 to 4:1)
and usually begin between the ages of 4 and 8. Motor tics usually precede vocal
tics, with initial simple motor tics involving the face, head, or neck. Tics can be
highly variable and fluctuate, and an individual’s tic repertoire evolves over time.
Statistically, the maximum severity of tics tends to occur between 8 and 12 years
of age,16 with tics declining in severity throughout the teenaged to early
adulthood years.16,17 An approximate rule of thirds suggests that one-third of tics
disappear, one-third improve, and one-third continue to fluctuate.18 Although
many adults have reported the resolution of tics, formal assessments have
indicated otherwise.19 Assumptions that adult tic severity can be predicted by
childhood tic severity, childhood fine motor skills, and reduced childhood
MRI caudate volumes require additional confirmation. Tics can persist into
adulthood and, for some, can be severe and debilitating.20 In addition,
symptom worsening has occurred in adults after prolonged periods of clinical
remission.21 Adult-onset tic disorders (formally, other specified tic disorder
with onset after age 18 years) have been reported and often include severe
symptoms, greater social morbidity, and poorer response to medications
compared to childhood-onset tic disorders.22
Neurologic examination and neuroimaging studies are typically normal.
“Soft” neurologic findings may include coordination issues, synkinesis
(involuntary muscular movements accompanying voluntary movements), and
motor restlessness, especially in individuals with ADHD.
DIAGNOSIS
The diagnosis of a tic disorder is based on historical features and observation of the
tics; there is no definitive diagnostic laboratory test. Tics must be differentiated
from drug-induced movements (akathisia, dystonia, parkinsonism), obsessive-
compulsive behaviors, hyperactivity, antisocial behaviors, and stereotypies.23
In contrast to tics, which are commonly associated with a premonitory urge,
u Provisional tic disorder designates an individual whose tics (motor or vocal) have been
present for less than 1 year since first tic onset, and onset is before age 18 years. The
disturbance cannot be attributable to the psychological effects of a substance or other
medical conditions. For example, tics can result as a direct consequence of a variety
of neurodegenerative disorders (eg, neuroacanthocytosis, Huntington disease,
neurodegeneration with brain iron accumulation), neurocutaneous syndromes,
and Creutzfeldt-Jakob disease.3,28 Tics have also been reported in association with
infections, Sydenham chorea, toxins (carbon monoxide), stroke, head and peripheral
trauma, and surgery. Drugs reported to induce tics include cocaine, lamotrigine, and
neuroleptics.29 Significant data exist refuting the concept that stimulant medications
are precipitating agents for tics.30
u Chronic motor or vocal tic disorder indicates the presence of either motor or vocal tics, but
not both, for longer than 1 year, without regard for tic frequency. Onset is before 18 years
of age, and tics cannot be attributed to the use of drugs or other medical condition. Note
that multiple characteristics overlap in individuals with chronic motor tic disorder and
individuals with Tourette syndrome.31
u Tourette disorder is also called Tourette syndrome, and both have very similar formal
criteria,27,32 except that when originally proposed, the age of onset for Tourette syndrome
was before 21 years of age as compared to before 18 years of age for Tourette disorder.
Other criteria include the presence of multiple motor tics and at least one vocal tic, a
waxing and waning course, a duration of longer than 1 year, and tics that are neither
substance-induced nor due to a general medical condition.
The last two categories apply to tic disorders that cause clinical
distress/impairment but do not meet criteria for either of the aforementioned
three tic disorders or any neurodevelopmental disorder.
u Other specified tic disorder is a diagnosis used when the clinician chooses to
communicate the reason why the individual failed to meet the criteria for a tic disorder,
(eg, other specified tic disorder with an age of onset of older than 18 years).
u Unspecified tic disorder is a diagnosis used when the criteria for a tic disorder are not met,
and there is insufficient information to make a more specific diagnosis.
CONTINUUMJOURNAL.COM 939
EPIDEMIOLOGY
Simple tics are relatively common in childhood, with reports of prevalence
(the number of cases in the population at a given time) being 6% to 12% (range of
4% to 24%).36,37 For Tourette syndrome, which occurs worldwide with common
features in all cultures and races, prevalence estimates have been variable,
with estimates ranging from 0.3% to 1%. In two meta-analyses, prevalence in
children was 0.52%38 and 0.77%39 but increased to 1.06% when only boys were
considered.39 Another estimate is that an additional 1% to 3% of children and
adolescents have a mild unidentified form. The variations in reported prevalence
are believed to be associated with differences in assessment methods and
measures. Several studies have documented that Tourette syndrome is common
in children with autistic spectrum disorders40; however, no correlation with
the severity of autistic symptoms has been seen. Mortality rates are reportedly
higher in Tourette syndrome and chronic motor or vocal tic disorder, irrespective
of the presence of comorbidities.41 Parents of children with chronic tic disorders
have higher rates of psychiatric illnesses, greater in mothers than fathers,
although whether this is associated with parental stress or environmental,
genetic, or other factors is unclear.42
COMORBIDITIES
Most individuals with Tourette syndrome (an estimated 86% to 90%) have at
least one comorbid/coexisting neuropsychological problem.43 These additional
issues add an extra clinical burden, and, for some, the clinical impact may be
greater than that caused by tics. Health-related quality of life assessments
have shown that outcome is predicted by the presence of comorbidities,
such as ADHD and obsessive-compulsive disorder (OCD), rather than tic
severity.44,45 Coexisting neuropsychological issues add a significant additional
burden to patients with Tourette syndrome or chronic motor or vocal tic
disorder.44,46 It has also been suggested that less severe tic phenotypes have
lower rates of comorbidity.47 In a study examining parent attribution of
impairment in home activities, non–tic-related concerns were a greater
problem.48
Longitudinal studies in patients with Tourette syndrome have suggested a
decline in both ADHD and OCD during adolescence although other
psychopathologies persist.17 Recognizing the common presence and detrimental
CONTINUUMJOURNAL.COM 941
CASE 3-1 A 9-year-old boy presented for a neurologic consultation for tics. He had
developed eye-blinking tics at age 7 and subsequently gradually
developed a variety of other motor tics, including head turning, neck
stretching, and facial grimacing, and vocal tics that included throat
clearing and grunting sounds. Over the years, his tics had a waxing and
waning course. They were worse when he was stressed or fatigued.
Nothing clearly made them better, and they were not present during
sleep. He denied having a premonitory urge. At the time of evaluation,
motor tics were occurring approximately once per hour but at times more
frequently. The movements were not interfering with his daily activities.
Vocal tics occurred about once every 3 hours, were quieter than his
normal voice, and did not interfere with his communication. He had
received only a few comments in the academic setting, his tics were not
causing any physical issues nor disrupting his classroom, and he had been
on no prior tic-suppressing therapy.
The patient had a history of attention deficit hyperactivity disorder
since age 4 and was started on a stimulant medication (amphetamine) at
age 6 years and 9 months. He had no history of obsessive-compulsive
behaviors, anxiety, or mood issues. He did, however, have disruptive
behaviors, with yelling, crying out, and banging on objects, and was easily
angered when he did not get his way.
The patient was the product of an uncomplicated pregnancy and
delivery, his early development was normal, and his general health was
good. His family history was positive for childhood tics in his father. His
neurologic examination was normal other than the occasional tics that
were observed.
Academic Difficulties
Intellectual function is typically normal in Tourette syndrome; however, some
children have executive function issues, differences between performance and
verbal IQ testing, impairment of visual-perceptual achievement, and a reduction
of visual-motor skills.46 A variety of factors account for poor school performance
in children with Tourette syndrome, including disruptive tics, psychosocial
difficulties, ADHD, OCD, learning disabilities, and use of medications.51,65
Individuals with Tourette syndrome or chronic motor or vocal tic disorder are
more likely to academically underachieve, even after accounting for various
comorbidities.66
Sleep Disorders
Sleep disorders, including difficulties falling and staying asleep, restlessness,
arousals, and parasomnias, are common in Tourette syndrome.67,68 Sleep issues
have been associated with the presence of comorbidities such as ADHD, anxiety,
mood disorders, or OCD69; however, others have suggested that sleep problems
in Tourette syndrome are not fully explainable by these associated conditions.67
The effective management of sleep problems has been reported to improve tic
control in both severity and impact on life (CASE 3-1).70
ETIOLOGY
The precise etiology for Tourette syndrome remains undetermined; however,
there is strong agreement that it is associated with polygenic and environmental
vulnerability factors.
Genetics
Tourette syndrome is currently classified as a polygenic inherited disorder,
suggesting that a combination of a variety of genes (some common, some with
a low effect or rare, and others having a larger effect) and environmental
factors are all involved in its transmission.71–73 An inherited nature for Tourette
CONTINUUMJOURNAL.COM 943
PATHOPHYSIOLOGY
Pathophysiological models of Tourette syndrome typically include the disruption of
specific circuits involved in motor activity and their associated neurotransmitters.
Circuits
A series of parallel cortical–basal ganglia–thalamocortical circuits provide a
framework for understanding the pathophysiology of tics and associated
behaviors (FIGURE 3-1).
CONTINUUMJOURNAL.COM 945
FIGURE 3-1
Cortical–basal ganglia–thalamocortical circuit.
D1 = dopamine D1 receptor; D2 = dopamine D2 receptor; GPe = globus pallidus externus; GPi = globus
pallidus internus; Mthal = motor thalamus; MSN = medium-sized spiny neuron; NAC = nucleus accumbens;
SMA = supplementary motor area; SNpr = substantia nigra pars reticulata; STN = subthalamic nucleus.
internus/substantia nigra pars reticulata region, in turn, affect the firing of cells in
the thalamus that project to the cortex. In sum, the direct pathway facilitates motor
activity via disinhibition of thalamocortical neurons and the indirect pathway
reduces motor activity by increasing the inhibition of thalamocortical neurons.
OTHER. Preliminary imaging and animal model studies also provide support for
involvement of the amygdala, hippocampus, ventral striatum, thalamus, midbrain,
and cerebellum. A favored proposal suggests the presence of a complex circuit in
which a failure anywhere within the cortical–basal ganglia–thalamocortical circuit,
or even one involving inputs to the circuit, can lead to an aberrant message arriving
at the primary motor cortex and enabling the tic.84
Neurotransmitter Abnormality
Strong evidence supports involvement of cortical–basal ganglia–thalamocortical
circuits in the pathophysiology of tic disorders. Several neurotransmitters,
including dopamine, glutamate, GABA, serotonin, acetylcholine, norepinephrine,
endogenous cannabinoids, opioids, histamine, and adenosine, are active
participants within these circuits and may be dynamic factors in the
pathophysiology of tics. Hence, an additional area of controversy is whether a
specific neurotransmitter or combination of neurotransmitters is relevant in the
pathogenesis of tics. Several published reviews discuss the evidence supporting
individual neurotransmitter abnormalities in Tourette syndrome.84,85 In general,
data used to support a particular neurotransmitter hypothesis are derived from
clinical responses to specific classes of medications; genetic protocols; CSF, blood,
CONTINUUMJOURNAL.COM 947
TREATMENT
The establishment of an effective therapeutic plan requires the careful initial
assessment of tics, determining the presence of co-occurring issues, and clarifying
the resulting impairment of each issue. Further, it is essential to clarify whether
tics or associated problems, such as ADHD, OCD, anxiety, school problems, or
behavioral disorders, represent the greatest impairment.
The first step in treatment (FIGURE 3-2) is education of the patient, his/her
family, and the school or workplace about the diagnosis, its potential coexisting
issues, and indications for therapy. Tics have no cure and fluctuate, and supportive
FIGURE 3-2
Approaches to the treatment of tics.
VMAT-2 = vesicular monoamine transporter-2.
a
Approved by the US Food and Drug Administration for the treatment of tics.
b
Medications are under investigation.
c
Medications are not available in the United states.
and gradually increased as needed. Patients should be carefully followed and ● In general, a two-tiered
have periodic evaluations assessing medication efficacy, side effects, and the approach to the use of
requirement for continued therapy. Recognizing that treatment is symptomatic, pharmacotherapy is
if tics remain under good control for a significant period, a gradual taper of the recommended for treating
tics, with use of tier 1
medication during a nonstressful time should be considered. Currently, only medications for milder tics
pimozide, haloperidol, and aripiprazole have US Food and Drug Administration and use of tier 2 medications
(FDA) approval for use as tic-suppressing agents. The extent of supporting reserved for more difficult
evidence for medications has been reviewed, and individual drug selection is to control symptoms.
often based on physician experience.91
CONTINUUMJOURNAL.COM 949
Brain Stimulation
Repetitive transcranial magnetic stimulation is a noninvasive technique that
uses brief repetitive intense magnetic fields generated by a coil placed over the
scalp to produce an electromagnetic field in the underlying brain region. Several
small trials in Tourette syndrome using low-frequency inhibiting repetitive
transcranial magnetic stimulation (1 Hz) suggest that it can have a beneficial
effect, especially if the supplementary motor area is targeted.106 A single
double-blind randomized placebo trial, however, showed no significant
difference between active and sham treatment.107 A small study using cathodal
transcranial direct current stimulation was unsuccessful in two-thirds of patients
with severe tics.108
Deep brain stimulation is a stereotactic treatment that has significant potential
for the treatment of tics.109,110 The centromedian parafascicular complex of the
thalamus and anteromedial globus pallidus internus have been the most
commonly stimulated sites, but the optimal target has yet to be determined.
Although most reports describe a beneficial effect, interpretation is confounded
by variations in methodologies, differences in complications, variable use of
standard outcome measures, and the lack of a control population. Suggested
criteria for the use of deep brain stimulation in patients with tics include
the following:
u Disabling tics with a Yale Global Tic Severity Scale score of more than 35/50
u Failed behavioral therapy (CBIT)
u Failed medication trials from pharmacologic groups including α-adrenergic agonists
(guanfacine, clonidine) and dopamine antagonists (including one typical and one
atypical antipsychotic), plus one additional category (vesicular monoamine
transporter-2 inhibitors)
u Evaluation by a multidisciplinary team (eg, neurologist, neurosurgeon, psychiatrist,
neuropsychologist, deep brain stimulation programmer)
u Treated and stable comorbid conditions110
CONTINUUMJOURNAL.COM 951
Deep brain stimulation for patients younger than 18 years of age should have
additional institutional approval (CASE 3-2).
CONCLUSION
Tourette syndrome is a heterogeneous disorder with variable and fluctuating
motor and vocal tics as well as frequent co-occurring neuropsychiatric
CASE 3-2 A 19-year-old man was referred for a second opinion regarding the
treatment of Tourette syndrome. He was diagnosed with Tourette
syndrome at 8 years of age. His history included numerous intermittent
and fluctuating motor tics (ocular deviations, forceful head jerks,
shoulder shrugs, facial and body contortions, abdominal jerks, hitting,
jumping, and copropraxia) and vocal tics (coprolalia, echolalia, palilalia,
shouts, and screams).
Over the past several weeks, his tics had dramatically increased without
a clear precipitating event. Both motor and vocal tics were occurring every
few minutes. The motor tics were extremely forceful and complex and
included violent head flexion and extension movements that were causing
persistent neck discomfort. Recent x-rays of his neck were normal. His
vocal tics were very loud and interfered with his social activities. He had
recently left college and returned home because of his discomfort.
He was on 20 mg/d aripiprazole after clonidine, guanfacine, pimozide,
haloperidol, and risperidone had failed to control his tics. He was also on
sertraline and clonazepam for anxiety and obsessive-compulsive disorder.
On examination, he was extremely anxious and had frequent motor tics,
including very painful exaggerated head thrusts, and coprolalia.
Neurologic examination showed marked discomfort with neck flexion and
extension and cervical spine tenderness. Tone, strength, sensation, and
reflexes were normal.
VIDEO LEGENDS
VIDEO 3-1 VIDEO 3-4
Tourette syndrome. Video shows a boy with Dystonic, vocal, and holding (blocking) tics. Video
Tourette syndrome demonstrating frequent shows a boy with Tourette syndrome exhibiting a
blinking, alternating facial contractions, tongue dystonic extension of his neck associated with
protrusions, and mouth openings. repetitive expiratory grunting sounds. In addition, he
links.lww.com/CONT/A284 demonstrates a holding (blocking) tic, during which
time he stops breathing and is completely motionless.
Reproduced with permission from Singer HS.3 links.lww.com/CONT/A287
© 2016 Elsevier.
Reproduced with permission from Singer HS.3
VIDEO 3-2 © 2016 Elsevier.
Dystonic tics. Video shows a girl exhibiting facial
tics manifested by repetitive sustained contractions VIDEO 3-5
of facial muscles, resulting in grimacing and neck Complex motor and phonic tics. Video shows a
muscle contractions typical of dystonic tics. 17-year-old girl with severe Tourette syndrome
links.lww.com/CONT/A285 exhibiting complex motor tics and loud screaming
phonic tics.
Reproduced with permission from Singer HS.3 links.lww.com/CONT/A288
© 2016 Elsevier.
Reproduced with permission from Singer HS.3
VIDEO 3-3 © 2016 Elsevier.
Repetitive complex movements. Video shows a
boy with Tourette syndrome displaying repetitive, VIDEO 3-6
complex movements produced by contractions of Complex motor and phonic tics. Video shows a boy
his shoulder and trunk muscles, preceded by an with severe Tourette syndrome exhibiting complex
intense premonitory sensation of a chill. motor and loud phonic tics, including coprolalia.
links.lww.com/CONT/A286 links.lww.com/CONT/A289
Reproduced with permission from Singer HS.3 Reproduced with permission from Singer HS.3
© 2016 Elsevier. © 2016 Elsevier.
USEFUL WEBSITE
TOURETTE ASSOCIATION OF AMERICA
The Tourette Association of America provides
educational materials and information about
research efforts, support groups, and other
services to assist patients and their families
in coping with the problems associated with
Tourette syndrome.
tourette.org
CONTINUUMJOURNAL.COM 953
REFERENCES
1 Gilles dela Tourette G. Étude sur une affection 14 Banaschewski T, Woerner W, Rothenberger A.
nerveuse caractèrisée par de l’incoordination Premonitory sensory phenomena and suppressibility
motrice accompagnée d’écholalie et de coprolalie of tics in Tourette syndrome: developmental
(jumping, latah, myriachit). Paris: Aux bureaux du aspects in children and adolescents. Dev Med
Progrès médical : V.-A. Delahaye et Lecrosnier; Child Neurol 2003;45(10):700–703. doi:10.1017/
1885. 68 p. (Publications du Progrès médical). S0012162203001294.
2 Kushner HI. Medical fictions: the case of the 15 Prado HS, Rosário MC, Lee J, et al. Sensory
cursing marquise and the (re)construction of phenomena in obsessive-compulsive disorder and
Gilles de la Tourette’s syndrome. Bull Hist Med tic disorders: a review of the literature. CNS Spectr
1995;69(2):224–254. 2008;13(5):425–432. doi:10.1017/S1092852900016606.
3 Singer HS. Tics and Tourette syndrome. In: Singer 16 Shprecher DR, Gannon K, Agarwal N, et al. Elucidating
HS, Mink J, Gilbert DL, Jankovic J. Movement the nature and mechanism of tic improvement in
disorders in childhood. 2nd ed. London, UK: tourette syndrome: a pilot study. Tremor Other
Elsevier, 2016:81–109. Hyperkinet Mov (N Y) 2014;4:217. doi:10.7916/
D8TH8JQQ.
4 Cavanna AE, Robertson MM, Critchley HD. Catatonic
signs in Gilles de la Tourette syndrome. Cogn 17 Groth C, Mol Debes N, Rask CU, et al. Course of
Behav Neurol 2008;21(1):34–37. doi:10.1097/ Tourette syndrome and comorbidities in a
WNN.0b013e318165a9cf. large prospective clinical study. J Am Acad
5 Robertson MM, Trimble MR, Lees AJ. Self-injurious Child Adolesc Psychiatry 2017;56(4):304–312.
behaviour and the Gilles de la Tourette syndrome: doi:10.1016/j.jaac.2017.01.010.
a clinical study and review of the literature. 18 Erenberg G, Cruse RP, Rothner AD. The natural
Psychol Med 1989;19(3):611–625. doi:10.1017/ history of Tourette syndrome: a follow-up study.
S0033291700024211. Ann Neurol 1987;22(3):383–385. doi:10.1002/
6 Cheung MY, Shahed J, Jankovic J. Malignant ana.410220317.
Tourette syndrome. Mov Disord 2007;22(12): 19 Pappert EJ, Goetz CG, Louis ED, et al. Objective
1743–1750. doi:10.1002/mds.21599. assessments of longitudinal outcome in Gilles de la
7 Freeman RD, Zinner SH, Müller-Vahl KR, et al. Tourette’s syndrome. Neurology 2003;61(7):
Coprophenomena in Tourette syndrome. 936–940. doi:10.1212/01.WNL.0000086370.10186.7C.
Dev Med Child Neurol 2009;51(3):218–227.
20 Jankovic J, Gelineau-Kattner R, Davidson A.
doi:10.1111/j.1469-8749.2008.03135.x.
Tourette’s syndrome in adults. Mov Disord 2010;
8 Robertson MM, Eapen V, Singer HS, et al. Gilles 25(13):2171–2175. doi:10.1002/mds.23199.
de la Tourette syndrome. Nat Rev Dis Primers
21 Schaefer SM, Chow CA, Louis ED, Robakis D. Tic
2017;3:16097. doi:10.1038/nrdp.2016.97.
exacerbation in adults with Tourette syndrome: a
9 Lin H, Katsovich L, Ghebremichael M, et al. case series. Tremor Other Hyperkinet Mov (N Y)
Psychosocial stress predicts future symptom 2017;7:450. doi:10.7916/D8FF3Z1Q.
severities in children and adolescents with
22 Eapen V, Lees AJ, Lakke JP et al. Adult-onset tic
Tourette syndrome and/or obsessive-compulsive
disorders. Mov Disord 2002;17(4):735–740.
disorder. J Child Psychol Psychiatry 2007;48(2):
doi:10.1002/mds.10180.
157–166. doi:10.1111/j.1469-7610.2006.01687.x.
23 Kompoliti K, Goetz CG. Hyperkinetic movement
10 Hoekstra PJ, Steenhuis MP, Kallenberg CG,
disorders misdiagnosed as tics in Gilles de la
Minderaa RB. Association of small life events with
Tourette syndrome. Mov Disord 1998;13(3):
self reports of tic severity in pediatric and adult tic
477–480. doi:10.1002/mds.870130317.
disorder patients: a prospective longitudinal study.
J Clin Psychiatry 2004;65(3):426–431. 24 Mahone EM, Bridges D, Prahme C, Singer HS.
Repetitive arm and hand movements (complex
11 Horesh N, Zimmerman S, Steinberg T, et al. Is
motor stereotypies) in children. J Pediatr 2004;
onset of Tourette syndrome influenced by life
145(3):391–395. doi:10.1016/j.jpeds.2004.06.014.
events? J Neural Transm (Vienna) 2008;115(5):
787–793. doi:10.1007/s00702-007-0014-3. 25 Van Borsel J, Tetnowski JA. Fluency disorders in
genetic syndromes. J Fluency Disord 2007;32(4):
12 Cohrs S, Rasch T, Altmeyer S, et al. Decreased
279–296. doi:10.1016/j.jfludis.2007.07.002.
sleep quality and increased sleep related
movements in patients with Tourette’s syndrome. 26 Baizabal-Carvallo JF, Jankovic J. The clinical features
J Neurol Neurosurg Psychiatry 2001;70(2):192–197. of psychogenic movement disorders resembling
doi:10.1136/jnnp.70.2.192. tics. J Neurol Neurosurg Psychiatry 2014;85(5):
573–575. doi:10.1136/jnnp-2013-305594.
13 Kwak C, Dat Vuong K, Jankovic J. Premonitory
sensory phenomenon in Tourette’s syndrome. 27 American Psychiatric Association. Diagnostic and
Mov Disord 2003;18(12):1530–1533. doi:10.1002/ statistical manual of mental disorders, 5th ed
mds.10618. (DSM-5). Washington, DC: American Psychiatric
Association, 2013.
CONTINUUMJOURNAL.COM 955
55 Lombroso PJ, Scahill L. Tourette syndrome and 69 Kirov R, Kinkelbur J, Banaschewski T, Rothenberger
obsessive–compulsive disorder. Brain Dev 2008; A. Sleep patterns in children with attention-
30(4):231–237. doi:10.1016/j.braindev.2007.09.001. deficit/hyperactivity disorder, tic disorder,
and comorbidity. J Child Psychol Psychiatry
56 Rickards H, Robertson M. A controlled study of
2007;48(6):561–570. doi:10.1111/j.1469-7610.
psychopathology and associated symptoms in
2007.01729.x.
Tourette syndrome. World J Biol Psychiatry 2003;
4(2):64–68. doi:10.3109/15622970309167953. 70 Ghosh D, Rizkala A, Khuhro A. Management of
sleep problems helps improve tic control in children
57 Storch EA, Hanks CE, Mink JW, et al. Suicidal
with Tourette Syndrome/chronic tic disorder.
thoughts and behaviors in children and
Poster presented at: 47th Child Neurology
adolescents with chronic tic disorders. Depress
Society Meeting; Chicago IL, Annals of Neurology,
Anxiety 2015;32(10):744–753. doi:10.1002/da.22357.
84: issue 522, Oct 2018, S370.
58 Budman CL, Rockmore L, Stokes J, Sossin M.
71 Wang S, Mandell JD, Kumar Y, et al. De novo
Clinical phenomenology of episodic rage in
sequence and copy number variants are strongly
children with Tourette syndrome. J Psychosom
associated with Tourette disorder and implicate
Res 2003;55(1):59–65. doi:10.1016/S0022-
cell polarity in pathogenesis. Cell Rep 2018;24(13):
3999(02)00584-6.
3544. doi:10.1016/j.celrep.2018.12.024.
59 Mol Debes NM. Co-morbid disorders in Tourette
72 Fernandez TV, State MW, Pittenger C. Tourette
syndrome. Behav Neurol 2013;27(1):7–14.
disorder and other tic disorders. In: Geschwind
doi:10.3233/BEN-120275.
DH, Paulson HL, Klein C, editors. Handbook of
60 Mathews C, Waller J, Glidden D, et al. Self injurious Clinical Neurology. Amsterdam, the Netherlands:
behaviour in Tourette syndrome: correlates Elsevier, 2018:343–354.
with impulsivity and impulse control. J Neurol
73 Brainstorm Consortium; Anttila V, Bulik-Sullivan B,
Neurosurg Psychiatry 2004;75(8):1149–1155.
et al. Analysis of shared heritability in common
doi:10.1136/jnnp.2003.020693.
disorders of the brain. Science 2018;360(6395):
61 Silvestri PR, Baglioni V, Cardona F, Cavanna AE. 048991. doi:10.1126/science.aap8757.
Self-concept and self-esteem in patients with 74 Mataix-Cols D, Isomura K, Pérez-Vigil A, et al.
chronic tic disorders: a systematic literature review. Familial risks of Tourette syndrome and chronic
Eur J Paediatr Neurol 2018;22(5):749–756. tic disorders. A population-based cohort study.
doi:10.1016/j.ejpn.2018.05.008. JAMA Psychiatry 2015;72(8):787–793. doi:10.1001/
62 Cavanna AE, Robertson MM, Critchley HD. jamapsychiatry.2015.0627.
Schizotypal personality traits in Gilles de la 75 Pauls DL, Abramovitch A, Rauch SL, Geller DA.
Tourette syndrome. Acta Neurol Scand 2007;116(6): Obsessive-compulsive disorder: an integrative
385–391. doi:10.1111/j.1600-0404.2007.00879.x. genetic and neurobiological perspective. Nat Rev
63 Ghanizadeh A, Mosallaei S. Psychiatric disorders and Neurosci 2014;15(6):410–424. doi:10.1038/nrn3746.
behavioral problems in children and adolescents 76 Georgitsi M, Willsey AJ, Mathews CA, et al.
with Tourette syndrome. Brain Dev 2009;31(1): The genetic etiology of Tourette syndrome:
15–19. doi:10.1016/j.braindev.2008.03.010. large-scale collaborative efforts on the precipice
64 Jankovic J, Kwak C, Frankoff R. Tourette’s syndrome of discovery. Front Neurosci 2016;10:351.
and the law. J Neuropsychiatry Clin Neurosci 2006; doi:10.3389/fnins.2016.00351.
18(1):86–95. doi:10.1176/jnp.18.1.86. 77 Scharf JM, Moorjani P, Fagerness J, et al. Lack of
65 Singer HS, Schuerholz LJ, Denckla MB. Learning association between SLITRK1var321 and Tourette
difficulties in children with Tourette syndrome. syndrome in a large family-based sample.
J Child Neurol 1995;10(suppl 1):S58–S61. doi:10.1177/ Neurology 2008;70(16 pt 2):1495–1496. doi:10.1212/
08830738950100S112. 01.wnl.0000296833.25484.bb.
66 Pérez-Vigil A, Fernández de la Cruz L, Brander G, 78 Lai Wing Sun K, Correia JP, Kennedy TE. Netrins:
et al. Association of Tourette syndrome and versatile extracellular cues with diverse
chronic tic disorders with objective indicators functions. Development 2011;138(11):2153–2169.
of educational attainment: a population-based doi:10.1242/dev.044529.
sibling comparison study. JAMA Neurol 2018; 79 Browne HA, Hansen SN, Buxbaum JD, et al.
75(9):1098–1105. doi:10.1001/jamaneurol.2018.1194. Familial clustering of tic disorders and
67 Lee WT, Huang HL, Wong LC, et al. Tourette obsessive-compulsive disorder. JAMA
syndrome as an independent risk factor for Psychiatry 2015;72(4):359–366. doi:10.1001/
subsequent sleep disorders in children: a jamapsychiatry.2014.2656.
nationwide population-based case–control Study. 80 Yu D, Mathews CA, Scharf JM, et al. Cross-
Sleep 2017;40(3). doi:10.1093/sleep/zsw072. disorder genome-wide analyses suggest a
complex genetic relationship between
68 Ghosh D, Rajan PV, Das D, et al. Sleep disorders in
Tourette’s syndrome and OCD. Am J Psychiatry
children with Tourette syndrome. Pediatr Neurol
2015;172(1):82–93. doi:10.1176/appi.ajp.2014.
2014;51(1):31–35. doi:10.1016/j.pediatrneurol.
13101306.
2014.03.017.
CONTINUUMJOURNAL.COM 957
By Elan D. Louis, MD, MS, FAAN C O N T I N U UM A U D I O
I NT E R V I E W A V A I L AB L E
ONLINE
ABSTRACT VIDEO CONTENT
PURPOSE OF REVIEW: Tremor may be defined as an involuntary movement that A V AI L A B L E O N L I N E
H
carbamazepine, clonazepam,
umans have been documenting their tremors for thousands of ethosuximide, and phenytoin
years.1 Tremor is defined as an involuntary movement that is both for the treatment of orthostatic
rhythmic (ie, regularly recurrent) and oscillatory (ie, rotating tremor; pregabalin for the
treatment of neuropathic
around a central plane).2 Tremor may manifest in a vast array of tremor; benzodiazepines for the
ways. As such, the clinical phenomenology is very rich, and it treatment of parkinsonian
resting tremor; and
should be no surprise that numerous methods for classifying tremors exit. Thus,
phenobarbital to treat the side
tremors may be classified based on speed (ie, frequency measured in hertz), effects of acute nausea and
regions of the body affected (eg, arm, voice, head), activation state in which unsteadiness that occur as
a result of other treatments
tremor manifests (eg, when the body part is at rest), occurrence of associated for tremor.
medical conditions (eg, hyperthyroidism), and the brain region from which the
tremor arises (eg, basal ganglia, cerebellum). Because there are many ways to
classify and divide tremor, a large nomenclature applies to tremor. The richness © 2019 American Academy
of the clinical phenomenology and its associated nomenclature can be daunting of Neurology.
CONTINUUMJOURNAL.COM 959
to the clinician. The goal of this article is to provide the reader with a basic
approach to the diagnosis and management of the patient with tremor. This
approach includes a medical history, a focused neurologic examination,
diagnosis, and, finally, treatment.
Medical History
The first set of questions should be directed at determining whether the tremor is
one that occurs with action or at rest. It is best to begin with an initial question
that is open-ended (eg, “Can you tell me about your tremor?” or “What type
of tremor do you have?” or “When do you notice tremor?”). After this initial
question, more specific questions, such as “Does your hand shake when you are
writing?” or “Does your hand shake when you are trying to eat something?” may
be asked to further ascertain whether the tremor is an action tremor or a resting
tremor. This is then followed by additional questions that elicit information on
the following items:
u The body areas that seem to be shaking (eg, arms, head, voice)
u The limb positions that bring on the tremor and, conversely, those that seem to lessen it
u The age at which tremor began
u How the tremor has changed over the years
u The presence of other involuntary movements
u The presence of other neurologic symptoms aside from tremor
u The presence of pulling sensations or discomfort in the body part that is shaking
u The use of medications that seem to produce or exacerbate tremor
u Dietary factors that exacerbate tremor (eg, coffee and other forms of caffeine)
u Symptoms of thyroid diseases (eg, weight loss, heat intolerance)
u Family history of “shaking” or tremor (eg, the presence of affected first-degree relatives
is often reported by patients with essential tremor, among whom the pattern of
inheritance may resemble that of an autosomal dominant disease)
Neurologic Examination
After the medical history, a detailed and focused neurologic examination should
be performed. First, the examiner should ask the patient to raise his or her arms
against gravity, with the palms down in front and then in the wing-beat position
with the hands facing one another in the midline. If a postural tremor is present
during sustained arm extension, the examiner should assess the following:
● An initial step in
Next, the examiner should attempt to elicit kinetic tremor—a tremor that
evaluating patients with
occurs during voluntary movements. Thus, the examiner may ask the patient to tremor is to determine
perform the finger-nose-finger maneuver, pour water between cups, draw whether the tremor is
spirals, or write a sentence. The examiner should assess the following items: primarily present at rest or
with activity.
u Does the tremor have an intentional component (ie, does the tremor worsen as the limb
● The key feature of
approaches a target [eg, during the finger-nose-finger maneuver])?
essential tremor is kinetic
u Are dystonic movements or postures present (eg, do some of the fingers flex, extend, or tremor.
twist during the finger-nose-finger maneuver)?
● The kinetic tremor of
u What is the relative severity of the kinetic tremor that is being observed to that which was
essential tremor is typically
observed during sustained posture (above)?
slightly asymmetric.
Next, the examiner should assess whether there is any tremor at rest in the ● Approximately one-half
patient’s arms or legs. Tremor at rest in the arms can be assessed while the patient of patients with essential
is seated, standing, walking, and lying down. Resting tremor in the legs can be tremor exhibit intention
tremor during the finger-
assessed while the patient is seated or lying down. In addition, tremor while nose-finger maneuver.
standing (ie, orthostatic tremor) may be assessed while the patient is standing in
a stationary position.
Finally, the examiner may assess for tremor in the head (ie, neck) (while the
patient is seated and lying down), jaw (with the patient’s mouth closed and then
with the mouth held open), facial muscles (eg, forehead, cheek), chin, tongue,
and voice (during sustained phonation and during speech).
DIAGNOSIS
The history and physical examination are first used to establish whether the main
type of tremor is an action tremor (ie, postural, kinetic, or intention tremor) or a
tremor at rest. Indeed, this is a primary point of divergence: those diseases in
which action tremor is the predominant tremor versus those diseases in which
resting tremor is the predominant tremor, each of which will be discussed in
turn, beginning with the former because these are both of a larger variety and
more prevalent.
Essential Tremor
The central clinical feature of essential tremor is kinetic tremor. This is generally
observed during numerous activities of daily living, ranging from eating to
writing, and may be elicited on neurologic examination during a variety of
maneuvers (eg, finger-nose-finger maneuver, spiral drawing, pouring water
between two cups) (VIDEO 4-1, links.lww.com/CONT/A278). Rather than being
totally symmetric, the tremor is usually slightly asymmetric, affecting one arm
more than the other. In approximately 5% of patients, this tremor is markedly
asymmetric or unilateral.3 In approximately 50% of patients with essential
CONTINUUMJOURNAL.COM 961
CASE 4-1 A 68-year-old woman presented with tremor, which she noticed when
holding an eating utensil and when writing. The tremor had begun about
5 years previously, and it seemed to be getting worse, little by little, with
each passing year. Her father and one of her two sisters had had a similar
tremor. Although she was unaware of it herself, one of her children told
her he sometimes noticed a mild, side-to-side head tremor.
She had previously been treated with propranolol but was unable to
tolerate more than 80 mg/d because her heart rate dropped below
60 beats/min. Furthermore, this dose had resulted in only a modest (ie, 10%)
reduction in her tremor. She had also taken primidone, 50 mg in the morning,
but the initial dose made her nauseated, and she did not take it again.
On examination, the tremor was noticeable when she drew spirals and
performed the finger-nose-finger maneuver, and it worsened slightly as she
approached her nose during this maneuver. She also had a postural tremor,
but it was considerably less than the tremor observed while she used her
hands to write. She was started on topiramate 25 mg/d and was gradually
increased to 200 mg/d, resulting in a moderate reduction in the amplitude
of her tremor.
COMMENT This case illustrates the slowly progressive and often familial nature of
essential tremor, the fact that the central defining clinical feature of
essential tremor is kinetic tremor, and the presence of head tremor in some
patients with essential tremor, particularly women.
CONTINUUMJOURNAL.COM 963
type of postural tremor that commences after a brief latency of several seconds
and is another feature of PD. The use of dopamine transporter (DAT) imaging
can be useful in distinguishing patients with essential tremor from those with
parkinsonism, although its use is supplemental to clinical information derived
from the history and examination.11,12
Aside from a mild dystonic posture, noted above, dystonic postures, movements,
or tremor are not features of essential tremor. In addition, dystonic tremor
is often neither rhythmic nor oscillatory. The patient should be assessed for
the presence of neck dystonia, which is characterized by head tilt or rotation,
hypertrophy of the sternocleidomastoid or other neck muscles, the presence of a
tremor null-point, or a sensory trick by history (ie, a maneuver such as touching
the chin or back of the head that lessens the tremor).
Scanning or dysarthric speech or nystagmus may be present in patients with
spinocerebellar ataxias; however, these are not features of essential tremor.
Hyperthyroidism can be assessed by clinical history (eg, symptoms of weight loss
or heat intolerance) as can the use of medications (eg, lithium, prednisone,
valproate) or other substances (eg, tobacco, caffeine) that may produce or
exacerbate action tremor.
A difficult differential is between that of mild essential tremor and enhanced
physiologic tremor, although the presence of neck tremor should exclude
the latter. Computerized tremor analysis with inertial loading can assist with this
differential, although this is often not available outside of research-oriented
tertiary referral centers. In patients with a tremor of central origin (eg, essential
tremor), the primary tremor frequency should not change with inertial
loading; in patients with enhanced physiologic tremor, the frequency will
reduce. Other features that support an essential tremor diagnosis are the
presence of essential tremor in one or more first-degree relatives. A reported
reduction in tremor with ethanol use is often used as a diagnostic tool; however,
this is not very specific and of limited utility. Indeed, patients with most
tremor disorders often experience a reduction in tremor following ethanol
consumption.
The main motivators for treatment in essential tremor are embarrassment
and functional disability. Beta-blockers (especially propranolol) and primidone,
alone or in combination, are the most effective pharmacologic agents, although
many patients choose to discontinue these medications because of their limited
efficacy and side effects.
Propranolol has been used in doses up to 360 mg/d, although doses in excess
of 80 mg/d to 100 mg/d are rarely tolerated in patients who are elderly, with
the main issue being bradycardia. A conservative starting dose is 20 mg/d,
and this is gradually increased as noted above. Asthma is only a relative
contraindication to the use of propranolol, and propranolol use should be
considered on a case-by-case basis. Primidone can be given in doses up to
1500 mg/d, although lower doses (eg, starting with 25 mg and gradually
increasing to 500 mg/d) are often effective. Acute nausea or unsteadiness is
observed in approximately 25% of patients, irrespective of the starting dose, and
in the author’s experience, preloading with phenobarbital (ie, 30 mg 2 times a
day for 3 days) is one method to avoid this unwanted side effect. Propranolol
and primidone may result in mild to moderate reduction in the amplitude of
tremor in 30% to 70% of patients with essential tremor. If the tremor is mild, it
may be abolished.
CONTINUUMJOURNAL.COM 965
Dystonic Tremor
A range of tremors may occur among patients who have been diagnosed with
dystonia, and a challenging differential is between the diagnosis of essential tremor
and the diagnosis of dystonic tremor.18–20 In patients who have been diagnosed with
dystonia, tremor may occur both in limbs that exhibit dystonic postures or
movements and in limbs that do not exhibit these. Furthermore, the tremor may
occur in limbs at rest as well as in limbs that are active (ie, during sustained posture
or during voluntary movements). What complicates matters is that, as noted above,
patients with long-standing and clinically advanced essential tremor may develop
mild dystonic posturing of the hand during arm extension. As a result, considerable
debate exists as to where essential tremor as a disease ends and where dystonia as a
disease begins and vice versa. One further point is that the tremor in patients
with dystonia is not always regularly recurrent. This raises the issue in these
patients as to whether the “tremor” is indeed, in the strictest sense of the word, a
tremor. This author sometimes uses the term tremulous rather than tremor to
describe such movements.
With this uncertainly in mind, when a clinician is confronted with an
individual patient, several issues should be taken into consideration. First, what
CASE 4-2 A 56-year-old woman presented with a chief complaint of head tremor
with some mild associated right-sided neck pain. The pain had begun
10 years previously. For 4 years, she had also felt an intermittent pulling
sensation in the neck region. She further noted that her head had been
turning to one side and sometimes it even felt a little shaky. She did not
report any tremor in her hands.
On examination, she had a mild to moderate postural tremor of the
right arm, with a little bit of flexed posturing of her index and middle
fingers. On the finger-nose-finger maneuver, no tremor was observable.
Her right sternocleidomastoid muscle was slightly hypertrophic, and her
head tended to preferentially turn to the left and shake intermittently;
the shakiness was irregular. This head tremor persisted even when she lay
down on her back on the examining table.
She was treated with IM botulinum toxin injections to several neck
muscles, which helped diminish her symptoms, although they did not
resolve completely.
COMMENT This case illustrates several important features of dystonic head (neck)
tremor: the tremor is often nonrhythmic, and it often persists in the
recumbent position. Furthermore, it may be accompanied by pain or pulling
sensations of the neck.
Orthostatic Tremor
This is a rare syndrome characterized by unsteadiness on standing and high-
frequency tremor in the legs.25,26 The typical onset is in the sixth decade of life.
CONTINUUMJOURNAL.COM 967
Although most cases are sporadic, rare familial cases have been reported. Patients
more often note unsteadiness while standing rather than tremor per se. Because of
these symptoms, patients typically avoid situations in which they have to stand
still (eg, standing in lines). Indeed, they are eventually forced to sit down or walk
after a short time (ie, seconds to minutes), depending on the severity of the
disease. As the disease progresses, the tremor may begin to encroach upon the
stance phase of walking. On examination, the patient may see or feel a rapid
(14 Hz to 16 Hz) fine tremor in the calves. Due to its high frequency and low
amplitude, orthostatic tremor may be difficult to appreciate on visual inspection.
In some cases, the tremor may be heard when a stethoscope is placed over the
affected calf, sounding like a distant helicopter. The EMG indicates the presence
of a 14 Hz to 16 Hz synchronous tremor in the leg (especially in calf ) muscles.
A slower, larger-amplitude tremor may also be superimposed on top of this
tremor, perhaps representing a subharmonic of the high-frequency tremor, and
this can be more disabling for patients than the faster tremor. Numerous cases
occur in the setting of comorbid PD.
The treatment of orthostatic tremor is challenging.25 Many agents have
been used and often to little avail. The most commonly used agents are
clonazepam (0.5 mg/d to 4 mg/d), gabapentin (300 mg/d to 1800 mg/d), and
carbidopa/levodopa (25 mg/100 mg per day to 250 mg/1000 mg per day). Many
other agents have also been tried, including propranolol, primidone, phenytoin,
carbamazepine, ethosuximide, baclofen, and acetazolamide, although given the
rarity of the disorder no large-scale clinical trials have been conducted. DBS
surgery can also provide benefit to some patients.
Cerebellar Tremor
The term cerebellar tremor has classically been used to describe tremor that can
occur in patients with spinocerebellar ataxias and other classical disorders
originating in the cerebellum.27 In modern times, cerebellar tremor has
become equated exclusively with intention tremor.28 This is a tremor that
occurs with goal-directed movement (eg, finger-to-nose maneuver) and
worsens when approaching a target. However, cerebellar tremors (ie, tremors
of cerebellar origin) do not always present exclusively as intention tremor.
Indeed, the clinical phenomenology of tremor of cerebellar origin is
heterogeneous, and it extends beyond that of intention tremor to include
postural tremor, kinetic tremor, resting tremor, and orthostatic tremor.28 This
heterogeneity is consistent with the seminal work of Holmes,29 who described
a variety of tremors aside from intention tremor in the setting of cerebellar
lesions.30
On examination, patients with classically defined cerebellar tremor often
have other cerebellar signs, including saccadic eye movement abnormalities,
dysarthric or scanning speech, gait ataxia, and hypotonia. When these patients are
examined, it is important, although often difficult, to separate the tremor
(ie, rhythmic oscillatory movements) from problems with force and timing of
motion (ie, dysmetria); both may occur during the finger-nose-finger maneuver,
but the former generally improves with DBS surgery whereas the latter
might worsen.
A number of medications have been used to treat cerebellar tremor, although
their efficacy is limited. The most effective treatment for severe cerebellar
tremor is thalamic DBS surgery, with the caveat noted above.31
CONTINUUMJOURNAL.COM 969
Parkinson Disease
Tremor in patients with PD is classically a tremor at rest. The resting tremor
is generally asymmetric, affecting one side of the body (ie, arm, leg, or both)
preferentially; it typically begins in one limb. In patients with upper limb tremor,
the tremor typically involves distal joints (eg, fingers and wrist) rather than
proximal joints (eg, elbow or shoulder). Aside from the arms, tremor may affect
the jaw, although in contrast to essential tremor, it is more often noted when
the patient’s mouth is closed and relaxed rather than while the patient is
speaking. In patients with PD, tremor rarely affects the head.
The treatment of parkinsonian resting tremor includes the use of anticholinergic
agents (trihexyphenidyl 2 mg/d to 10 mg/d), amantadine (up to 300 mg/d), as
well as carbidopa/levodopa (up to 500/2000 mg/d of carbidopa/levodopa),
dopamine agonists, and rasagiline.55,56 DBS surgery for tremor is reserved for
patients with PD who have severe tremor and who are refractory to medications.
Although resting tremor is one of the hallmark features of PD, a large proportion
of patients also have postural or kinetic tremor (or both) of the arms. Sometimes
the postural and kinetic tremors have a reemergent quality; this so-called
“reemergent tremor” surfaces after a latency of 1 or several seconds, has a
frequency that is similar to that of the resting tremor in PD, and often attains
amplitudes greater than that seen in patients with essential tremor (VIDEO 4-5,
links.lww.com/CONT/A282).57 This tremor tends to increase in severity (ie, it
crescendos) with sustained posture or during the course of repetitive movements
during which much of the limb is immobile (eg, while pouring water between
two cups, during which much of the movement is proximal rather than distal).
CONTINUUMJOURNAL.COM 971
The treatment of this type of tremor is similar to the treatment of the resting
tremor of PD, although it is less responsive to medications than resting tremor.
CONCLUSION
Tremors are among the most common movement disorders. The diagnosis of
these disorders is challenging. The approach to a patient with tremor involves a
history and careful neurologic examination focused on the nuances of clinical
phenomenology. When generating the differential diagnosis, it is important to
first consider whether the primary type of tremor is an action tremor or a resting
tremor. As is true for the diagnosis of most disorders of involuntary movement,
arriving at the correct diagnosis is often based on pattern recognition.
VIDEO LEGENDS
VIDEO 4-1 VIDEO 4-4
Essential tremor. Video shows a man with essential Holmes tremor. Video show a woman with Holmes
tremor exhibiting a severe kinetic tremor while tremor exhibiting a unilateral, slow tremor at rest,
pouring water between two cups. The tremor is which worsens during sustained posture.
slightly asymmetric and is worse on the right. links.lww.com/CONT/A281
links.lww.com/CONT/A278
Courtesy of Sule Tinaz, MD, PhD.
© 2019 American Academy of Neurology. © 2019 American Academy of Neurology.
1 Louis ED. Essential tremor. Arch Neurol 2000;57: 13 Mittal SO, Machado D, Richardson D, et al.
1522–1524. Botulinum toxin in essential hand tremor—a
randomized double-blind placebo-controlled
2 Bhatia KP, Bain P, Bajaj N, et al. Consensus
study with customized injection approach.
Statement on the classification of tremors. from
Parkinsonism Relat Disord 2018;pii:S1353-
the task force on tremor of the International
8020(18)30278–5. doi:10.1016/j.parkreldis.2018.06.019.
Parkinson and Movement Disorder Society.
Mov Disord 2018;33(1):75–87. doi:10.1002/ 14 Dallapiazza RF, Lee DJ, De Vloo P, et al. Outcomes
mds.27121. from stereotactic surgery for essential tremor.
J Neurol Neurosurg Psychiatry 2018;pii:jnnp-2018-
3 Phibbs F, Fang JY, Cooper MK, et al. Prevalence
318240. doi:10.1136/jnnp-2018-318240.
of unilateral tremor in autosomal dominant
essential tremor. Mov Disord 2009;24(1):108–111. 15 Elble RJ, Higgins C, Elble S. Electrophysiologic
doi:10.1002/mds.22113. transition from physiologic tremor to essential
tremor. Mov Disord 2005;20(8):1038–1042.
4 Louis ED. The primary type of tremor in essential
doi:10.1002/mds.20487.
tremor is kinetic rather than postural: cross-
sectional observation of tremor phenomenology 16 Morgan JC, Kurek JA, Davis JL, Sethi KD. Insights
in 369 cases. Eur J Neurol 2013;20(4):725–727. into pathophysiology from medication-induced
doi:10.1111/j.1468-1331.2012.03855.x. tremor. Tremor Other Hyperkinet Mov (N Y) 2017;
7:442. doi:10.7916/D8FJ2V9Q.
5 Sternberg EJ, Alcalay RN, Levy OA, Louis ED.
Postural and intention tremors: a detailed clinical 17 Raethjen J, Lemke MR, Lindemann M, et al.
study of essential tremor vs. Parkinson’s disease. Amitriptyline enhances the central component
Front Neurol 2013;4:51. doi:10.3389/fneur. of physiological tremor. J Neurol Neurosurg
2013.00051. Psychiatry 2001;70(1):78–82.
6 Rao AK, Gilman A, Louis ED. Balance confidence 18 Defazio G, Conte A, Gigante AF, et al. Is tremor in
and falls in nondemented essential tremor dystonia a phenotypic feature of dystonia?
patients: the role of cognition. Arch Phys Med Neurology 2015;84:1053–1059. doi:10.1212/WNL.
Rehabil 2014;95(10):1832–1837. doi:10.1016/ 0000000000001341.
j.apmr.2014.04.001.
19 Defazio G, Gigante AF, Abbruzzese G, et al. Tremor
7 Louis ED, Agnew A, Gillman A, et al. Estimating in primary adult-onset dystonia: prevalence and
annual rate of decline: prospective, longitudinal associated clinical features. J Neurol Neurosurg
data on arm tremor severity in two groups of Psychiatry 2013;84(10):404–408. doi:10.1212/
essential tremor cases. J Neurol Neurosurg WNL.0000000000001341.
Psychiatry 2011;82(7):761–765. doi:10.1136/jnnp.
20 Pandey S, Sarma N. Tremor in dystonia. Parkinsonism
2010.229740.
Relat Disord 2016;29:3–9. doi:10.1016/j.parkreldis.
8 Hardesty DE, Maraganore DM, Matsumoto JY, 2016.03.024.
Louis ED. Increased risk of head tremor in women
21 Agnew A, Frucht SJ, Louis ED. Supine head tremor:
with essential tremor: longitudinal data from the
a clinical comparison of essential tremor and
Rochester Epidemiology Project. Mov Disord
spasmodic torticollis patients. J Neurol Neurosurg
2004;19(5):529–533. doi:10.1002/mds.20096.
Psychiatry 2012;83(2):179–181. doi:10.1136/jnnp-
9 Robakis D, Louis ED. Head tremor in essential 2011-300823.
tremor: “yes-yes”, “no-no”, or “round and
22 Rana AQ, Vaid HM. A review of primary writing
round”? Parkinsonism Relat Disord 2016;22:
tremor. Int J Neurosci 2012;122(3):114–118.
98–101. doi:10.1016/j.parkreldis.2015.11.002.
doi:10.3109/00207454.2011.635827.
10 Jain S, Lo SE, Louis ED. Common misdiagnosis
23 Hai C, Yu-ping W, Hua W, Ying S. Advances in
of a common neurological disorder: how are we
primary writing tremor. Parkinsonism Relat Disord
misdiagnosing essential tremor? Arch Neurol
2010;16(9):561–565. doi:10.1016/j.parkreldis.
2006;63(8):1100–1104. doi:10.1001/archneur.
2010.06.013.
63.8.1100.
24 Papapetropoulos S, Singer C. Treatment of
11 Oravivattanakul S, Benchaya L, Wu G, et al.
primary writing tremor with botulinum toxin type
Dopamine transporter (DaT) scan utilization in a
A injections: report of a case series. Clin
movement disorder center. Mov Disord Clin
Neuropharmacol 2006;29(6):364–367.
Pract 2015;3(1):31–35. doi:10.1002/mdc3.12261.
doi:10.1097/01.WNF.0000236765.00785.9C.
12 Waln O, Wu Y, Perlman R, et al. Dopamine transporter
25 Yaltho TC, Ondo WG. Orthostatic tremor: a review
imaging in essential tremor with and without
of 45 cases. Parkinsonism Relat Disord 2014;20(7):
parkinsonian features. J Neural Transm (Vienna)
723–725. doi:10.1016/j.parkreldis.2014.03.013.
2015;122(11):1515–1521. doi:10.1007/s00702-015-
1419-z. 26 Benito-León J, Domingo-Santos Á. Orthostatic
tremor: an update on a rare entity. Tremor Other
Hyperkinet Mov (N Y) 2016;6:411. doi:10.7916/D81N81BT.
CONTINUUMJOURNAL.COM 973
CONTINUUMJOURNAL.COM 975
The Dystonias
C O N T I N UU M A UD I O By H. A. Jinnah, MD, PhD
I NT E R V I E W A V AI L A B L E
ONLINE
ABSTRACT
CITE AS:
PURPOSE OF REVIEW: This article provides a summary of the state of the art in
CONTINUUM (MINNEAP MINN)
2019;25(4, MOVEMENT DISORDERS): the diagnosis, classification, etiologies, and treatment of dystonia.
976–1000.
[email protected]. diagnosis by focusing on key clinical features that help distinguish the
many subtypes. In the past few years, major advances have been made in
RELATIONSHIP DISCLOSURE:
the discovery of new genes as well as advances in our understanding of the
Dr Jinnah has received personal
compensation for serving on biological processes involved. These advances have led to major changes
the advisory boards of and in strategies for diagnosis of the inherited dystonias. An emerging trend is
as a consultant for Abide
Therapeutics, Inc; Allergan, Inc;
to move away from heavy reliance on the phenotype to target diagnostic
CoA Therapeutics; the testing toward a broader approach that involves large gene panels or
International Neurotoxin Society; whole exome sequencing.
the International Parkinson and
Movement Disorders Society;
Medtronic; the Parkinson's SUMMARY: The dystonias are a large family of phenotypically and
Foundation; Psyadon etiologically diverse disorders. The diagnosis of these disorders depends
Pharmaceuticals Inc; Retrophin,
Inc; and Saol Therapeutics. on clinical recognition of characteristic clinical features. Symptomatic
Dr Jinnah has received grant treatments are useful for all forms of dystonia and include oral
support from the Benign
Essential Blepharospasm
medications, botulinum toxins, and surgical procedures. Determination of
Research Foundation, Cavion, etiology is becoming increasingly important because the number of
Cure Dystonia Now, the Dystonia disorders is growing and more specific and sometimes disease-modifying
Study Group, Ipsen Group,
National Institutes of Health
therapies now exist.
(NIH), and Retrophin, Inc. He also
is principle investigator for the
Dystonia Coalition, which
receives the majority of its
support through NIH grant INTRODUCTION
T
TR001456 from the Office of Rare he dystonias are a diverse family of disorders that share an underlying
Diseases Research at the
National Center for Advancing
phenomenon of excessive contractions of specific muscle groups
Translational Sciences and leading to abnormal movements.1 Any region of the body can be
previously received support affected, and the overt manifestations depend on the severity and
through grant NS065701 from the
National Institutes of
distribution of muscles involved. In its mildest forms, abnormalities
Neurological Disorders and appear as slight distortions of otherwise normal movements. In patients who
Stroke. The Dystonia Coalition are more affected, abnormal movements have a more obvious appearance of
has received additional material
or administrative support from cramping, stiffening, jerking, or twisting. The most severe cases of dystonia
industry sponsors (Allergan, Inc are associated with fixed abnormal postures or joint deformities with
and Merz Pharmaceuticals) as
severe disability.
well as private foundations (the
American Dystonia Society, The causes of dystonia are similarly diverse.2 Some types of dystonia are
Continued on page 1000 associated with overt neuropathologic abnormalities of the brain that can be
© 2019 American Academy detected by neuroimaging or postmortem histopathologic studies, such as focal
of Neurology. lesions or degenerative changes. Some types of dystonia are acquired whereas
DIAGNOSIS OF DYSTONIA
Dystonia is easy to recognize in its most classic expressions, in which patients
exhibit twisting movements and abnormal postures affecting many regions of
the body. Less severe expressions are often misdiagnosed. Several studies have
revealed that the average time from onset of symptoms to diagnosis can take
many years, even for the most common subtypes (TABLE 5-1).3–7 Part of the
reason for delayed diagnosis is that the definition of dystonia and the many
syndromes included under this umbrella term have evolved over the years.
Definition of Dystonia
The definition of dystonia has evolved considerably since it was first described
more than 100 years ago. In 2013, an international panel of experts agreed to the
following working definition:
Dystonia is defined as a movement disorder characterized by sustained
or intermittent muscle contractions causing abnormal, often repetitive,
movements, postures, or both. Dystonic movements are typically patterned
and twisting, and may be tremulous. Dystonia is often initiated or worsened
by voluntary action and associated with overflow muscle activation.1
One of the main reasons for delayed diagnosis is lack of appreciation that
abnormal movements in dystonia can fall into two broad categories that often
overlap. They can be slow and twisting and sometimes with abnormal postures
that appear to be fixed, such as the abnormal postures and slow but repetitive
deviations of the head in cervical dystonia or the overt twisting of a foot.
Alternatively, dystonic movements can be rapid and jerky, including the
periocular spasms of blepharospasm, voice breaks of laryngeal dystonia, dystonic
tremor, and myoclonic dystonia. In some patients, movements may be rapid and
a
This table describes the average length of time from symptom onset to diagnosis of dystonia.
CONTINUUMJOURNAL.COM 977
Clinical Evaluation
The many types of dystonia are
grouped according to two main
axes: a clinical axis and an
etiologic axis (FIGURE 5-1). The
clinical history and examination
(axis I) should address four
dimensions that include age at
onset, body region affected,
specific temporal features, and
whether associated clinical FIGURE 5-1
Classification of the dystonias. The many types of
problems are present.1 Age at dystonia are classified according to two
onset is important because independent axes. One axis relates to clinical
subtypes that emerge in infancy features, and the other relates to etiology.
Laboratory Investigations
Each of the four clinical dimensions described above is important for describing a
syndromic pattern, which helps guide laboratory investigations to delineate
etiology, which falls in axis II (FIGURE 5-2). Universal algorithms for laboratory
investigations in the dystonias are challenging because of the remarkable
heterogeneity of clinical manifestations and causes. A comprehensive review
described more than 100 dystonic disorders organized into 18 tables based on
specific clinical features.2 Although several different algorithms have been
proposed,10–14 none is complete. Following are some general guidelines for
laboratory investigations.
For dystonias that first emerge in later adulthood (patients older than
40 years of age), laboratory investigations depend on body distribution,
whether additional neurologic features are present, and temporal aspects. For
the most common adult-onset focal or segmental dystonias, such as cervical
dystonia or blepharospasm, diagnostic testing is usually unrevealing. For
laryngeal dystonia, laryngoscopy is recommended to rule out structural
defects of the vocal apparatus.15 For the less common adult-onset cases with
hemidystonia or generalized dystonia, neuroimaging can be useful to reveal a
structural cause. Neuroimaging is also useful in patients with adult-onset
dystonia with rapid or severe progression of symptoms or if dystonia is
combined with other neurologic features, such as parkinsonism or ataxia. For
CONTINUUMJOURNAL.COM 979
FIGURE 5-2
Workup of dystonia. The evaluation begins with clinical evaluation according to four main
clinical domains. The resulting syndromic pattern can be used to guide the approach to the
diagnostic workup.
most cases that emerge in later adulthood, genetic testing is not usually
conducted unless multiple family members are affected or a specific
syndromic pattern leads to suspicion for an inherited disorder. EEG is not
usually conducted unless dystonia appears in paroxysmal attacks. EMG is not
needed unless a neuromuscular disorder that may resemble dystonia is
suspected, such as myotonia or stiff person syndrome.
For dystonias that first emerge at an earlier age (in patients younger than
40 years), some laboratory investigations are almost always valuable.2 The
experts to remember all the red flags. ● For almost all classic
A related strategy is to delineate a group of clinical abnormalities that form inherited dystonic disorders
a recognizable syndromic pattern to guide diagnostic testing. An example in children, late-onset cases
of this is Lesch-Nyhan disease, where patients present with early-onset or less severe cases are
known to occur in adults.
generalized dystonia, intellectual disability, tendencies toward self-harm, and
elevations of serum uric acid.16,17 This syndrome is unique; it is associated only ● Elucidating etiology is
with the HPRT1 gene, so single gene or enzyme testing is efficient.18 However, important because specific
many other disorders present with overlapping syndromes with many causes. treatments are available for
several types of dystonia.
It can be challenging even for experts to remember all syndromes, and partial
syndromes are hard to recognize. For almost all of the classic phenotypes
described for inherited disorders of children, adult-onset variant forms with
partial syndromes may occur. Furthermore, syndromic patterns are sometimes
misleading. An example involves the ATM gene, which is classically associated
with ataxia-telangiectasia in children. However, mutations in this gene can
sometimes cause isolated focal or generalized dystonia, sometimes in adults and
sometimes without telangiectasias.19 In the era of shotgun genetic testing that
may involve whole exome sequencing or large multigene panels, unexpected
results are common.20,21
A third strategy sometimes recommended is focusing laboratory
investigations specifically on disorders for which specific treatments exist.
The rationale for this strategy is that obtaining a definitive etiologic diagnosis
for dystonia often does not change clinical management because most are
managed symptomatically. According to this strategy, laboratory testing
should focus only on a smaller number of treatable disorders, such as Wilson
disease, for which specific treatments are available. This strategy is unwise.
Even when the cause does not have a specific treatment, many patients and
families find that obtaining a definitive etiologic diagnosis is useful for ending
an often long diagnostic odyssey.20,22–24 The information also provides valuable
information for counseling regarding the prognosis for the individual affected and
the potential risk for family members. A more important weakness of this
strategy is that the number of disorders with specific treatments has been
growing rapidly in recent years.
A recent review summarized more than 30 inherited disorders with specific
therapeutic interventions, and dystonia occurred in more than half of those
disorders.25 In these disorders, dystonia often is part of a more complex picture
with multiple neurologic and systemic problems. However, sometimes dystonia
is the sole initial presentation, and the diagnosis is delayed because the whole
syndrome has not yet evolved. At the same time, early treatment often is critical
to prevent permanent neurologic sequelae. The list of treatable disorders has
CONTINUUMJOURNAL.COM 981
CAUSES OF DYSTONIA
There have been enormous advances in elucidating the varied causes of dystonia
and in understanding the different biological mechanisms involved. This section
summarizes the major conceptual advances in three areas: genetic, physiologic,
and neuroanatomic.
Genetic Basis
Historically, most genes have been identified by a laborious process that
involved collecting large families, correlating the disease phenotype with known
genetic markers spread across the genome, and then sequencing DNA in the
chromosomal region with the best links between the phenotype and the known
markers. To aid this process of gene discovery, nomenclature was developed
that was based on linkage to chromosomal locations, or loci. Disorders with
dystonia were given the prefix DYT followed by a number, such as DYT1, DYT2,
DYT3, and so on.
This nomenclature has numerous flaws.29 In some cases, different DYT
loci ultimately were linked with the same gene. In other cases, multiple genes
were linked with the same DYT locus. Several DYT entries were found to be
erroneous or were never verified. The nomenclature also was clinically
misleading because it incorrectly implied that dystonia was a significant
feature of any disorder with a DYT label. The most serious drawback of this
nomenclature was that it was incomplete. It did not include dystonia genes
that were found before the DYT convention was established. Wilson disease
CONTINUUMJOURNAL.COM 983
is one of many examples that was never included in the DYT lists despite the
fact that dystonia occurs in nearly all cases with neurologic involvement.
In fact, more than 100 inherited conditions exist in which dystonia may be a
major feature, a presenting feature, a dominant feature, or part of a more
complex syndrome.2 Only a small fraction of these disorders was assigned a
DYT label.
The next generation of genetic methods has led to major changes in our
approach to identifying genes for dystonia. A linkage-based nomenclature is
no longer central to gene discovery, and new genes are no longer consistently
being assigned DYT labels. Instead, a new nomenclature has been proposed.29
In the new nomenclature, the number is replaced by the gene name. For
example, TOR1A-associated dystonia (DYT1 according to the old nomenclature)
would be called DYT-TOR1A. Additionally, when the phenotype is mixed,
multiple prefixes may be combined, such as DYT for dystonia, PARK
for parkinsonism, or SCA for spinocerebellar ataxia. For example, dopa-
responsive dystonia (DYT5 in the old nomenclature) would be called
DYT/PARK-GCH1, DYT/PARK-TH, or DYT/PARK-SPR. The new convention
more accurately acknowledges the frequent occurrence of parkinsonism with
dystonia in dopa-responsive dystonia as well as at least three different
causative genes.
This new nomenclature is still evolving. It requires expertise in neurogenetics
and can be challenging to apply in routine clinical practice. For the practicing
neurologist, it is most useful to group genes by pattern of inheritance
(TABLE 5-3). The pattern of inheritance is important for genetic counseling,
and knowing the gene has implications for mechanism-specific treatments,
such as for Wilson disease. However, it is important to recognize that some of
the most common dystonia genes are dominant but inherited with reduced
penetrance (eg, GCH1, TOR1A, THAP1). This phenomenon may make
inheritance patterns difficult to recognize. In addition, considerable phenotypic
variation can occur in individual members of the same family with the same
genetic defect. For example, individual members of the same family can have
severe childhood-onset generalized dystonia, adolescent-onset segmental
dystonia, or adult-onset focal dystonia. This phenomenon can make it
Pattern of
Inheritance Genes
Autosomal ANO3, ATP1A3, CIZ1, GCH1, GLUT1, GNAL, PNKD, PRRT2, SCA3, SGCE,
dominant SLC2A1, THAP1, TOR1A, TUBB4
Autosomal AADC, ATP7B, COL6A3, CYP27A1, GCDH, HPCA, KMT2B, NPC1 or NPC2,
recessive PCCA or PCCB, PLA2G6, PRKRA, SLC19A3, SLC39A14, SPR, TH
a
These patterns can be challenging to recognize when families are smaller and because several dominantly
inherited dystonia genes have partial penetrance.
Dopamine signaling GCH1, TH, SPR, PTPS, AADC, VMAT2, PARKIN, PINK1, HPRT, GNAL
a
Dystonia genes relate to numerous biological processes, and only a few of the main genes are shown.
CONTINUUMJOURNAL.COM 985
transcription, degenerative disorders, and others. It seems unlikely that all types
of dystonia are caused by a single molecular or cellular process. Instead, it is more
likely that these processes converge to affect specific neuroanatomic pathways
responsible for dystonia.
Acquired processes that may cause dystonia include vascular, infectious,
immunologic, or structural; drugs or toxins may also cause dystonia. Two
acquired forms of dystonia are especially important to recognize. The first is
tardive dystonia caused by dopamine-receptor–blocking drugs such as
neuroleptics or metoclopramide. These drugs more commonly provoke
repetitive oral and lingual movements of tardive dyskinesia. However, some
patients develop predominantly dystonic manifestations. The most common
patterns involve the craniocervical regions or backward arching of the trunk or
neck. The second acquired form of dystonia is functional (psychogenic)
dystonia, which can closely mimic organic dystonia. Features suggestive of
functional dystonia include nonpatterned movements (tendency to change body
regions or primary muscles involved over time), abrupt onset, attenuation with
distraction, or other unusual accompanying features (CASE 5-1).
Neuroanatomic Basis
Historically, abnormalities in the basal ganglia have been considered the cause of
all forms of dystonia.33 In fact, very good evidence suggests that the defects in the
basal ganglia can cause dystonia. One example is dopa-responsive dystonia,
which can be caused by inherited defects that affect the production of dopamine
COMMENT This patient may have functional (psychogenic) dystonia because of the
abrupt onset, nonpatterned nature of her movements over weeks and
even over the course of the examination, and distractibility.
Neuromodulation surgery should not be recommended for functional
dystonia. Although oral medications can sometimes help at least
transiently, best results are obtained with multidisciplinary care that
involves counseling and physical therapy.
TREATMENT
It is not feasible to use a universal treatment algorithm for all types of dystonia
because so many different subtypes exist. However, some general principles
are useful. As described above, a careful diagnostic evaluation is an essential
starting point because treatments are available to target the causal mechanisms
for some subtypes (TABLE 5-5).25 Other management options include counseling,
physical therapy, oral medications, botulinum neurotoxin (BoNT) injections,
and neurosurgical procedures.
Counseling
Counseling is an important starting place in management. Psychiatric
comorbidities are common, including depression, anxiety, and social
withdrawal.9 Many patients are misdiagnosed for years, leading to frustration
and mistrust of medical providers. Counseling is important for identifying any
comorbidities and regaining trust.
In addition, it is important to recognize that few therapies are curative for
dystonia. Most therapies are symptomatic, and the best outcomes are often
achieved with an empirical trial-and-error approach, which takes time and can
be frustrating. An early and frank discussion that sets realistic expectations for
treatment is essential. In addition, the Useful Websites section at the end of
this article lists several online resources that patients can turn to for more
information.
CONTINUUMJOURNAL.COM 987
Typical Age at
Disorder Gene Onseta Typical Clinical Featuresb Treatment
Abetalipoproteinemia MTTP Childhood to Progressive ataxia, chorea, Early treatment with vitamin E
(Bassen-Kornzweig early dystonia (often and reduced-fat diet can
syndrome) adulthood oromandibular), seizures, prevent or reduce symptoms
acanthocytosis, retinitis
pigmentosa, fat
malabsorption syndrome
Aromatic L-amino acid AADC Infancy Motor delay with hypotonia Dopamine agonists and
decarboxylase deficiency and dystonia, oculogyric monoamine oxidase inhibitors
crises, autonomic can partly reverse symptoms in
dysfunction some patients
Ataxia with vitamin E TTPA Childhood to Ataxia, visual loss, Early treatment with vitamin E
deficiency early neuropathy; occasionally can prevent or reduce symptoms
adulthood patients present instead
with dystonia
Biotin-thiamine– SLC19A3 Childhood Encephalopathic crisis Biotin and thiamine can reverse
responsive basal ganglia leading to generalized or prevent symptoms
disorder dystonia
Biotinidase deficiency BTD Infancy Encephalopathy with motor Early treatment with biotin can
delay, dystonia, seizures, prevent or reduce symptoms
visual and auditory
impairment, skin rash
Cerebral folate FLR1, Early childhood Developmental delay, Folinic acid can prevent or
deficiency SLC46A1 to ataxia, dystonia, seizures, reduce symptoms
adolescence and neuropsychiatric
disturbances
Cerebrotendinous CYP27A1 Late childhood Ataxia, spasticity, dementia, Chenodeoxycholic acid may
xanthomatosis to adulthood dystonia, myoclonus, and prevent progression or reverse
tendon xanthomas some symptoms
Cobalamin deficiencies Multiple Infancy Encephalopathy with motor Cobalamin derivatives or protein
(inherited subtypes A delay, ataxia, spasticity, restriction or both can mitigate
through G) dystonia, seizures, and bone symptoms
marrow abnormalities
Coenzyme Q10 deficiency Multiple Any age Varied phenotypes of Coenzyme Q10 can prevent or
progressive ataxia or reduce symptoms
encephalopathy,
sometimes with dystonia
Cerebral creatine GAMT, Infancy Global delay, myopathy, Creatine with or without arginine
deficiency type 3 AGAT generalized dystonia restriction can mitigate
symptoms
Dopa-responsive GCH1 Early childhood Dystonia often combined Levodopa can reverse
dystonia, classic to late with parkinsonism symptoms
adulthood
Typical Age at
Disorder Gene Onseta Typical Clinical Featuresb Treatment
Dopa-responsive TH, PTPS, Infancy to Dystonia often combined Levodopa, 5-hydroxytryptophan,
dystonia, complicated SPR adolescence with parkinsonism, or tetrahydrobiopterin or a
oculogyric crises, and combination of them can
autonomic disturbances completely or partly reverse
symptoms
Dystonia with brain SLC30A10, Childhood Progressive dystonia with Chelation therapy can prevent or
manganese accumulation SLC39A14 parkinsonism, liver disease, at least partly reverse symptoms
and polycythemia
Galactosemia GALT, Childhood to Ataxia and tremor, lactose Lactose restriction can prevent
GALK1, early intolerance, sometimes with or mitigate symptoms
GALE adulthood mild dystonia
Glucose transporter type SLC2A1 Childhood to Developmental delay, Ketogenic diet or triheptanoin
1 deficiency adolescence seizures; sometimes can prevent or reduce symptoms
paroxysmal exertional
dystonia
Glutaric aciduria type 1 GCDH Early childhood Developmental delay with Avoiding or treating intercurrent
to early encephalopathic crisis illness with lysine restriction can
adulthood leading to generalized prevent encephalopathic crises
dystonia
Maple syrup urine disease BCKDHA, Childhood Intermittent Leucine restriction with or
BCKDHB, encephalopathy and ataxia; without thiamine can prevent or
DBT sometimes with focal or mitigate symptoms
paroxysmal dystonia
Methylmalonic aciduria MUT Childhood Developmental delay, renal Avoiding or treating intercurrent
insufficiency, pancytopenia, illness with protein restriction
generalized dystonia after can prevent encephalopathic
encephalopathic crisis crises
Niemann-Pick disease NPC1, NPC2 Early childhood Dementia, ataxia, spasticity, N-butyl-deoxynojirimycin can
type C to early seizures, supranuclear gaze prevent or mitigate some
adulthood palsy; sometimes with symptoms
progressive generalized
dystonia
Propionic aciduria PCCA, Early childhood Developmental delay with Avoiding or treating intercurrent
PCCB to generalized dystonia after illness with protein restriction
adolescence encephalopathic crisis can prevent encephalopathic
crises
CONTINUUMJOURNAL.COM 989
Typical Age at
Disorder Gene Onseta Typical Clinical Featuresb Treatment
Pyruvate dehydrogenase Multiple Infancy Progressive generalized or Thiamine, ketogenic diet, and
deficiency paroxysmal dystonia dichloroacetate can mitigate
symptoms
Rapid-onset dystonia- ATP1A3 Early childhood Psychomotor delay with Avoiding or treating intercurrent
parkinsonism to late bulbar or generalized illness can prevent
adulthood dystonia after encephalopathic crises
encephalopathic crisis
Wilson disease ATP7B Early childhood Liver disease, Kayser- Zinc or tetrathiomolybdate can
to late Fleischer rings, progressive prevent or reduce symptoms
adulthood dystonia, cognitive and
neuropsychiatric
abnormalities
a
For most childhood-onset disorders, rarely patients may present instead in adulthood.
b
Partial phenotypes are common.
Medication Class
and Example Indications Typical Dosing Common Side Effects
Anticholinergic
Trihexyphenidyl Any dystonia Start with 1 mg to 2 mg each night at Impaired mentation, dry mouth,
bedtime and increase by 1 mg to 2 mg dry eyes, constipation, urinary
every 2–7 days to maximum of 100 mg/d retention, blurry vision
in 3–4 divided doses
Dopaminergic
Carbidopa/ All childhood or early- Start with a half to 1 tablet of 25 mg/ Nausea, orthostasis, sleep
levodopa adult onset dystonia 100 mg and increase every 2–7 days to a disturbance
maximum levodopa dose of 20 mg/kg/d
in 3–4 divided doses
γ-Aminobutyric acid–mediated
Clonazepam Any dystonia Start with 0.5 mg to 1.0 mg each night at Impaired mentation or
bedtime and increase by 0.5 mg to 1 mg coordination, drowsiness,
every 2–7 days to maximum of 6 mg/d fatigue, withdrawal reactions
in 3–4 divided doses
Oral Medications
Many different oral medications are offered to patients with dystonia40,41; no
medications are approved for the treatment of dystonia by the US Food and Drug
Administration (FDA), so all uses are off-label. No medication has been subject
to large-scale, double-blinded, placebo-controlled trials. Although evidence-
based reviews have been published, the use of oral medications is based largely
on anecdotal experience and a few small nonblinded trials, retrospective reviews,
and expert consensus (TABLE 5-6).
CONTINUUMJOURNAL.COM 991
COMMENT This case emphasizes the importance of a levodopa trial in all patients with
early-onset dystonia, even those with a long-standing diagnosis of
“cerebral palsy.” Cerebral palsy does not typically worsen over time, and
the brain MRI often shows characteristic changes due to hypoxia-ischemia.
Despite many years of disability, levodopa can still produce dramatic
benefits in dopa-responsive dystonia. The most common error is to conduct
a levodopa trial with a very small dose. Current recommendations are to
push the levodopa dose to 20 mg/kg/d before concluding the trial failed.
CONTINUUMJOURNAL.COM 993
diplopia, and dry eyes. For laryngeal dystonia, the most common side effect is a
hoarse voice.
Systemic side effects are uncommon. Some patients describe a flulike
syndrome for 3 to 5 days after their treatments.65 A recent study has raised
concern for a high prevalence of antibodies to BoNT in treated patients.66
However, the patients in this study were all actively being treated with BoNT,
with typical good therapeutic responses, so the clinical significance of these
antibodies remains unclear. The development of true immune-mediated
functional resistance to BoNT is rare, so alternative explanations should be
sought when patients do not respond, before ascribing poor outcomes
to antibodies.67,68
Surgical Procedures
Multiple surgical options are available for the treatment of dystonia when more
conservative therapies fail. Currently, the most commonly offered procedure
involves neuromodulation of brain activity via deep brain stimulation. Ablative
procedures involving select brain regions or peripheral targets are applied in
some circumstances.
CONTINUUMJOURNAL.COM 995
CONCLUSION
In the past decade, there have been enormous strides in the appreciation of the
many clinical manifestations of the dystonias, how these manifestations should
be classified for optimal diagnostic and therapeutic value, their underlying
biological mechanisms, and how they should be treated. All types of dystonia are
treatable at least symptomatically, and several have treatments that target
underlying mechanisms. As our understanding of mechanisms continues to
evolve, it is likely that the number of dystonic disorders with more specific
treatments will continue to grow.
ACKNOWLEDGMENTS
Special appreciation goes to Laura Scorr, MD, for providing feedback on this
review. Dr Jinnah has received grant support from the National Institutes of
Health (NIH) and is principal investigator for the Dystonia Coalition, which
receives support through NIH grant TR001456 from the Office of Rare Diseases
Research at the National Center for Advancing Translational Sciences, and
previously received support through grant NS065701 from the National
Institutes of Neurological Disorders and Stroke.
USEFUL WEBSITES
DYSTONIA COALITION
NATIONAL SPASMODIC TORTICOLLIS ASSOCIATION
The Dystonia Coalition has researchers and
The National Spasmodic Torticollis Association
advocacy groups in the field working together to
provides information on the signs and symptoms
further research about dystonias.
and treatment options of cervical dystonia.
rarediseasesnetwork.org/cms/dystonia
torticollis.org
DYSTONIA MEDICAL RESEARCH FOUNDATION
The Dystonia Medical Research Foundation provides
support to people living with all types of dystonia.
dystonia-foundation.org
1 Albanese A, Bhatia K, Bressman SB, et al. 15 Ludlow CL, Domangue R, Sharma D, et al.
Phenomenology and classification of dystonia: Consensus-based attributes for identifying
a consensus update. Mov Disord 2013;28(7): patients with spasmodic dysphonia and other
863–873. doi:10.1002/mds.25475. voice disorders. JAMA Otolaryngol Head Neck
Surg 2018;144(8):657–665. doi: 10.1001/
2 Fung VS, Jinnah HA, Bhatia K, Vidailhet M.
jamaoto.2018.0644.
Assessment of patients with isolated or
combined dystonia: an update on dystonia 16 Jinnah HA, Ceballos-Picot I, Torres RJ, et al.
syndromes. Mov Disord 2013;28(7):889–898. Attenuated variants of Lesch-Nyhan disease. Brain
doi:10.1002/mds.25549. 2010;133(pt 3):671–689. doi:10.1093/brain/awq013.
3 Powell AT, Bidewell JW, Walker AC. Diagnosing 17 Jinnah HA, Visser JE, Harris JC, et al. Delineation of
idiopathic dystonia: must it take so long? Aust the motor disorder of Lesch-Nyhan disease. Brain
Health Rev 1995;18(3):120–131. 2006;129(pt 5):1201–1217. doi:10.1093/brain/awl056.
4 Jog M, Chouinard S, Hobson D, et al. Causes for 18 Fu R, Ceballos-Picot I, Torres RJ, et al.
treatment delays in dystonia and hemifacial Genotype-phenotype correlations in
spasm: a Canadian survey. Can J Neurol Sci 2011; neurogenetics: Lesch-Nyhan disease as a model
38(5):704–711. doi:10.1017/S0317167100012270. disorder. Brain 2013;137(pt 5):1282–1303. doi:
10.1093/brain/awt202.
5 Macerollo A, Superbo M, Gigante AF, et al.
Diagnostic delay in adult-onset dystonia: data 19 Levy A, Lange AE. Ataxia-telangiectasia: a review
from an Italian movement disorder center. J Clin of movement disorders, clinical features, and
Neurosci 2015;22(3):608–610. doi:10.1016/j. genotype correlations. Mov Disord 2018;33(8):
jocn.2014.09.014. 1238–1247. doi:10.1002/mds.27319.
6 Tiderington E, Goodman EM, Rosen AR, et al. 20 Zech M, Jech R, Wagner M, et al. Molecular
How long does it take to diagnose cervical diversity of combined and complex dystonia:
dystonia? J Neurol Sci 2013;335(1–2):72–74. insights from diagnostic exome sequencing.
doi:10.1016/j.jns.2013.08.028. Neurogenetics 2017;18(4):195–205. doi:10.1007/
s10048-017-0521-9.
7 Creighton FX, Hapner E, Klein A, et al. Diagnostic
delays in spasmodic dysphonia: a call for 21 van Egmond ME, Lugtenberg CHA, Brouwer OF,
clinician education. J Voice 2015;29(5):592–594. et al. A post hoc study on gene panel analysis for
doi:10.1016/j.jvoice.2013.10.0228. the diagnosis of dystonia. Mov Disord 2017;32(4):
569–575. doi:10.1002/mds.26937.
8 Patel N, Hanfelt J, Marsh L, et al. Alleviating
manoeuvres (sensory tricks) in cervical dystonia. 22 Lazaridis KN, Schahl KA, Cousin MA, et al.
J Neurol Neurosurg Psychiatry 2014;85(8): Outcome of whole exome sequencing for
882–884. doi:10.1136/jnnp-2013-307316. diagnostic odyssey cases of an individualized
medicine clinic: the Mayo Clinic experience.
9 Zurowski M, McDonald W, Fox S, Marsh L.
Mayo Clin Proc 2016;91(3):297–307. doi:10.1016/j.
Psychiatric comorbidities in dystonia: emerging
mayocp.2015.12.018.
concepts. Mov Disord 2013;28(7):914–920.
doi:10.1002/mds.25501. 23 Sawyer SL, Hartley T, Dyment DA, et al. Utility of
whole-exome sequencing for those near the
10 Rossi M, Balint B, Millar Vernetti P, et al.Genetic
end of the diagnostic odyssey: time to address
dystonia-ataxia syndromes: clinical spectrum,
gaps in care. Clin Genet 2016;89(3):275–284.
diagnostic approach, and treatment options.
doi:10.1111/cge.12654.
Mov Disord Clin Pract 2018;5(4):373–382. doi:
10.1002/mdc3.12635. 24 Johnson NE. Whole-exome sequencing in
neurologic practice: reducing the diagnostic
11 van Egmond ME, Kuiper A, Eggink H, et al.
odyssey. Neurol Genet 2015;1(4):e37. doi:10.1212/
Dystonia in children and adolescents: a
NXG.0000000000000037.
systematic review and a new diagnostic
algorithm. J Neurol Neurosurg Psychiatry 2014; 25 Jinnah HA, Albanese A, Bhatia KP, et al. Treatable
86(7):774–781. doi:10.1136/jnnp-2014-309106. inherited rare movement disorders. Mov Disord
2018;33(1):21–35. doi:10.1002/mds.27140.
12 Charlesworth G, Bhatia KP, Wood NW. The genetics
of dystonia: new twists in an old tale. Brain 2013; 26 Wickremaratchi MM, Knipe MD, Sastry BS, et al.
136(pt 7):2017–2037. doi:10.1093/brain/awt138. The motor phenotype of Parkinson's disease in
relation to age at onset. Mov Disord 2011;26(3):
13 Camargos S, Cardoso F. New algorithm for
457–463. doi:10.1002/mds.23469.
the diagnosis of hereditary dystonia.
Arq Neuropsiquiatr 2012;70(9):715–717. 27 Yoon WT. Comparison of dystonia between
doi:10.1590/S0004-282X2012000900013. Parkinson's disease and atypical parkinsonism:
the clinical usefulness of dystonia distribution
14 Bertram KL, Williams DR. Diagnosis of dystonic
and characteristics in the differential diagnosis
syndromes—a new eight-question approach.
of parkinsonism. Neurol Neurochir Pol 2018;52(1):
Nat Rev Neurol 2012;8(5):275–283. doi:10.1038/
48–53. doi:10.1016/j.pjnns.2017.11.004.
nrneurol.2012.39.
CONTINUUMJOURNAL.COM 997
28 Schneider SA, Bhatia KP, Hardy J. Complicated 43 Lee WW, Jeon BS. Clinical spectrum of
recessive dystonia parkinsonism syndromes. dopa-responsive dystonia and related disorders.
Mov Disord 2009;24(4):490–499. doi:10.1002/ Curr Neurol Neurosci Rep 2014;14(7):461.
mds.22314. doi:10.1007/s11910-014-0461-9.
29 Marras C, Lang A, van de Warrenburg BP, et al. 44 Kurian MA, Gissen P, Smith M, et al. The
Nomenclature of genetic movement disorders: monoamine neurotransmitter disorders: an
recommendations of the International Parkinson expanding range of neurological syndromes.
and Movement Disorder Society Task Force. Mov Lancet Neurol 2011;10(8):721–733. doi:10.1016/
Disord 2016;31(4):436–457. doi:10.1002/mds.26527. S1474-4422(11)70141-7.
30 Weisheit CE, Pappas SS, Dauer WT. Inherited 45 Charlesworth G, Mohire MD, Schneider SA, et al.
dystonias: clinical features and molecular Ataxia telangiectasia presenting as dopa-
pathways. Handb Clin Neurol 2018;147:241–254. responsive cervical dystonia. Neurology 2013;
doi:10.1016/B978-0-444-63233-3.00016-6. 81(13):1148–1151. doi:10.1212/WNL.0b013e3182a55fa2.
31 Balint B, Mencacci NE, Valente EM, et al. 46 Wilder-Smith E, Tan EK, Law HY, et al.
Dystonia. Nat Rev Dis Primers 2018;4(1):25. Spinocerebellar ataxia type 3 presenting as an
doi:10.1038/s41572-018-0023-6. L-DOPA responsive dystonia phenotype in a
Chinese family. J Neurol Sci 2003;213(1–2):25–28.
32 Quartarone A, Hallett M. Emerging concepts in
doi:10.1016/S0022-510X(03)00129-1.
the physiological basis of dystonia. Mov Disord
2013;28(7):958–967. doi:10.1002/mds.25532. 47 Greene P, Shale H, Fahn S. Experience with high
dosages of anticholinergic and other drugs in the
33 Neychev VK, Gross RE, Lehéricy S, et al.The
treatment of torsion dystonia. Adv Neurol 1988;
functional neuroanatomy of dystonia.
50:547–556.
Neurobiol Dis 2011;42(2):185–201. doi:10.1016/
j.nbd.2011.01.026. 48 Fehlings D, Brown L, Harvey A, et al.
Pharmacological and neurosurgical interventions
34 Prudente CN, Hess EJ, Jinnah HA. Dystonia as a
for managing dystonia in cerebral palsy: a
network disorder: what is the role of the
systematic review. Dev Med Child Neurol 2018;
cerebellum? Neuroscience 2014;260:23–35.
60(4):356–366. doi:10.1111/dmcn.13652.
doi:10.1016/j.neuroscience.2013.11.062.
49 Harvey AR, Baker LB, Reddihough DS, et al.
35 Nibbeling EA, Delnooz CC, de Koning TJ, et al.
Trihexyphenidyl for dystonia in cerebral palsy.
Using the shared genetics of dystonia and ataxia
Cochrane Database Syst Rev 2018;5: CD012430.
to unravel their pathogenesis. Neurosci Biobehav
doi:10.1002/14651858.CD012430.pub2.
Rev 2017;75:22–39. doi:10.1016/j.neubiorev.
2017.01.033. 50 Houser MK, Soland VL, Bhatia KP, et al. Paroxysmal
kinesigenic choreoathetosis: a report of 26
36 Jinnah HA, Neychev V, Hess EJ. The anatomical
patients. J Neurol 1999;246(2):120–126. doi:10.1007/
basis for dystonia: the motor network model.
s004150050318.
Tremor Other Hyperkinet Mov (N Y) 2017;7:506.
doi:10.7916/D8V69X3S. 51 Demirkiran M, Jankovic J. Paroxysmal dyskinesias:
clinical features and classification. Ann Neurol 1995;
37 Shaikh AG, Zee DS, Crawford JD, Jinnah HA.
38(4):571–579. doi:10.1002/ana.410380405.
Cervical dystonia: a neural integrator disorder.
Brain 2017;139(pt 10):2590–2599. doi:10.1093/ 52 Simpson DM, Hallett M, Ashman EJ, et al.
brain/aww141. Practice guideline update summary: botulinum
neurotoxin for the treatment of blepharospasm,
38 Hutchinson M, Isa T, Molloy A, et al. Cervical
cervical dystonia, adult spasticity, and headache:
dystonia: a disorder of the midbrain network for
report of the Guideline Development
covert attentional orienting. Front Neurol 2014;
Subcommittee of the American Academy of
5:54. doi:10.3389/fneur.2014.00054.
Neurology. Neurology 2016;86(19):1818–1826.
39 Prudente CN, Zetterberg L, Bring A, et al. doi:10.1212/WNL.0000000000002560.
Systematic review of rehabilitation in focal
53 Castelão M, Marques RE, Duarte GS, et al.
dystonias: classification and recommendations.
Botulinum toxin type A therapy for cervical
Mov Disord Clin Pract 2018;5(3):237–245.
dystonia. Cochrane Database Syst Rev 2017;12:
doi:10.1002/mdc3.12574.
CD003633. doi:10.1002/14651858.CD003633.pub3.
40 Pirio Richardson S, Wegele AR, Skipper B, et al.
54 Hallett M, Albanese A, Dressler D, et al.
Dystonia treatment: patterns of medication use
Evidence-based review and assessment of
in an international cohort. Neurology 2017;88(6):
botulinum neurotoxin for the treatment of
1–8. doi:10.1212/WNL.0000000000003596.
movement disorders. Toxicon 2013;67:94–114.
41 Jinnah HA, Factor S. Diagnosis and treatment of doi:10.1016/j.toxicon.2012.12.004.
dystonia. In: Jankovic J, editor. Neurologic clinics.
55 Truong D, Dressler D, Hallet M, Zachary C. Manual
Elsevier, 2015:77–100. doi:10.1016/j.ncl.
of botulinum toxin. Cambridge: Cambridge
2014.09.002.
University Press, 2009.
42 Wijemanne S, Jankovic J. Dopa-responsive
56 Jost W, Valerius KP. Pictorial atlas of botulinum
dystonia—clinical and genetic heterogeneity.
toxin injection: dosage, localization, application.
Nat Rev Neurol 2015;11(7):414–424. doi:10.1038/
Berlin: Quintessence Publishing Company, 2008.
nrneurol.2015.86.
CONTINUUMJOURNAL.COM 999
DISCLOSURE
Continued from page 976 ethopropazine, gabapentin, lithium, metaxalone,
methocarbamol, nabilone orphenadrine,
Beat Dystonia, the Benign Essential Blepharospasm procyclidine, riluzole, tizanidine, trihexyphenidyl,
Research Foundation, Cure Dystonia Now, Dystonia and zolpidem for the treatment of dystonia.
Europe, Dystonia Inc, Dystonia Ireland, the Dystonia Dr Jinnah discusses the unlabeled/investigational
Medical Research Foundation, the Foundation for use of thiamine for the treatment of biotin-
Dystonia Research, the National Spasmodic thiamine–responsive basal ganglia disorder;
Dysphonia Association, and the National Spasmodic folinic acid for the treatment of cerebral folate
Torticollis Association). deficiency; chenodeoxycholic acid for the
treatment of cerebrotendinous xanthomatosis;
UNLABELED USE OF 5-hydroxytryptophan and tetrahydrobiopterin for
PRODUCTS/INVESTIGATIONAL the treatment of dopa-responsive dystonia;
USE DISCLOSURE: cyclic pyranopterin monophosphate for the
Dr Jinnah discusses the unlabeled/investigational treatment of molybdenum cofactor deficiency;
use of alprazolam, amphetamines, baclofen, N-butyl-deoxynojirimycin for the treatment of
benzodiazepines, benztropine, biperiden, Niemann-Pick disease type C; tetrabenazine for
botulinum neurotoxins, carbamazepine, the treatment of oromandibular dystonia; and
carbidopa/levodopa, carisoprodol, deutetrabenazine, tetrabenazine, and
chlordiazepoxide, chlorzoxazone, clonazepam, valbenazine and for the treatment of tardive
cyclobenzaprine, cyproheptadine, diazepam, dystonia.
By Pichet Termsarasab, MD C O N T I N U UM A U D I O
I NT E R V I E W A V A I L AB L E
ONLINE
ABSTRACT VIDEO CONTENT
PURPOSE OF REVIEW: Thisarticle provides an overview of the approach A V AI L A B L E O N L I N E
UNLABELED USE OF
PRODUCTS/INVESTIGATIONAL
USE DISCLOSURE:
INTRODUCTION
Dr Termsarasab discusses the
C
horea is derived from the Greek word χορεία, meaning dance. unlabeled/investigational
It is characterized by the random and flowing quality of the use of the current
recommended treatments of
movements, giving it a dancelike appearance. Randomness is the chorea, none of which are
key phenomenologic feature in the identification of chorea. The approved by the US Food and
movements typically flit from one body region to another in an Drug Administration except the
use of deutetrabenazine for the
unpredictable fashion. treatment of chorea associated
The differential diagnosis of chorea is broad. However, with clinical features with Huntington disease and
including demographic data, time course, associated medical and neurologic tardive dyskinesia,
tetrabenazine for the treatment
features, and known prevalence, the search for the etiology of chorea can be of chorea associated with
performed efficiently. A “shotgun approach” can be reserved for when no diagnostic Huntington disease, and
valbenazine for the treatment of
clues are present. This article discusses the general diagnostic approach to chorea,
tardive dyskinesia.
clinical clues to common and important choreic disorders, and therapeutic principles.
The first section of the article discusses general phenomenologic features of
chorea and how to differentiate chorea from other movement disorders. The next © 2019 American Academy
section covers the general approach to chorea, beginning with the three body of Neurology.
CONTINUUMJOURNAL.COM 1001
APPROACH TO CHOREA
Given an extensive differential diagnosis, chorea can be challenging to many
clinicians. Nevertheless, there are some important clinical features that can
serve as diagnostic clues to the specific diagnosis. These include three body
distributions and other crucial features.
FIGURE 6-1
Body distribution as a phenomenologic clue in chorea. The differential diagnoses are
demonstrated in each distribution.
PKAN = pantothenate kinase–associated neurodegeneration.
a
Locations outside the subthalamic nucleus can also be involved.
CONTINUUMJOURNAL.COM 1003
CASE 6-1 A 77-year-old woman presented with abnormal movements of her left
arm, left leg, and left face. She had a history of type 2 diabetes mellitus
and poor compliance. She had run out of her medications and not taken
them for 2 weeks before developing abnormal flinging movements of her
left arm upon waking up in the morning, followed a few days later by
movements in her left leg and left face. The movements had gradually
become worse over the week, prompting her to seek medical attention.
On presentation to the hospital, examination revealed left
hemichorea/hemiballism involving her left arm, left leg, and left face
(VIDEO 6-2, links.lww.com/CONT/A352). The chorea abated during sleep.
Blood testing on presentation showed a glucose level of 445 mg/dL and
hemoglobin A1c of 12%. She had no ketosis.
The diagnosis of left hemichorea/hemiballism secondary to nonketotic
hyperglycemia was made, which was initially managed by IV and
subcutaneous insulin. Her oral diabetic medications were resumed, with
improvement in her blood glucose level. MRI of the brain revealed a
hyperintense signal on T1-weighted images in the right (contralateral)
putamen (FIGURE 6-2A). A faint hyperdense signal could also be seen on a
CT scan without contrast in the same region (FIGURE 6-2B).
Her chorea was not adequately improved with blood glucose control
alone, and haloperidol was started at 0.5 mg/d with about 50%
improvement in her hemichorea. The dose was then increased to 0.5 mg
2 times a day with further improvement. She required low-dose
haloperidol for several weeks before the chorea subsided.
COMMENT The acute temporal profile in this patient is suggestive of an acquired cause
of chorea. Hemichorea/hemiballism can be a manifestation of systemic
disorders such as nonketotic hyperglycemia or polycythemia vera.
Nonketotic hyperglycemia is common among Asian populations, especially
women (this patient was from Thailand). Blood glucose control serves as a
specific treatment, but most patients will also need symptomatic
therapies, as chorea may take several weeks or months to resolve.
FIGURE 6-2
Imaging of the patient in CASE 6-1 with nonketotic hyperglycemia. A, Axial T1-weighted MRI
shows hyperintense signal in the right putamen (red arrow), contralateral to the side of
hemichorea. B, CT scan of the same patient demonstrates mild hyperdensity in the same
region (white arrow).
CONTINUUMJOURNAL.COM 1005
DIAGNOSTIC APPROACH
Useful diagnostic clues include demographic data (eg, age, gender, and
ethnicity); family history; coexisting movement phenomenology such as
dystonia, myoclonus, or tics; and associated clinical features, such as seizures,
neuropathy, and myopathy. Given the broad differential diagnosis of chorea, the
clinical approach can be challenging to clinicians. A proposed practical clinical
approach is illustrated in FIGURE 6-3. The three most crucial features are time
course, age group, and known prevalence.
As a general rule, acquired or sporadic causes of chorea usually present with
an acute or subacute temporal profile, whereas genetic causes are chronic
(longer than 1 year in duration). It is crucial to identify acquired causes, as many
of them are treatable. Autoimmune etiologies should always be included in the
differential diagnosis, especially in subacute presentations; these disorders
typically respond to immunotherapies, including IV immunoglobulin (IVIg),
steroids, other immunosuppressive agents, and plasma exchange. In recent
years, the number of autoimmune neurologic disorders has been expanding
with advances in neuroimmunology and identification of novel autoantibodies.
CONTINUUMJOURNAL.COM 1007
Category Examples
Tumors
Demyelinating lesions
Hypoglycemia
Hypocalcemia
Hyponatremia/hypernatremia
Uremia
Hyperthyroidism
Hypoparathyroidism/hyperparathyroidism
Infectious Toxoplasmosis
HIV encephalopathy
Prion diseases
Drug-induced Levodopa
Cocaine (“crack-dancing”)
Amphetamine
Anticonvulsants
Lithium
Anticholinergics
Neuroleptic withdrawal
Postpump chorea
CONTINUUMJOURNAL.COM 1009
Sydenham chorea is still the most common cause of chorea in the pediatric
population, after choreoathetoid cerebral palsy. Patients typically develop
chorea 2 to 3 months after group A β-hemolytic streptococcal throat infection.
Therefore, when chorea is already present, throat culture is usually negative
and not helpful. Furthermore, antibody testing usually performed in clinical
practice (including antistreptolysin O [ASO] and antideoxyribonuclease B [anti-
DNase B]), although having higher diagnostic yields than throat culture, does not
have high sensitivity. In addition to chorea, patients may have tics and
neurobehavioral features, such as anxiety, irritability, attention deficit
hyperactivity disorder, and obsessive-compulsive behaviors, that can sometimes
be disruptive. Thus, the term Sydenham disease has been proposed to cover the
entire spectrum of its clinical features. Some patients have severe hypotonia
along with florid chorea, called chorea paralytica.
Recognition of Sydenham chorea is important, since patients will require a cardiac
workup, particularly echocardiography, and secondary antibiotic prophylaxis
with long-term penicillin, the duration of which depends on the severity of
carditis. Treatment of Sydenham chorea is discussed further in the treatment
section of this article. Sydenham chorea can recur in 20% to 50% of patients, and
persistent symptoms after 2-year follow-up have been reported in up to 50%.18
Chorea gravidarum refers to chorea that initially emerges, reemerges, or
exacerbates during pregnancy. It is a descriptive terminology rather than specific
diagnostic entity, as underlying etiologies such as HD, systemic lupus
erythematosus, antiphospholipid antibody syndrome, and hyperthyroidism should
always be sought. Patients with a history of Sydenham chorea in childhood can have
reemergence of chorea during pregnancy. However, thorough investigations for
other potential underlying causes of chorea in these patients should not be
neglected. Another hormonal-related chorea is chorea that may emerge or reemerge
during the use of oral contraceptive pills, and the same principles should be applied.
Chorea gravidarum can improve spontaneously in the third trimester or shortly
after the patient gives birth,19 and treatment may not be required. Nevertheless, the
most crucial “don’t-miss” step is searching for an underlying etiology.
The workup for acquired causes of chorea can be extensive. If diagnostic clues
are present, a targeted approach can be pursued. In clinical practice, if no
diagnostic clues are present, the first-tier testing includes complete blood cell
count, thyroid function tests, liver function tests, serum electrolytes, serum
calcium, antinuclear antibody, anti–double-stranded DNA antibody, lupus
anticoagulant, and antiphospholipid antibody syndrome workup. When patients
present with a subacute temporal profile, antibody testing in both serum and CSF
should be considered in addition to routine CSF studies.
Huntington Disease
HD is by far the most common genetic chorea in adults (TABLE 6-2). Geographic
variability is seen, with higher prevalence in some regions because of founder
effects, such as in Venezuela around Lake Maracaibo. Clinical features can be
grouped into three major categories: movement disorders, cognitive impairment,
Another eye movement abnormality in HD is an impaired antisaccade task. This ● Delayed initiation of
can be seen by asking the patient to look to the side contralateral to the side on saccades is a hallmark eye
which the examiner is holding up a finger. Patients with HD tend to look to the movement abnormality in
ipsilateral side, reflecting frontal disinhibition. Huntington disease.
HD is an autosomal dominant disorder due to CAG repeat expansion in the
● Senile chorea should not
HTT (also known as IT15) gene on chromosome 4p encoding the huntingtin be used as a diagnosis, and
protein. Individuals with 40 or more CAG repeats have complete penetrance an underlying etiology
(FIGURE 6-4), with an inverse correlation between the number of repeats and the should be sought.
age at onset. Individuals with between 36 and 39 CAG repeats are manifesting
● Children with Huntington
carriers with incomplete penetrance; not all individuals with CAG repeats in this disease typically do not
range will develop HD manifestations during their lifetime. Patients with lower present with chorea but
numbers of repeats tend to have presentation of chorea in late life, which has rather parkinsonism,
been called senile chorea. However, this term is considered obsolete and should dystonia, and seizures.
not be used as a diagnosis. A careful search for underlying etiologies, including ● Age at onset in
HD, should be conducted. Generally, individuals with fewer than 36 CAG repeats Huntington disease is
(35 or fewer) will not manifest the symptoms or signs of HD. Nevertheless, rare determined by the number
case reports exist of manifest HD with an intermediate number (27 to 35) of CAG of CAG repeats, genetic
modifiers, and
repeats. Anticipation tends to occur when unstable CAG repeats are inherited
environmental factors.
from the father, because of CAG-repeat instability during spermatogenesis.
Children with manifest HD typically have 50 to 60 CAG repeats or more. In
contrast to the adult-onset form, juvenile HD (onset at younger than 20 years of
age) typically presents with parkinsonism (also known as the Westphal variant)
and dystonia rather than chorea, as well as seizures. Nevertheless, not only the
number of CAG repeats but also genetic modifiers and environmental factors
contribute to the age at onset24; therefore, the age at onset in an individual cannot
be accurately predicted exclusively from the number of CAG repeats.
CONTINUUMJOURNAL.COM 1011
C9orf72 disease Autosomal GGGGCC repeat Phenotypic variability: some Recently found
dominant expansion in C9orf72 patients have frontotemporal to be the most
gene (C9orf72) dementia-amyotrophic lateral common cause
sclerosis of Huntington
disease
Pyramidal features (hyperreflexia)
phenocopies
SCA17 (HDL-4) Autosomal TBP (TATA-box binding Ataxia, dystonia The second most
dominant protein) common cause of
Cognitive impairment,
Huntington
neuropsychiatric features
disease
phenocopies after
C9orf72 disease
CONTINUUMJOURNAL.COM 1013
Neurodegeneration Pantothenate
with brain iron kinase–associated
accumulation neurodegeneration
is rarely reported to
cause chorea
Neuroferritinopathy Autosomal FTL (ferritin light chain) Dystonia with predilection to lower
dominant cranial region
Low serum ferritin (not all cases)
MRI gradient recalled echo
(GRE), or susceptibility-weighted
imaging (SWI): cystic degeneration
in caudate and putamen; “pencil
sign” (cortical lining of iron) has
also been reported
HDL = Huntington disease–like; MRI = magnetic resonance imaging; SCA17 = spinocerebellar ataxia type 17.
a
Modified with permission from Termsarasab P.11 © 2017 American Academy of Neurology.
CONTINUUMJOURNAL.COM 1015
studies and clinical trials for disease-modifying therapies; however, ethical issues
and impact on patients and families should be taken into consideration.
FIGURE 6-4
Relationship between CAG repeat length and Huntington disease phenotype.
Figure courtesy of Thananan Thammongkolchai, MD.
The two main disorders in this group are chorea-acanthocytosis and McLeod
syndrome. Chorea-acanthocytosis is due to mutations in the VPS13A gene
CONTINUUMJOURNAL.COM 1017
FIGURE 6-5
Neuroimaging in choreic disorders. A, Axial T2* MRI shows hypointensity in the bilateral
striatum (red arrows), thalami (yellow arrowheads), and cortical surface (green arrow),
representing iron accumulation in a patient with aceruloplasminemia. B, Axial T2-weighted
MRI shows cystic degeneration of the bilateral basal ganglia (blue arrows) and hyperintense
signal with a hypointense rim at the bilateral thalami (red arrowheads) in a patient with
neuroferritinopathy. C, Axial susceptibility-weighted imaging (SWI) shows “cortical pencil
lining” representing superficial iron accumulation in a patient with neuroferritinopathy.
Panel A reprinted with permission from Fujita K, et al, Neurology.32 © 2013 American Academy of Neurology.
Panel B reprinted with permission from Ohta E, Takiyama Y, Neurol Res Int.33 © 2012 The Authors. Panel C
reprinted with permission from Batla A, et al, Neurology.34 © 2015 American Academy of Neurology.
encoding for chorein. However, because of the large size of the gene with 76 exons,
gene sequencing is not feasible. Using Western blot to detect chorein deficiency is
another diagnostic method. McLeod syndrome is due to mutations in the XK gene
on the X chromosome encoding for the Kx antigen on the surface of red blood
cells. The mutations lead to absent Kx antigen and reduced Kell antigen. Kx is not a
part of the Kell antigen system, but molecular structural interaction between these
two proteins leads to reduction of Kell protein when Kx antigen is absent. The
McLeod phenotype can be discovered in blood banks during blood screening and
can be detected by using anti-Kx and anti-Kell antibodies.
While acanthocytes are a hallmark of these disorders, three caveats are
important to note here. First, acanthocytes are not specific to these two disorders
and can also be seen in others as noted above. These disorders have also been
included under the umbrella of neuroacanthocytosis syndromes.36 Second, a
special technique using a wet unfixed preparation of an isotonically diluted blood
sample, as described in detail by Storch and colleagues,37 is required for higher
sensitivity of acanthocyte detection in peripheral blood smear. Although the yield
of acanthocyte detection from routine peripheral blood smear is lower, it should
still be examined, with an understanding of its limitations. Third, even with the
appropriate technique, acanthocytes can be absent in neuroacanthocytosis
syndromes; thus, the absence of acanthocytes on peripheral blood smear does not
exclude these disorders.
In chorea-acanthocytosis, dystonia has a predilection to involve the lower
cranial region, and feeding dystonia is one of the characteristic features. This can
lead to poor nutritional status. Lip and tongue biting can be due to feeding
dystonia and possibly coexisting obsessive-compulsive behaviors. Intermittent
CONTINUUMJOURNAL.COM 1019
COMMENT Chorea, along with feeding dystonia, seizures, and coexisting neuropathy
or myopathy (evidenced by absent or reduced deep tendon reflexes and
elevated creatine kinase) are very suggestive of chorea-acanthocytosis, an
autosomal recessive form of neuroacanthocytosis syndromes. Although
present in this patient, acanthocytes in the peripheral blood smear are not
always seen even with a special technique using a wet unfixed preparation
of an isotonically diluted blood sample. Delayed initiation of saccades,
similar to that seen in Huntington disease, can be seen in chorea-
acanthocytosis; however, it usually comes in the later stages compared to
Huntington disease. Vertical supranuclear gaze palsy is an unusual feature
in chorea-acanthocytosis. Therapies for choreic disorders are not
restricted to the treatments of chorea. Although specific therapies are not
yet available, and symptomatic treatment is not required if chorea is mild
and nonbothersome, patients such as this one could benefit from
multidisciplinary care by a physical medicine and rehabilitation specialist
and physical, occupational, and speech therapists.
CONTINUUMJOURNAL.COM 1021
Selected Clinical
Pattern of Gene (Protein Clues (Other
Disorder Inheritance Encoded) Than Chorea) Remarks/Caveats
Classic benign Autosomal NKX2-1 (formerly Also called Some may have
hereditary chorea dominant known as TITF-1) brain-lung-thyroid paradoxical
syndrome or gene (thyroid syndrome (or BLT response to
NKX2-1–related transcription factor-1) syndrome) levodopa
benign hereditary
Hypothyroidism and
chorea
pulmonary disease
(eg, respiratory
distress or interstitial
lung disease) can
coexist
Typically
nonprogressive, but
not always benign
(considered
relatively more
benign, compared to
Huntington disease)
Patients may have
developmental
delay or short
stature
Selected Clinical
Pattern of Gene (Protein Clues (Other
Disorder Inheritance Encoded) Than Chorea) Remarks/Caveats
CONTINUUMJOURNAL.COM 1023
Selected Clinical
Pattern of Gene (Protein Clues (Other
Disorder Inheritance Encoded) Than Chorea) Remarks/Caveats
Selected Clinical
Pattern of Gene (Protein Clues (Other
Disorder Inheritance Encoded) Than Chorea) Remarks/Caveats
CSF = cerebrospinal fluid; CT = computed tomography; EMG = electromyography; MELAS = mitochondrial encephalomyopathy, lactic acidosis,
and strokelike episodes; MRI = magnetic resonance imaging; NA = not applicable.
a
Modified with permission from Termsarasab P.11 © 2017 American Academy of Neurology.
CONTINUUMJOURNAL.COM 1025
Other rarer genes related to benign hereditary chorea syndromes are discussed
here only briefly. PDE10A mutations can be either autosomal dominant or
recessive. The dominant forms or de novo mutations typically present with
chorea at around 5 to 10 years of age and show bilateral striatal hyperintense
signals on MRI (hence, also called childhood-onset bilateral striatal necrosis), but
no intellectual impairment is seen.45 The recessive forms are more severe.
Clinical features include infantile-onset chorea and motor and language
developmental delay but, interestingly, no striatal hyperintensity on MRI.46
Gain-of-function mutations in the GNAO1 gene can present with chorea,47
whereas loss-of-function mutations cause Ohtahara syndrome, a form of early
infantile epileptic encephalopathy. Relatively new genes reported in chorea
associated with epilepsy include FOXG1 (in which patients can have prominent
orofacial chorea/dyskinesia),52 GRIN153,54 and FRRS1L.55
CASE 6-4 A 6-year-old boy came to the clinic for follow-up of his choreic disorder.
He was born full term without any prenatal and perinatal complications.
However, during the first 2 years of life, he was diagnosed with
hypothyroidism and “asthma.” Around 4.5 years of age, he was evaluated
in the movement disorder clinic because of abnormal movements
compatible with generalized chorea, which had gradually become more
noticeable in the past 1.5 years. Multiple family members on his maternal
side had thyroid problems and asthma; however, no abnormal movement
was reported in these family members. The diagnosis of benign
hereditary chorea was suspected, and it was confirmed by the mutation
in the NKX2-1 (formerly known as TITF-1) gene. He had mild intellectual
disability and had participated in a special school program. His chorea
had been stable in the past 2 years. He was trialed on levodopa at the age
of 6 years up to 5 mg/kg/d for symptomatic control of chorea. However,
because no improvement was seen, it was subsequently discontinued.
Examination revealed mild to moderate generalized chorea involving
all extremities, trunk, neck, and lower facial region, as well as mild
bilateral foot dystonia demonstrated as mild foot inversion when
walking. The dystonic component was much less prominent than chorea.
Treatment options, including tetrabenazine, were discussed with his
parents.
TREATMENT
The first question before initiating treatment is “Does chorea need to be treated?”
(TABLE 6-4). When chorea is mild and nonbothersome and does not interfere
with a patient’s daily activities, symptomatic treatment is not required. In some
choreic disorders, such as HD, lack of awareness of the deficit by patients as a
possible coexisting neuropsychiatric feature may limit evaluation of chorea
severity solely from patients’ reports. Thus, the clinician’s and family’s
evaluation of chorea severity should also be incorporated into clinical decisions.
When clinicians decide to treat, two main categories of therapies should be
considered: specific therapies and symptomatic therapies (TABLE 6-5).
Specific Therapies
Many acquired choreas are treatable, so correct diagnosis of these “don’t-miss”
disorders is a crucial step before using corresponding specific therapies.
Examples include blood sugar control in nonketotic hyperglycemia, phlebotomy
and hydroxyurea in polycythemia vera, antibiotics in central nervous system
(CNS) infection such as CNS toxoplasmosis, and immunotherapies in
autoimmune choreas, including systemic lupus erythematosus.
Symptomatic Therapies
Symptomatic therapies include pharmacologic treatment and deep brain
stimulation (DBS).
Step 1: Does chorea need to be treated? When it is mild and nonbothersome, treatment is not
required.
Step 2: Is specific treatment available?
Step 3: If chorea is not controlled by specific treatment or specific treatment is not available,
what symptomatic therapies should be selected?
Step 4: Are there any other symptoms than chorea that require treatment or surveillance?
Patients can benefit from multidisciplinary approach.
Step 5: Does any family member need further evaluation or genetic counseling?
CONTINUUMJOURNAL.COM 1027
Specific Therapies
◆ Blood sugar control in nonketotic hyperglycemia
◆ Phlebotomy and hydroxyurea in polycythemia vera
◆ Treatment for hyperthyroidism
◆ Immunotherapies in autoimmune choreas
Symptomatic Therapies
◆ Pharmacologic therapies
◇ Presynaptic dopamine depletors
→ Tetrabenazine and reserpine
→ New-generation vesicular monoamine transporter 2 inhibitors
– Deutetrabenazine
– Valbenazine
◇ Postsynaptic dopamine receptor blockers (dopamine receptor blocking agents,
neuroleptics or antipsychotics)
→ Typical antipsychotics
– Haloperidol
→ Atypical antipsychotics
– Olanzapine
– Risperidone
– Quetiapine
– Clozapine
◇ Others
→ Antiepileptics
– Valproic acid
– Carbamazepine
Deep Brain Stimulation
◆ Reported in Huntington disease, chorea-acanthocytosis, and some acquired choreas
Multidisciplinary Approach
◆ Psychiatrist; physical medicine and rehabilitation specialist; physical, occupational, and
speech therapists
◆ Geneticist, genetic counselor
◆ Social worker
FIGURE 6-7
Sites of action of the main pharmacologic therapies in chorea. 1, Presynaptic dopamine-
depleting agents are vesicular monoamine transporter 2 (VMAT2) inhibitors. 2, Postsynaptic
dopamine receptor blocking agents (DRBAs) act by blocking mainly D2 receptors.
Figure courtesy of Thananan Thammongkolchai, MD.
CONTINUUMJOURNAL.COM 1029
DEEP BRAIN STIMULATION. DBS in chorea remains a moving target. Most studies
have been done in HD70 and chorea-acanthocytosis.71 These are mostly case
reports or small case series,72 which can pose some biases, as cases with poor
outcome are unlikely to be reported. The most common target is the globus
pallidus internus (GPi), and stimulation at the ventral GPi is known to have an
antidyskinetic effect from extensive DBS experience in Parkinson disease.
Further clarification on DBS efficacy, candidacy, and appropriate targets can be
elucidated from future studies and randomized controlled trials.
Factors other than chorea also should be considered when selecting DBS as a
treatment. First, coexisting cognitive and neuropsychiatric features can even be
more debilitating than chorea and other motor features and are unlikely to be
responsive to DBS. Secondly, chorea in HD and chorea-acanthocytosis can
gradually be replaced by parkinsonism in the later stages of the diseases. Thus, the
CONTINUUMJOURNAL.COM 1031
natural history of the disease can contribute to chorea reduction, and different
stimulation or programming strategies may be required in different stages (eg, more
dorsal electrodes in GPi to control parkinsonism). Experimental genetic therapies,
especially huntingtin-lowering treatment, are under active investigation,73,74
and more data will be required before application to clinical practice.
A holistic approach is also crucial in the care of patients with chorea. Many patients
with chorea experience not only motor symptoms but also cognitive and
neuropsychiatric features as well as psychosocial issues. A multidisciplinary team
approach that includes psychiatrists, physical medicine and rehabilitation specialists,
physical therapists, occupational therapists, speech therapists, social workers, and
geneticists and genetic counselors can be beneficial. Furthermore, in some disorders,
especially HD, the unit of treatment is an entire family rather than an individual patient.
Caregiver burden and the stress of at-risk family members should also be supported.
CONCLUSION
The etiology of chorea can be categorized into acquired and genetic causes.
Among the most useful features in the diagnostic approach are time course, age
group, and known prevalence in the population. Other clinical diagnostic clues
can help guide the investigation. Genetic counseling should be considered before
sending genetic testing in chorea. Management should include not only
symptomatic treatment of chorea but also other associated medical, neurologic,
and psychiatric aspects. Family and caregiver support should be also taken into
consideration when caring for patients with chorea.
VIDEO LEGENDS
VIDEO 6-1 VIDEO 6-4
Hemiballism secondary to central nervous system Sydenham chorea. Video shows an 8-year-old boy
toxoplasmosis. Video shows a 54-year-old man with Sydenham chorea who presented with an
with left hemiballism exhibiting large-amplitude acute temporal profile of severe generalized
movements involving the predominantly proximal chorea. He also displays hypotonia and milkmaid’s
arm and leg. On MRI, fluid-attenuated inversion grip. He is unable to stand unassisted.
recovery (FLAIR) images show numerous multifocal links.lww.com/CONT/A354
hyperintense lesions that also involve the basal
ganglia. Reproduced with permission from Frucht SJ.17
links.lww.com/CONT/A351 © 2013 Springer Science+Business Media.
VIDEO 6-3
Lupus chorea. Video shows a 29-year-old woman
with acute generalized chorea and encephalopathy
as her first presentation of systemic lupus
erythematosus. Both chorea and her mental status
improved after 3 days of treatment with IV steroids
and tetrabenazine 75 mg/d.
links.lww.com/CONT/A353
Courtesy of Steven Frucht, MD.
© 2019 American Academy of Neurology.
REFERENCES
1 Morris JGL, Jankelowitz SK, Fung VSC, et al. 11 Termsarasab P. Hyperkinetic movement disorders:
Athetosis I: historical considerations. Mov Dis videodiagnosis and treatment. Presented at:
2002;17:1278–1280. doi:10.1002/mds.10267. 69th Annual Meeting of the American Academy
of Neurology; April 22–28, 2017; Boston, MA.
2 Postuma RB, Lang AE. Hemiballism: revisiting a
classic disorder. Lancet Neurol 2003;2:661–668. 12 Damato V, Balint B, Kienzler AK, Irani SR. The
doi:10.1016/S1474-4422(03)00554-4. clinical features, underlying immunology, and
treatment of autoantibody-mediated movement
3 Cosentino C, Torres L, Nunez Y, Suarez R,
disorders. Mov Disord 2018;33(9):1376–1389.
Velez M, Flores M. Hemichorea/hemiballism
doi:10.1002/mds.27446.
associated with hyperglycemia: report of
20 cases. Tremor Other Hyperkinet Mov (N Y) 13 Varley JA, Webb AJS, Balint B, et al. The
2016;6:402. doi:10.7916/D8DN454P. movement disorder associated with NMDAR
antibody-encephalitis is complex and characteristic:
4 Lew J, Frucht SJ, Kremyanskaya M, et al.
an expert video-rating study. J Neurol Neurosurg
Hemichorea in a patient with JAK2V617F blood
Psychiatry 2018;pii:jnnp-2018–318584. doi:10.1136/
cells. Blood 2013;121(7):1239–1240. doi:10.1182/
jnnp-2018-318584.
blood-2012-12-468751.
14 Sabater L, Gaig C, Gelpi E, et al. A novel non-
5 Ryan C, Ahlskog JE, Savica R. Hyperglycemic
rapid-eye movement and rapid-eye-movement
chorea/ballism ascertained over 15 years at a
parasomnia with sleep breathing disorder
referral medical center. Parkinsonism Relat
associated with antibodies to IgLON5: a case
Disord 2018;48:97–100. doi:10.1016/j.
series, characterisation of the antigen, and post-
parkreldis.2017.12.032.
mortem study. Lancet Neurol 2014;13(6):575–586.
6 Walker RH. Untangling the thorns: advances doi:10.1016/S1474-4422(14)70051-1.
in the neuroacanthocytosis syndromes.
15 Gaig C, Graus F, Compta Y, et al. Clinical
J Mov Disord 2015;8(2):41–54. doi:10.14802/
manifestations of the anti-IgLON5 disease.
jmd.15009.
Neurology 2017;88(18):1736–1743. doi:10.1212/
7 Fekete R, Jankovic J. Upper facial chorea in WNL.0000000000003887.
Huntington disease. J Clin Mov Disord 2014;1:7.
16 Gelpi E, Höftberger R, Graus F, et al.
doi:10.1186/2054-7072-1-7.
Neuropathological criteria of anti-IgLON5-
8 Hensman Moss DJ, Poulter M, Beck J, et al. related tauopathy. Acta Neuropathol 2016;132(4):
C9orf72 expansions are the most common 531–543. doi:10.1007/s00401-016-1591-8.
genetic cause of Huntington disease
17 Frucht SJ, editor. Movement disorder
phenocopies. Neurology 2014;82(4):292–299.
emergencies. 2nd edition. New York, NY:
doi:10.1212/WNL.0000000000000061.
Humana Press, 2013.
9 Van Mossevelde S, van der Zee J, Gijselinck I,
18 Cardoso F, Vargas AP, Oliveira LD, et al.
et al. Clinical evidence of disease anticipation in
Persistent Sydenham's chorea. Mov Disord 1999;
families segregating a C9orf72 repeat expansion.
14(5):805–807. doi:10.1002/1531-8257(199909)14:
JAMA Neurol 2017;74(4):445–452. doi:10.1001/
5<805::AID-MDS1013>3.0.CO;2-P.
jamaneurol.2016.4847.
19 Robottom BJ, Weiner WJ. Chorea gravidarum.
10 Krause A, Mitchell C, Essop F, et al. Junctophilin 3
Handb Clin Neurol 2011;100:231–235. doi:10.1016/
(JPH3) expansion mutations causing Huntington
B978-0-444-52014-2.00015-X.
disease like 2 (HDL2) are common in South
African patients with African ancestry and a 20 Termsarasab P, Frucht SJ. The “stutter-step”: a
Huntington disease phenotype. Am J Med Genet peculiar gait feature in advanced Huntington's
B Neuropsychiatr Genet 2015;168(7):573–585. disease and chorea-acanthocytosis. Mov Disord
doi:10.1002/ajmg.b.32332. Clin Pract 2018;5:223–224. doi:10.1002/
mdc3.12586.
CONTINUUMJOURNAL.COM 1033
21 Bates GP, Dorsey R, Gusella JF, et al. Huntington 36 Peikert K, Danek A, Hermann A. Current state of
disease. Nat Rev Dis Primers 2015;1:15005. knowledge in chorea-acanthocytosis as core
doi:10.1038/nrdp.2015.5. neuroacanthocytosis syndrome. Eur J Med Genet
2017;61(11):699–705. doi:10.1016/j.ejmg.2017.12.007.
22 McColgan P, Tabrizi SJ. Huntington's disease: a
clinical review. Eur J Neurol 2018;25(1):24–34. 37 Storch A, Kornhass M, Schwarz J. Testing for
doi:10.1111/ene.13413. acanthocytosis: a prospective reader-blinded
study in movement disorder patients. J Neurol
23 Termsarasab P, Thammongkolchai T, Rucker JC,
2005;252(1)84–90. doi:10.1007/s00415-005-0616-3.
Frucht SJ. The diagnostic value of saccades in
movement disorder patients: a practical guide 38 Schneider SA, Lang AE, Moro E, et al.
and review. J Clin Mov Disord 2015;2:14. doi: Characteristic head drops and axial extension in
10.1186/s40734-015-0025-4. advanced chorea-acanthocytosis. Mov Disord
2010;25(10):1487–1491. doi:10.1002/mds.23052.
24 Gusella JF, MacDonald ME, Lee JM. Genetic
modifiers of Huntington's disease. Mov Disord 39 Bhidayasiri R, Jitkritsadakul O, Walker RH. Axial
2014;29(11):1359–1365. doi:10.1002/mds.26001. sensory tricks in chorea-acanthocytosis: insights
into phenomenology. Tremor Other Hyperkinet
25 MacLeod R, Tibben A, Frontali M, et al.
Mov (N Y) 2017;7:475. doi:10.7916/D8PV6RWW.
Recommendations for the predictive genetic
test in Huntington's disease. Clin Genet 2013; 40 Gras D, Jonard L, Roze E, et al. Benign hereditary
83(3):221–231. doi:10.1111/j.1399-0004.2012.01900.x. chorea: phenotype, prognosis, therapeutic
outcome and long term follow-up in a large
26 Nance MA. Genetic counseling and testing for
series with new mutations in the TITF1/NKX2-1
Huntington's disease: a historical review. Am J
gene. J Neurol Neurosurg Psychiatry 2012;83(10):
Med Genet B Neuropsychiatr Genet 2017;174(1):
956–962. doi:10.1136/jnnp-2012-302505.
75–92. doi:10.1002/ajmg.b.32453.
41 Peall KJ, Lumsden D, Kneen R, et al. Benign
27 Reilmann R, Leavitt BR, Ross CA. Diagnostic
hereditary chorea related to NKX2.1: expansion
criteria for Huntington's disease based on natural
of the genotypic and phenotypic spectrum.
history. Mov Disord 2014;29(11):1335–1341. doi:
Dev Med Child Neurol 2014;56(7):642–648.
10.1002/mds.26011.
doi:10.1111/dmcn.12323.
28 Schneider SA, Bird T. Huntington's disease,
42 Peall KJ, Kurian MA. Benign hereditary chorea: an
Huntington's disease look-alikes, and benign
update. Tremor Other Hyperkinet Mov (N Y) 2015;
hereditary chorea: what's new? Mov Disord Clin
5:314. doi:10.7916/D8RJ4HM5.
Pract 2016;3:342–354. doi:10.1002/mdc3.12312.
43 Mencacci NE, Erro R, Wiethoff S, et al. ADCY5
29 Burke JR, Wingfield MS, Lewis KE, et al. The
mutations are another cause of benign
Haw river syndrome: dentatorubropallidoluysian
hereditary chorea. Neurology 2015;85(1):80–88.
atrophy (DRPLA) in an African–American family.
doi:10.1212/WNL.0000000000001720.
Nat Genet 1994;7(4):521–524. doi:10.1038/
ng0894-521. 44 Chen DH, Méneret A, Friedman JR, et al. ADCY5-
related dyskinesia: broader spectrum and genotype-
30 Pearson TS. More than ataxia: hyperkinetic
phenotype correlations. Neurology 2015;85(23):
movement disorders in childhood autosomal
2026–2035. doi:10.1212/WNL.0000000000002058.
recessive ataxia syndromes. Tremor Other
Hyperkinet Mov (N Y) 2016;6:368. doi:10.7916/ 45 Mencacci NE, Kamsteeg EJ, Nakashima K, et al.
D8H70FSS. De novo mutations in PDE10A cause
childhood-onset chorea with bilateral striatal
31 Margolis RL, Holmes SE. Huntington’s
lesions. Am J Hum Genet 2016;98(4):763–771.
disease–like 2: a clinical, pathological, and
doi:10.1016/j.ajhg.2016.02.015.
molecular comparison to Huntington’s disease.
Clin Neurosci Res 2003;3(3):187–196. 46 Diggle CP, Sukoff Rizzo SJ, Popiolek M, et al.
doi.org/10.1016/S1566-2772(03)00061-6. Biallelic mutations in PDE10A lead to loss of
striatal PDE10A and a hyperkinetic movement
32 Fujita K, Osaki Y, Harada M, et al. Brain and
disorder with onset in infancy. Am J Hum Genet
liver iron accumulation in aceruloplasminemia.
2016;98(4):735–743. doi:10.1016/j.ajhg.2016.03.015.
Neurology 2013;81(24):2145–2146. doi: 10.1212/01.
wnl.0000437304.30227.bd. 47 Saitsu H, Fukai R, Ben-Zeev B, et al. Phenotypic
spectrum of GNAO1 variants: epileptic
33 Ohta E, Takiyama Y. MRI findings in
encephalopathy to involuntary movements with
neuroferritinopathy. Neurol Res Int 2012;2012:
severe developmental delay. Eur J Hum Genet
197438. doi: 10.1155/2012/197438.
2016;24(1):129–134. doi:10.1038/ejhg.2015.92.
34 Batla A, Adams ME, Erro R, et al. Cortical pencil
48 Kurian MA, Jungbluth H. Genetic disorders of
lining in neuroferritinopathy: a diagnostic cluc.
thyroid metabolism and brain development.
Neurology 2015;84(17):1816–1818. doi: 10.1212/
Dev Med Child Neurol 2014;56(7):627–634.
WNL.0000000000001511.
doi:10.1111/dmcn.12445.
35 Jung HH, Danek A, Walker RH.
49 Fernandez M, Raskind W, Wolff J, et al. Familial
Neuroacanthocytosis syndromes. Orphanet J
dyskinesia and facial myokymia (FDFM): a novel
Rare Dis 2011;6:68. doi:10.1186/1750-1172-6-68.
movement disorder. Ann Neurol 2001;49(4):
486–492. doi:10.1002/ana.98.
CONTINUUMJOURNAL.COM 1035
Ataxia
C O N T I N UU M AUDIO By Sheng-Han Kuo, MD
INTERVIEW AVAILABLE
ONLINE
VIDEO CONTENT ABSTRACT
A VA I L A B L E O N L I N E PURPOSE OF REVIEW: This
article reviews the symptoms, laboratory and
neuroimaging diagnostic tests, genetics, and management of cerebellar
ataxia.
CITE AS:
CONTINUUM (MINNEAP MINN) develop cerebellar ataxia. In addition, several ongoing clinical trials for
2019;25(4, MOVEMENT DISORDERS): Friedreich ataxia and spinocerebellar ataxia will likely result in novel
1036–1054.
symptomatic and disease-modifying therapies for ataxia. Antisense
Address correspondence to oligonucleotides for spinocerebellar ataxias associated with CAG repeat
Dr Sheng-Han Kuo, 650 West
168th St, Room 305, New York,
expansions might be a promising therapeutic strategy.
NY 10032, [email protected].
SUMMARY: Cerebellar ataxias include heterogeneous disorders affecting
RELATIONSHIP DISCLOSURE:
Dr Kuo has received personal
cerebellar function, leading to ataxic symptoms. Step-by-step diagnostic
compensation for serving on the workups with genetic investigations are likely to reveal the underlying
medical advisory board of the causes of ataxia. Some disease-specific therapies for ataxia exist, such as
International Essential Tremor
Foundation and on the research vitamin E for ataxia with vitamin E deficiency and thiamine for Wernicke
advisory board of the National encephalopathy, highlighting the importance of recognizing these forms of
Ataxia Foundation. Dr Kuo has
ataxia. Finally, genetic diagnosis for patients with ataxia will accelerate
received research/grant support
from the American Brain clinical trials for disease-modifying therapy and will have prognostic value
Research Training Fellowship, and implications for family planning for these patients.
American Parkinson Disease
Association, International
Essential Tremor Foundation, the
Louis V. Gerstner Jr Scholarship, INTRODUCTION
T
Continued on page 1054
he causes of cerebellar ataxia are diverse, ranging from infectious and
immune mediated to degenerative. In addition, many genetic causes
UNLABELED USE OF
PRODUCTS/INVESTIGATIONAL of cerebellar ataxia exist. Therefore, the workup for cerebellar ataxia
USE DISCLOSURE: usually poses significant challenges to neurologists. In addition, the
Dr Kuo discusses the unlabeled/
treatment for cerebellar ataxia has been traditionally thought to be
investigational use of
amantadine, baclofen, ineffective, leaving many patients with ataxia untreated. Moreover, nonmotor
chlorzoxazone, and riluzole for symptoms, such as depression and mood disorders, are common in patients with
the treatment of cerebellar ataxia;
4-aminopyridine for the treatment
ataxia and are often underrecognized and undertreated.
of episodic ataxia type 2; deep To circumvent these challenges of the diagnosis and treatment for patients
brain stimulation for the with ataxia, this review provides step-by-step approaches and an overview for
treatment of spinocerebellar
ataxia type 2; miglustat for the ataxia. This review does not cover ataxia caused by strokes, tumors, or multiple
treatment of Niemann-Pick sclerosis since these lesion-related ataxias are easily identified by neuroimaging
disease type C; and varenicline and are discussed in other issues within the Continuum curriculum. The section
for spinocerebellar ataxia type 3.
on ataxia genetics only briefly covers the most common causes of ataxia and
© 2019 American Academy
does not provide a complete list of ataxic genes, which can be found in other
of Neurology. comprehensive reviews.1–3
CONTINUUMJOURNAL.COM 1037
therefore, these signs are helpful in differentiating cerebellar ataxia from sensory
ataxia, especially in the early stages of the disease when no cerebellar atrophy is
found on neuroimaging.
Patients with ataxia usually have scanning speech (ie, words are broken up
into separate syllables with disrupted normal speech patterns). The speed of
speech could become slow, and the volume of speech could be variable.
The three commonly used tasks for ataxic hand examination include (1)
finger-nose-finger test (the patient points repeatedly with his or her index finger
from his or her nose to the examiner’s finger), (2) finger chase (the patient’s
index finger follows the examiner’s moving index finger as precisely as possible),
and (3) fast alternating movements (the patient performs cycles of repetitive
TABLE 7-1 Acute, Subacute, Chronic, and Episodic Causes of Cerebellar Ataxia
CONTINUUMJOURNAL.COM 1039
CONTINUUMJOURNAL.COM 1041
TABLE 7-2 Brain MRI Findings Associated With Specific Forms of Ataxia
Cervical spinal cord atrophy Cervical spinal cord atrophy Friedreich ataxia
T2 hyperintensity in the bilateral inferior olivary Hypertrophic degeneration POLG ataxia, adult-onset Alexander
nuclei of the neurons in the inferior disease, ataxia with gluten sensitivity
olivary nuclei
Dentate nucleus calcification (CT)a Dentate nucleus calcification Spinocerebellar ataxia type 20
T2 gradient recalled echo showing linear Iron deposition on the Superficial siderosis
hypointensity around the cerebellum and surface of the brain
brainstem parenchyma
GENETICS OF ATAXIA
Genetic causes of ataxia are many. The Online Mendelian Inheritance in Man
(OMIM) can serve as a useful resource for up-to-date ataxia genetics.13 Following
is an overview of ataxia genetics based on the pattern of inheritance.
CONTINUUMJOURNAL.COM 1043
FIGURE 7-2
The diagnostic workflow for cerebellar ataxia.
AD = autosomal dominant; AR = autosomal recessive; FXTAS = fragile X tremor-ataxia syndrome; GAD =
glutamic acid decarboxylase; ILOCA = idiopathic late-onset cerebellar ataxia; MRI = magnetic resonance
imaging; MSA = multiple system atrophy; REM = rapid eye movement; SCA = spinocerebellar ataxia.
Common Autosomal Dominant Ataxias Associated With Repeat Expansions TABLE 7-3
CONTINUUMJOURNAL.COM 1045
are also common in Friedreich ataxia. Friedreich ataxia is caused by intronic GAA
repeat expansions of the FXN gene, which result in inefficient production of
frataxin protein and lead to disturbed mitochondrial function. About 5% of
patients with Friedreich ataxia have one allele carrying a point mutation in the
FXN gene and another allele with repeat expansions. Therefore, a neurologist
should still have a high suspicion of Friedreich ataxia if the expanded repeats are
detected in only one allele in patients with otherwise classical Friedreich ataxia.
COMMENT This case demonstrates that determination for repeat expansions is the
first step for autosomal dominant ataxia. Slow saccades are the key feature
for SCA2. Other than treating ataxia symptoms, parkinsonism in patients
with ataxia can sometimes respond to carbidopa/levodopa. Finally,
depressive symptoms are very common in patients with cerebellar ataxia,9
possibly due to the cerebellar cognitive affective syndrome,10 and should
be treated with antidepressants.
CONTINUUMJOURNAL.COM 1047
with upper or lower motor neuron signs, pes cavus, and scoliosis.26 Patients
with Niemann-Pick disease type C have characteristic vertical supranuclear
ophthalmoplegia (VIDEO 7-4, links.lww.com/CONT/A361)27 in addition to mental
retardation, dystonia, and cognitive decline.
X-linked Ataxia
The most common X-linked ataxia is fragile X tremor-ataxia syndrome, which is
caused by CGG repeat expansions within the FMR1 gene, leading to RNA toxic
function. A longer repeat expansion of the FMR1 gene, as the result of
anticipation, will lead to premature ovarian failure in the patients’ daughters and
mental retardation in patients’ grandsons. Thus, fragile X tremor-ataxia
syndrome should be considered in the context of a family history of mental
retardation and premature ovarian failure. Parkinsonism and autonomic
dysfunction can also occur; therefore, fragile X tremor-ataxia syndrome should
be in the differential diagnosis for multiple system atrophy. T2 hyperintensity in
the middle cerebellar peduncles and corpus callosum on brain MRI are
characteristics of fragile X tremor-ataxia syndrome and can help with the
diagnosis (FIGURE 7-3).28
Friedreich ataxia Repeat expansions in From childhood Pes cavus, scoliosis, square-wave jerks,
intron 1 of FXN gene to third decade hyporeflexia, spasticity (adult onset)
Sensory ataxic neuropathy, POLG mutations 20–60 years Ophthalmoplegia, dysarthria, ptosis,
dysarthria, and myoclonus, epilepsy
ophthalmoparesis (SANDO)
Ataxia with oculomotor apraxia APTX mutations <20 years Oculomotor apraxia, chorea, dystonia
type 1
Ataxia with oculomotor apraxia SETX mutations From childhood Oculomotor apraxia, chorea, dystonia,
type 2 to third decade elevated α-fetoprotein
Autosomal recessive cerebellar SYNE1 mutations From second to Spasticity, lower motor neuron signs
ataxia type 1 fifth decade
Autosomal recessive cerebellar ADCK3 mutations <10 years Mental retardation, myoclonus, epilepsy,
ataxia type 2 exercise intolerance
Niemann-Pick disease type C NPC1 and NPC2 From first to Vertical supranuclear ophthalmoplegia,
mutations third decade splenomegaly, dystonia, cognitive decline
CONTINUUMJOURNAL.COM 1049
clinicians should still keep these genes in mind. One example is the SPG7 mutation,
which has been observed in patients with idiopathic cerebellar ataxia.30
DEGENERATIVE ATAXIA
Degenerative forms of ataxia usually occur in patients who are older than
60 years of age and have no family history of ataxia. In this category, multiple
system atrophy and idiopathic late-onset cerebellar ataxia are the two most
common diseases.
Multiple system atrophy is characterized by cerebellar ataxia, parkinsonism,
autonomic instability (orthostatic hypotension; impotence; and urinary
frequency, urgency, and incontinence), and pyramidal signs.31 Depending on
the predominant features, multiple system atrophy can also be divided into
multiple system atrophy–parkinsonism type or multiple system atrophy–cerebellar
type. Multiple system atrophy–cerebellar type constitutes approximately 30% of
all multiple system atrophy cases.31 Patients with multiple system atrophy with
predominant cerebellar ataxia generally have a slower disease progression than
those with predominant parkinsonism. In the early stage of the disease, patients
might present with relatively isolated cerebellar ataxia. Therefore, the history
and examination should focus on searching for any signs of parkinsonian
features, autonomic dysfunction, and upper motor neuron signs. Rapid eye
movement sleep behavior disorder is often present, which can be very helpful
in indicating an underlying synucleinopathy associated with multiple system
atrophy.31 Respiratory stridor and obstructive sleep apnea sometimes are
present. Therefore, for patients with cerebellar ataxia onset later in life,
autonomic tests and sleep studies can provide additional evidence to support the
diagnosis of multiple system atrophy when clinical symptoms are not yet
evident. Brain MRI in patients with multiple system atrophy may show the hot
cross bun sign; this sign might not be evident in the early stage of the disease but
can show up later after the disease progresses.
Multiple system atrophy is a disease with a relatively fast progression. However,
idiopathic late-onset cerebellar ataxia is a disease with a slow progression,
and it usually does not compromise the life span. Patients with idiopathic
late-onset cerebellar ataxia usually will not develop other neurologic features
such as hyperreflexia or parkinsonism.
Practically, the presence of parkinsonism and autonomic dysfunction in
elderly patients with ataxia usually indicates a poor prognosis because the
diagnosis is likely to be multiple system atrophy. If no parkinsonism or
autonomic dysfunction is seen within 5 years after the onset of ataxia, these
patients are likely to follow a benign clinical course with the diagnosis of
idiopathic late-onset cerebellar ataxia.
TREATMENT OF ATAXIA
The treatment for ataxia can be divided into symptomatic and disease-modifying
therapies. Disease-modifying therapy exists for some causes of ataxia, especially
for acquired cases (eg, thiamine for Wernicke encephalopathy,32 gluten-free diet
for ataxia associated with gluten sensitivity, and IV immunoglobulin and plasma
exchange for anti-GAD ataxia and paraneoplastic ataxia). Disease-modifying
therapies for genetic ataxias include vitamin E replacement for ataxia associated
with vitamin E deficiency and abetalipoproteinemia, miglustat for Niemann-Pick
disease type C, and ketogenic diet for glucose transporter type 1 deficiency.33
CONTINUUMJOURNAL.COM 1051
levels and to improve the symptoms of mouse models of SCA2 and SCA3.47,48
These studies formulate a strong scientific rationale to test antisense
oligonucleotides in patients with ataxia in the near future.
CONCLUSION
The causes of cerebellar ataxia are many and can present as diagnostic challenges.
This article provides a step-by-step diagnostic guide for patients with ataxia. The
acquired causes of ataxia should first be considered, followed by genetic and
degenerative forms of ataxia. If genetic ataxia is suspected, pathologic repeat
expansions in the ataxic genes should be determined first, followed by assessing
for sequence alterations. Among late-onset cerebellar ataxias, multiple system
atrophy is most common and should be considered. Although the treatment for
cerebellar ataxia remains only modestly effective, several novel therapies are
currently being tested, including pharmacologic therapy, brain stimulation, and
antisense oligonucleotides.
USEFUL WEBSITES
ONLINE MENDELIAN INHERITANCE IN MAN (OMIM) FRIEDREICH’S ATAXIA RESEARCH ALLIANCE
The OMIM website provides useful guidance for This website discusses therapy development for
genetic mutations associated with cerebellar ataxia. Friedreich ataxia.
omim.org curefa.org/pipeline
VIDEO LEGENDS
VIDEO 7-1 VIDEO 7-3
Different degrees of gait ataxia. The first video Spinocerebellar ataxia type 2. The first video
segment shows an 18-year-old man with ataxia with segment shows the 55-year-old woman discussed
oculomotor apraxia type 2. Mild gait ataxia can in CASE 7-2 with spinocerebellar ataxia type 2. She
manifest with variable step length, side steps, and has limited upward gaze and slow saccadic eye
veering toward one side without a marked wide movements without nystagmus or hypermetric or
base. The second video segment shows a 37-year- hypometric saccades. She has dysmetria on finger-
old man with idiopathic cerebellar ataxia. In patients nose-finger tests and an overshoot in finger chase
with moderate ataxia, the gait becomes wide tests. She also has impaired rapid alternating
based with obvious variable stride lengths. The third movements. Her facial expression is hypomimic,
video segment shows a 32-year-old woman with and her gait is wide based and shows variable stride
spinocerebellar ataxia type 2. In the advanced length. The second video segment shows the
stages of ataxia, a wide-based gait is more evident, same patient 5 years later. Her symptoms have
and the stride length becomes shorter to progressed. She has ophthalmoparesis, particularly
compensate for ataxia. in vertical gaze, and her saccadic eye movements
links.lww.com/CONT/A358 have become very slow.
© 2019 American Academy of Neurology. links.lww.com/CONT/A360
© 2019 American Academy of Neurology.
VIDEO 7-2
Multiple system atrophy, cerebellar type. VIDEO 7-4
Video shows the 56-year-old woman discussed in Niemann-Pick disease type C. Video shows a
CASE 7-1 who developed imbalance and walked “as 30-year-old man with Niemann-Pick disease type C.
if she were drunk.” She veered toward one side He has relatively preserved smooth pursuit in the
while walking, had two episodes of syncope, horizontal direction without end-gaze nystagmus.
and her speech and hand dexterity were He can perform upward pursuit, but his downward
subsequently involved. On examination, she has pursuit movements are extremely slow. He also has
hypomimic facial expression, hypermetric relatively preserved horizontal saccades and has
saccades, dysmetria on finger-nose-finger tests, great difficulty in vertical saccades, particularly in
bradykinesia in finger taps, and ataxic gait with the downward direction.
variable footsteps. Her postmortem brain links.lww.com/CONT/A361
pathology demonstrated glial cytoplasmic © 2019 American Academy of Neurology.
inclusions along with olivopontocerebellar
atrophy, confirming the diagnosis of multiple
system atrophy, cerebellar type.
links.lww.com/CONT/A359
© 2019 American Academy of Neurology.
1 Ashizawa T, Öz G, Paulson HL. Spinocerebellar 15 Fogel BL, Lee H, Deignan JL, et al. Exome
ataxias: prospects and challenges for therapy sequencing in the clinical diagnosis of sporadic
development. Nat Rev Neurol 2018;14(10): or familial cerebellar ataxia. JAMA Neurol
590–605. doi:10.1038/s41582-018-0051-6. 2014;71(10):1237–1246. doi:10.1001/jamaneurol.
2014.1944.
2 Schöls L, Bauer P, Schmidt T, et al. Autosomal
dominant cerebellar ataxias: clinical features, 16 Ashizawa T, Figueroa KP, Perlman SL, et al.
genetics, and pathogenesis. Lancet Neurol 2004; Clinical characteristics of patients with
3:291–304. doi:10.1016/S1474-4422(04)00737-9. spinocerebellar ataxias 1, 2, 3 and 6 in the US; a
prospective observational study. Orphanet J
3 Anheim M, Tranchant C, Koenig M. The autosomal
Rare Dis 2013;8:177. doi:10.1186/1750-1172-8-177.
recessive cerebellar ataxias. N Engl J Med 2012;
366(7):636–646. doi:10.1056/NEJMra1006610. 17 Jacobi H, Bauer P, Giunti P, et al. The natural
history of spinocerebellar ataxia type 1, 2, 3, and
4 Luo L, Wang J, Lo RY, et al. The initial symptom
6: a 2-year follow-up study. Neurology 2011;77(11):
and motor progression in spinocerebellar
1035–1041. doi:10.1212/WNL.0b013e31822e7ca0.
ataxias. Cerebellum 2017;16(3):615–622.
doi:10.1007/s12311-016-0836-3. 18 Romano S, Coarelli G, Marcotulli C, et al. Riluzole
in patients with hereditary cerebellar ataxia: a
5 Moscovich M, Okun MS, Favilla C, et al. Clinical
randomised, double-blind, placebo-controlled
evaluation of eye movements in spinocerebellar
trial. Lancet Neurol 2015;14(10):985–991.
ataxias: a prospective multicenter study.
doi:10.1016/S1474-4422(15)00201-X.
J Neuroophthalmol 2015;35(1):16–21. doi:10.1097/
WNO.0000000000000167. 19 Velázquez-Pérez L, Rodríguez-Chanfrau J,
Garcia-Rodríguez JC, et al. Oral zinc sulphate
6 Kuo PH, Lo RY, Tanji K, Kuo SH. Clinical reasoning:
supplementation for six months in SCA2
a 58-year-old man with progressive ptosis and
patients: a randomized, double-blind, placebo-
walking difficulty. Neurology 2017;89(1):e1–e5.
controlled trial. Neurochem Res 2011;36(10):
doi:10.1212/WNL.0000000000004064.
1793–1800. doi:10.1007/s11064-011-0496-0.
7 Lin CY, Wang MJ, Tse W, et al. Serum antigliadin
20 Zesiewicz TA, Greenstein PE, Sullivan KL, et al. A
antibodies in cerebellar ataxias: a systematic
randomized trial of varenicline (Chantix) for the
review and meta-analysis. J Neurol Neurosurg
treatment of spinocerebellar ataxia type 3.
Psychiatry 2018;89(11):1174–1180. doi:10.1136/
Neurology 2012;78(8):545–550. doi:10.1212/
jnnp-2018-318215.
WNL.0b013e318247cc7a.
8 Tee BL, Longoria Ibarrola EM, Geschwind MD.
21 Lo RY, Figueroa KP, Pulst SM, et al. Coenzyme Q10
Prion diseases. Neurol Clin 2018;36(4):865–897.
and spinocerebellar ataxias. Mov Disord 2015;
doi:10.1016/j.ncl.2018.07.005.
30(2):214–220. doi:10.1002/mds.26088.
9 Lo RY, Figueroa KP, Pulst SM, et al. Depression
22 Zesiewicz TA, Wilmot G, Kuo SH, et al. Comprehensive
and clinical progression in spinocerebellar
systematic review summary: treatment of cerebellar
ataxias. Parkinsonism Relat Disord 2016;22:87–92.
motor dysfunction and ataxia: report of the
doi:10.1016/j.parkreldis.2015.11.021.
Guideline Development, Dissemination, and
10 Hoche F, Guell X, Vangel MG, et al. The cerebellar Implementation Subcommittee of the American
cognitive affective/Schmahmann syndrome Academy of Neurology. Neurology 2018;90(10):
scale. Brain 2018;141(1):248–270. doi:10.1093/ 464–471. doi:10.1212/WNL.0000000000005055.
brain/awx317.
23 Ashraf NS, Duarte-Silva S, Shaw ED, et al.
11 McFarland NR. Diagnostic approach to atypical Citalopram reduces aggregation of ATXN3 in a
parkinsonian syndromes. Continuum (Minneap YAC transgenic mouse model of Machado-Joseph
Minn) 2016;22(4, Movement Disorders):1117–1142. disease. Mol Neurobiol 2019;56(5):3690–3701.
doi:10.1212/CON.0000000000000347. doi:10.1007/s12035-018-1331-2.
12 Kuo PH, Kuo SH, Lo RY. Deferiprone reduces 24 Seidel K, Siswanto S, Brunt ER, et al. Brain pathology
hemosiderin deposition in superficial siderosis. of spinocerebellar ataxias. Acta Neuropathol
Can J Neurol Sci 2017;44(2):219–220. doi:10.1017/ 2012;124(1):1–21. doi:10.1007/s00401-012-1000-x.
cjn.2016.329.
25 Paulson HL, Shakkottai VG, Clark HB, Orr HT.
13 Online Mendelian Inheritance in Man: an online Polyglutamine spinocerebellar ataxias—from
catolog of human genes and genetic disorders. genes to potential treatments. Nat Rev Neurosci
omim.org. Updated April 23, 2019. Accessed 2017;18(10):613–626. doi:10.1038/nrn.2017.92.
June 4, 2019.
26 Mademan I, Harmuth F, Giordano I, et al.
14 Durr A. Autosomal dominant cerebellar ataxias: Multisystemic SYNE1 ataxia: confirming the high
polyglutamine expansions and beyond. Lancet frequency and extending the mutational and
Neurol 2010;9(9):885–894. doi:10.1016/S1474- phenotypic spectrum. Brain 2016;139(pt 8):e46.
4422(10)70183-6. doi:10.1093/brain/aww115.
CONTINUUMJOURNAL.COM 1053
27 Gupta DK, Blanco-Palmero VA, Chung WK, Kuo 38 Botez MI, Botez-Marquard T, Elie R, et al.
SH. Abnormal vertical eye movements as a clue Amantadine hydrochloride treatment in
for diagnosis of Niemann-Pick type C. Tremor heredodegenerative ataxias: a double blind
Other Hyperkinet Mov (N Y) 2018;8:560. study. J Neurol Neurosurg Psychiatry 1996;61(3):
doi:10.7916/D8XS7BGD. 259–264. doi:10.1136/jnnp.61.3.259.
28 Hagerman PJ, Hagerman RJ. Fragile X-associated 39 Bushart DD, Chopra R, Singh V, et al. Targeting
tremor/ataxia syndrome–and older face of the potassium channels to treat cerebellar ataxia.
fragile X gene. Nat Clin Pract Neurol 2007;3(2): Ann Clin Transl Neurol 2018;5(3):297–314.
107–112. doi:10.1038/ncpneuro0373. doi:10.1002/acn3.527.
29 Jen JC, Wan J. Episodic ataxias. Handb Clin 40 Fryer JD, Yu P, Kang H, et al. Exercise and genetic
Neurol 2018;155:205–215. doi:10.1016/B978-0-444- rescue of SCA1 via the transcriptional repressor
64189-2.00013-5. Capicua. Science 2011;334(6056):690–693.
doi:10.1126/science.1212673.
30 Pfeffer G, Pyle A, Griffin H, et al. SPG7 mutations
are a common cause of undiagnosed ataxia. 41 Ilg W, Brötz D, Burkard S, et al. Long-term effects
Neurology 2015;84(11):1174–1176. doi:10.1212/WNL. of coordinative training in degenerative cerebellar
0000000000001369. disease. Mov Disord 2010;25(13):2239–2246.
doi:10.1002/mds.23222.
31 Fanciulli A, Wenning GK. Multiple-system
atrophy. N Engl J Med 2015;372(3):249–263. 42 Multiple-System Atrophy Research
doi:10.1056/NEJMra1311488. Collaboration. Mutations in COQ2 in familial and
sporadic multiple-system atrophy. N Engl J Med
32 Kuo SH, Debnam JM, Fuller GN, de Groot J.
2013;369(3):233–244. doi:10.1056/NEJMoa1212115.
Wernicke's encephalopathy: an underrecognized
and reversible cause of confusional state in 43 Barca E, Kleiner G, Tang G, et al. Decreased
cancer patients. Oncology 2009;76(1):10–18. coenzyme Q10 levels in multiple system atrophy
doi:10.1159/000174951. cerebellum. J Neuropathol Exp Neurol 2016;75(7):
663–672. doi:10.1093/jnen/nlw037.
33 Braga Neto P, Pedroso JL, Kuo SH, et al. Current
concepts in the treatment of hereditary ataxias. 44 UMIN Clinical Trials Registry. Clinical trial
Arq Neuropsiquiatr 2016;74(3):244–252. UMIN000031771. upload.umin.ac.jp/cgi-open-
doi:10.1590/0004-282X20160038. bin/ctr_e/ctr_view.cgi?recptno=R000036134.
Updated July 27, 2018; Accessed June 4, 2019.
34 Aranca TV, Jones TM, Shaw JD, et al. Emerging
therapies in Friedreich's ataxia. Neurodegener 45 ClinicalTrials.gov. NCT02960893. clinicaltrials.
Dis Manag 2016;6(1):49–65. doi:10.2217/nmt.15.73. gov/ct2/show/NCT02960893. Updated
August 8, 2018. Accessed June 4, 2019.
35 Friedreich’s Ataxia Research Alliance. Research
pipeline. curefa.org/pipeline. Accessed June 4, 46 Hashimoto T, Muralidharan A, Yoshida K, et al.
2019. Neuronal activity and outcomes from thalamic
surgery for spinocerebellar ataxia. Ann Clin Transl
36 Strupp M, Kalla R, Claassen J, et al. A randomized
Neurol 2018;5(1):52–63. doi:10.1002/acn3.508.
trial of 4-aminopyridine in EA2 and related
familial episodic ataxias. Neurology 2011;77(3): 47 Scoles DR, Meera P, Schneider MD, et al.
269–275. doi:10.1212/WNL.0b013e318225ab07. Antisense oligonucleotide therapy for
spinocerebellar ataxia type 2. Nature 2017;
37 Tsunemi T, Ishikawa K, Tsukui K, et al. The effect
544(7650):362–366. doi:10.1038/nature22044.
of 3,4-diaminopyridine on the patients with
hereditary pure cerebellar ataxia. J Neurol Sci 48 McLoughlin HS, Moore LR, Chopra R, et al.
2010;292(1–2):81–84. doi:10.1016/j.jns.2010.01.021. Oligonucleotide therapy mitigates disease in
spinocerebellar ataxia type 3 mice. Ann Neurol
2018;84(1):64–77. doi:10.1002/ana.25264.
DISCLOSURE
Continued from page 1036
National Ataxia Foundation, for the National Institute
of Environmental Health Sciences pilot grant
ES009089, as principal investigator for studies from
the National Institute of Neurological Disorders and
Stroke (R01 NS104423, K08 NS083738), Parkinson’s
Foundation, and the Rare Disease Clinical Research
Network (RC1NS068897).
By John N. Caviness, MD, FAAN C O N T I N U UM A U D I O
I NT E R V I E W A V A I L AB L E
ONLINE
ABSTRACT
PURPOSE OF REVIEW: This
article offers clinicians a strategic approach for
making sense of a symptom complex that contains myoclonus. The article
presents an evaluation strategy that highly leverages the two major
classification schemes of myoclonus. The goal of this article is to link
evaluation strategy with diagnosis and treatment of myoclonus.
M
yoclonus is defined as a sudden, brief, lightninglike muscle PRODUCTS/INVESTIGATIONAL
USE DISCLOSURE:
contraction.1 Myoclonus may occur from an increase in Dr Caviness discusses the
contraction activity (positive myoclonus) or inhibition unlabeled/investigational use of
of contraction activity (negative myoclonus).1 To recognize anticholinergic medications,
botulinum toxin,
the visual appearance of a myoclonic jerk, the clinician should carbamazepine, clonazepam,
consider the quickest movement that can be performed with that body part. deep brain stimulation,
Indeed, the brief movement of myoclonus does not slow down, pause, or “hang levetiracetam, sodium oxybate,
and tetrabenazine for the
up.” Dystonic spasms or dyskinetic jerks may be sudden, but they are not treatment of myoclonus.
lightning brief. A tremor discharge may be brief but does not result in a jerk or
sudden movement. Indeed, the myoclonus phenotype is both a jerk (sudden) © 2019 American Academy
and the most transient (brief ) compared with any other movement disorder of Neurology.
CONTINUUMJOURNAL.COM 1055
phenotype. Although this definition has been quoted for decades, a study
found only moderate agreement on labeling a movement as myoclonus when
presented with a mixture of myoclonus, tics, and psychogenic jerks.2
The incidence of myoclonus in the clinic setting is estimated at 1.3 cases per
100,000 person-years, with a prevalence of 8.6 cases per 100,000 in the overall
population.3 These numbers are probably an underestimate because myoclonus
is most often secondary to another disorder. However, no single etiology or even
a small group of etiologies accounts for most of the cases. Indeed, myoclonus
has a bewildering number of types and etiologies.
To efficiently evaluate a patient with an unknown etiology for myoclonus,
clinicians must have an organized approach to myoclonus evaluation.
Fortunately, an efficient and pragmatic strategy exists to best define the
diagnosis, physiology, and treatment approach for a specific myoclonus case.
Central to this strategy are myoclonus classification schemes. Myoclonus has
popular classification schemes based on clinical presentation and etiology and a
separate scheme based on physiology.1,4
This article first covers the overall evaluation strategy for myoclonus. Second,
the physiologic classification of myoclonus is discussed. Next, clinical
presentation and etiologic classification and myoclonus types are reviewed.
Finally, symptomatic treatment strategies are discussed. This article focuses
on the connection between the evaluation (including classification) and
symptomatic treatment of myoclonus.
This testing evaluates for basic electrolyte and metabolic disorders known to
cause myoclonus. Although a metabolic etiology may present in the outpatient
clinic, metabolic and drug causes are particularly relevant in the hospital setting.
Moreover, myoclonus may appear after initial hospitalization because new
medications and progression of medical problems commonly occur during the
hospital course.
EEG should be performed in all cases of myoclonus unless the myoclonus
etiology is obvious since myoclonus most commonly arises from either cortical
or cortical-subcortical physiology, including seizures. The EEG also constitutes
the first step in the electrophysiologic evaluation of the myoclonus. For more
electrophysiologic definition, adding surface EMG channels to the EEG or
performing multichannel recording by using an EMG machine is recommended.
As discussed in the section on the physiologic classification of myoclonus, the
EMG discharge duration and pattern of myoclonus provide more exact
physiologic classification of the myoclonus.
Consideration of brain imaging is recommended, especially for symptomatic
presentations. If the distribution of myoclonus could be consistent with a
segmental or peripheral physiology or with propriospinal (nonsegmental)
myoclonus, then spinal cord or peripheral imaging, or both, should be considered.
The combination of these two steps will lead to the etiology of the myoclonus
in most cases. Moreover, if non–myoclonus signs and symptoms are present,
then appropriate evaluation for a unifying diagnosis should be undertaken. Only
after that evaluation should more comprehensive testing for the suspected
etiology of the myoclonus be undertaken.
CONTINUUMJOURNAL.COM 1057
results of steps one and two. Because myoclonus is caused by so many potential
etiologies, considering the best diagnosis when taking into account the
nonmyoclonus clinical features and all test evidence may be useful. To define the
physiology of the myoclonus, more electrophysiologic testing may be needed.
The list of eligible tests for more comprehensive testing is vast, which
reinforces the need for a targeted strategy and, ideally, a stepwise evaluation if
numerous tests are being considered. The broad categories of additional testing
may include specific genetic tests, whole exome sequencing, neuronal specific
antibodies (including paraneoplastic), additional specific toxins, and CSF
examination, among others.
Cortical Myoclonus
The classification of cortical myoclonus implies that intrinsic cortical
hyperexcitability is the major driver of the genesis of the patient’s myoclonus.
The abnormally excessive motor cortical activity occurs at any one instant in a
relatively small part of the motor homunculus. The excitation occurs in one part
of the homunculus, then in another, usually correlating with a multifocal
distribution of the myoclonus in the limbs. However, excitability spread is
common, subsequently activating adjacent muscles almost synchronously or
even bilateral muscles bisynchronously when the discharge transmits through
the corpus collosum. Myoclonus EMG discharges are brief (25 ms to 100 ms) in
duration and usually spread to antagonist and other contiguous muscle groups
(FIGURE 8-1A).
Chronic posthypoxic myoclonus (Lance-Adams syndrome) is a well-known
type of cortical myoclonus. The myoclonus in disorders classified as progressive
myoclonus epilepsy such as Unverricht-Lundborg disease and mitochondrial
conditions are classically cortical. Neurodegenerative illnesses affecting the
Subcortical/Nonsegmental Myoclonus
Two major patterns are seen in subcortical/nonsegmental myoclonus. In the first
pattern, the initial myoclonus EMG discharge corresponds to the subcortical
nidus level (at the brainstem or spinal cord) followed by simultaneous rostral and
caudal recruitment spread of muscle involvement. One example is brainstem
reticular myoclonus in which the first discharge arises in the cranial nerve XI
brainstem innervated muscles (trapezius and sternocleidomastoid). As the
CONTINUUMJOURNAL.COM 1059
Segmental Myoclonus
In segmental myoclonus the myoclonus EMG discharges usually last longer
than 100 ms and are highly rhythmic. By definition, the discharges are limited
to a few contiguous segments of the brainstem or spinal cord.11 The discharges
are either simultaneous with the other segments or at least time locked. The
discharges are fairly persistent and not usually affected by stimuli or exogenous
factors. Palatal segmental myoclonus is the most common location of segmental
myoclonus. Palatal segmental myoclonus is divided into essential and
symptomatic clinical types.12 Different types of focal lesions of the spinal cord
can cause spinal segmental myoclonus.12
Peripheral Myoclonus
The term peripheral myoclonus should be reserved for instances where the
movement phenotype is myoclonus, although it may coexist with other phenotypes
as well. However, the distribution is defined by one or more peripheral nervous
Essential Myoclonus
The clinical presentation of essential myoclonus is characterized by a chronic and
relatively stable course in which myoclonus is a prominent or the only symptom.
The jerks interfere with coordination and may become moderate, but the patient
usually compensates without disability (CASE 8-2).17 Nonmovement neurologic
systems are usually not affected, but they occur in some patients.18
CONTINUUMJOURNAL.COM 1061
CONTINUUMJOURNAL.COM 1063
ii Postthalamotomy
iii Tumor
iv Trauma
v Inflammation (eg, multiple sclerosis)
vi Moebius syndrome
vii Developmental
B Peripheral nervous system
i Trauma
ii Hematoma
iii Tumor/infiltration
10 Malabsorption
A Celiac disease
B Whipple disease
11 Eosinophilia-myalgia syndrome
12 Exaggerated startle syndrome
A Hereditary
B Sporadic
13 Hashimoto encephalopathy (steroid-responsive encephalopathy associated with
autoimmune thyroiditis [SREAT])
14 Multiple system degenerations (genetic disorders not falling into another category)
A Allgrove syndrome
B DiGeorge syndrome
C Angelman syndrome
D Familial cortical myoclonic tremor and epilepsy
15 Primary progressive myoclonus of aging
16 Myoclonus associated with sleep
A Generalized
B Propriospinal
17 Unknown
HIV = human immunodeficiency virus; LGI1 = leucine-rich glioma inactivated protein 1; CASPR2 = contactin-
associated proteinlike 2.
a
Modified with permission from Marsden CD, et al, Movement Disorders.1 © 1982 Butterworths.
b
The genetics and clinical details of these etiologies are complex and are not given in detail.
CONTINUUMJOURNAL.COM 1065
Epileptic Myoclonus
Epileptic myoclonus occurs in syndromes that are traditionally considered to
be epilepsy (ie, a chronic seizure disorder).7 In most instances, the myoclonus
is the seizure itself and is called a myoclonic seizure. Less commonly, the
CASE 8-1 A 53-year-old man presented for evaluation of whole-body jerks. He was
accompanied by his wife, who reported that her spouse sometimes had
violent whole-body jerks as he was falling asleep. These may have been
going on for years but seemed more apparent recently. He had recently
been diagnosed with diabetes mellitus, and his wife was worried that
these jerks were related to his diabetes mellitus. His current medications
were insulin, losartan, atorvastatin, and aspirin. The patient denied
trouble with falling asleep, insufficient sleep, or excessive daytime
somnolence. He had no other neurologic concerns, and his examination
was normal. His EEG was normal.
He was diagnosed with hypnic jerks presenting as physiologic sleep
myoclonus. The patient and his wife were told that this jerking was of no
concern at that time. They were instructed to return to clinic if any jerks
were disrupting his sleep or preventing her from sleeping. Reassurance
was given that this is a normal phenomenon that varies in occurrence and
size among the healthy population.
COMMENT The patient in this case has myoclonus symptoms characteristic of the
physiologic clinical presentation category. Myoclonus occurs normally
in humans. In this patient, no disabling or functional consequence
accompanied the myoclonus. In fact, from the standpoint of the patient,
the myoclonus was asymptomatic. There was no connection to the
diagnosis of diabetes mellitus or any medication that the patient was
taking. It is typical that the patient or patient’s family expresses concern
about the myoclonus and its possible associations. The evaluation should
be guided by associated symptoms, the neurologic examination, and
plausible differential diagnosis. The EEG was ordered to rule out seizures,
since such generalized jerks may be epileptic in nature.
A 24-year-old woman presented with a history of “jerking as far back as I CASE 8-2
can remember.” She experienced bilateral jerking of the arms with
muscle activation and random jerks of the neck. Although these
movements were bothersome, she related only mild disability with
using her hands and arms for handwriting, eating, and other hand
movements. She reported no other symptoms. Rare alcohol
consumption decreased the jerking. She had been previously diagnosed
with essential tremor, and the patient was being treated with propranolol
with modest effect. The patient was adopted, her family history was
unknown, and she worked as a graduate student.
Her examination revealed myoclonic continuous small-to-moderate
multifocal jerking in random directions occurring unpredictably in
bilateral proximal upper extremities during muscle activation. A slight left
torticollis was present. Postural activation of the left arm showed mild
hyperpronation of the forearm. The rest of the examination was normal.
Laboratory evaluation showed no electrolyte disturbance or other toxic-
metabolic or inflammatory abnormality. Imaging and EEG were normal.
She was diagnosed with myoclonus-dystonia syndrome presenting as
essential myoclonus (classified as subcortical-nonsegmental physiology).
Propranolol was discontinued with only a mild detrimental effect, and
clonazepam 1 mg twice daily decreased the myoclonic jerking by more than
50% per patient report.
Some patients, like the one described here, can have repetitive rhythmic COMMENT
myoclonic movements of the upper limbs resembling the postural tremor
of essential tremor. The fact that the myoclonus was the primary symptom
and was chronic with little disability makes essential myoclonus the correct
clinical classification category. The reference to myoclonus-dystonia
means that both myoclonus and dystonia coexist in the patient. However,
in some cases, there may be mostly myoclonus or mostly dystonia.
Although no genetic testing was done, an ε-sarcoglycan or another gene
mutation could have been found in this patient. However, currently, such
genetic testing would not affect treatment, although it affords a more exact
diagnosis and possible future treatment implications. If symptoms become
severe and not adequately controlled with medication (see the section on
treatment for subcortical myoclonus), deep brain stimulation could be
considered.
CONTINUUMJOURNAL.COM 1067
but may not be perfectly symmetrical. Although often occurring in the early
morning hours, the seizures arise unpredictably regardless of the muscle state,
relaxation, or activation. Other types of generalized seizures (eg, tonic-clonic)
are common in patients with juvenile myoclonic epilepsy. There may be
photosensitivity and predisposing factors such as sleep deprivation, alcohol use,
and other stresses.
Symptomatic Myoclonus
This category comprises the most common clinical presentation category and
the largest list of myoclonus etiologies.3 In this category, myoclonus is secondary
to a medical or neurologic illness. Seizures may be a significant part of the
patient’s illness, but the seizure is not the major myoclonus phenotype as in
epileptic myoclonus. Other neurologic systems are typically affected and may
CASE 8-3 A 57-year-old woman presented for evaluation of episodes of left arm
jerking. The patient stated that these symptoms had occurred for
decades as sporadic jerking episodes lasting a few seconds. Recently,
they were occurring as a series of repetitive jerks occurring for 15 to
45 seconds on most days. These episodes were unpredictable, and the
jerks disabled the use of the arm while they were occurring. She reported
no other symptoms. Her past medical history revealed no known history
of seizures, and her family history was unremarkable.
Her examination was unremarkable except for a witnessed jerking spell
characterized by abrupt repetitive moderate-to-large myoclonus of the
left arm, at a frequency of about a 1 time per second. Multiple muscles
were involved. After several seconds, the jerks became less frequent and
abruptly stopped.
Her laboratory evaluation was normal. Imaging was normal. Interictal EEG
findings were normal, but EEG during a jerking spell showed rhythmic,
sharply contoured theta activity occurring over the right central head region.
She was diagnosed with focal motor seizures presenting as epileptic
myoclonus (cortical physiology). She was treated with carbamazepine,
resulting in nearly complete remission of the jerking episodes.
COMMENT These episodes presented as unpredictable myoclonus from rest and likely
represented focal motor seizures with epileptic myoclonus as the correct
clinical presentation classification category. The lack of an interictal EEG
abnormality should not dissuade a clinician from a seizure diagnosis. The
gross EEG cortical correlate during a jerking episode confirmed a cortical
physiology for this myoclonus. It is important to define the electrophysiologic
pattern as much as possible. Because these were focal seizures,
carbamazepine was a reasonable treatment option in this patient;
however, it should be noted that myoclonic seizures with generalized spike
and wave may become worse with carbamazepine. This highlights the
importance of distinguishing myoclonus etiologies within the epileptic
myoclonus category with assistance from the EEG pattern.
● Epileptic myoclonus
PROGRESSIVE MYOCLONIC EPILEPSY SYNDROMES. Various storage diseases
etiologies are chronic
fall under the clinical syndrome of progressive myoclonic epilepsy.22 Progressive seizure disorders that have
myoclonic epilepsy is a chronic, progressive, neurologic syndrome that manifests myoclonus as a prominent
as some combination of myoclonus, seizures, ataxia, and dementia. These phenomenon.
disorders usually affect younger individuals and are often fatal. Several clinical
● Symptomatic myoclonus
differences exist between individual storage diseases in their age of onset, rate is secondary to another
of progression, details of clinical expression, and other clinical manifestations. disorder, neurologic or
The neuropathology in the brain is widespread. Genetic testing is used for an non-neurologic. Multiple
increasing number of etiologies. other symptoms and signs
are usually present or tied to
definable pathology.
NEURODEGENERATION. Myoclonus occurs in chronic degenerative disorders,
including spinocerebellar disorders, basal ganglia disorders, and dementing
diseases. The syndrome dyssynergia cerebellaris myoclonica (also known as
Ramsey Hunt syndrome) refers to an idiopathic progressive myoclonic ataxia.23
Progressive myoclonic ataxia can appear similar to progressive myoclonic
epilepsy, but progressive myoclonic ataxia involves ataxia more than the
seizures that are seen in progressive myoclonic epilepsy. A variety of
progressive myoclonic conditions have been noted to have a predominant
progressive myoclonic ataxia rather than progressive myoclonic epilepsy
phenotype.24 Thus, distinguishing between a progressive myoclonic epilepsy
and progressive myoclonic ataxia phenotype can assist with differential
diagnosis.
Cortical myoclonus occurs across the spectrum of Lewy body disorders.25 The
myoclonus in Parkinson disease is small amplitude and may be confused with
tremor.26 In dementia with Lewy bodies, the myoclonus is larger and occurs in
about 58% of patients.27 The myoclonus that occurs in patients with dementia
with Lewy bodies is more likely to occur at rest than the myoclonus in
Parkinson disease.
Multiple system atrophy manifests as varying degrees of parkinsonism, ataxia,
and autonomic dysfunction. In the parkinsonian presentation, postural
activation triggers small-amplitude myoclonus in about 36% of patients, whereas
a stimulus-induced myoclonus to touch or muscle stretch is not uncommon in
the cerebellar presentations.28
Myoclonic jerks, both action and stimulus sensitive, commonly occur in
corticobasal degeneration. The reported range of the prevalence of myoclonus
in corticobasal degeneration is wide but previously thought to occur in the
majority of clinical cases. However, an autopsy series yielded 27%, suggesting
a smaller occurrence in pathologically confirmed cases.29 The distribution
of the myoclonus in corticobasal degeneration is either asymmetric or
focal and is similar in distribution to the other clinical manifestations of
the disease.
Myoclonus occurs in about 43% of patients with Alzheimer disease.27 The
usual appearance is small multifocal distal jerking, but widespread or generalized
jerks may be present. Myoclonus is a hallmark of Creutzfeldt-Jakob disease and
CONTINUUMJOURNAL.COM 1069
COMMENT The subacute onset and presence of abnormal mental status suggest that
this patient’s presentation was consistent with symptomatic myoclonus as
the correct clinical presentation classification category. Antibodies to the
voltage-gated potassium channel complex include the subtypes leucine-
rich glioma inactivated protein 1 (LGI1) and contactin-associated proteinlike
2 (CASPR2). Such antibodies may cause multiple neurologic syndromes.20 It
should be realized that this syndrome may present similarly to Creutzfeldt-
Jakob disease, but once discovered, it may be treatable with
immunosuppressive therapy.21 In this case, the patient responded to
treatment of the autoimmune encephalitis, and no symptomatic treatment
of the myoclonus was necessary.
CONTINUUMJOURNAL.COM 1071
MYOCLONUS TYPES
Certain myoclonus entities have characteristic presentations on examination and
are ingrained in the vernacular of clinicians, and have designations that are long
ingrained in the literature. These entities may be idiopathic or have variable
TABLE 8-2 Drug Classes and Examples of Drugs Associated With Myoclonusa
Psychiatric Medications
◆ Cyclic antidepressants
◆ Selective serotonin reuptake inhibitors (SSRIs)
◆ Monoamine oxidase inhibitors
◆ Lithium preparations
◆ Antipsychotic agents (including tardive syndrome)
Anti-infectious Agents
◆ Penicillins
◆ Carbapenem classes
Narcotics
◆ Morphine
◆ Fentanyl
Anticonvulsants
◆ Phenytoin
◆ Valproic acid
Anesthetics
◆ Lidocaine
◆ Midazolam
Contrast Media
Cardiac Medications
◆ Antiarrhythmics (eg, amiodarone, flecainide)
Calcium Channel Blockers
a
Modified with permission from Caviness JN, Brown P, Lancet Neurol.34 © 2004 Elsevier.
Palatal Myoclonus
The term palatal myoclonus coincides with a segmental distribution of and
around the soft palate. Expectedly, the physiologic classification is segmental.
It is the most common cause of segmental myoclonus and a common cause of
myoclonus in general.3 Some experts prefer the term palatal tremor. However,
a literature search reveals that palatal myoclonus is used in the majority of cases.
It is recommended that the determination be based on the phenotypic
appearance of the repetitive movement. Some examples do have a very rhythmic
and continuous sinusoidal cadence, and it is reasonable to refer to this as
palatal tremor. For movements that are jerky or somewhat irregular, palatal
myoclonus is the correct term.
Palatal myoclonus can be due to a variety of etiologies.37 Essential palatal
myoclonus parallels the characteristics of an essential myoclonus clinical
presentation. In essential palatal myoclonus, the muscle agonist is the tensor
veli palatini, and the myoclonus is frequently associated with an ear-clicking
sound, and often disappears with sleep (CASE 8-5). In the more common
symptomatic palatal myoclonus, the palatal movement is due to contractions
of the levator veli palatini, has other symptoms related to the causative lesion,
and is more persistent. Symptomatic palatal myoclonus is believed to arise from
disruption within the Guillain-Mollaret triangle, a pathway connecting the red
nucleus to the inferior olivary nucleus (central tegmental tract), the inferior
olivary nucleus to the dentate nucleus (inferior cerebellar peduncle), and the
dentate nucleus to the red nucleus (superior cerebellar peduncle). Essential
cases are idiopathic with or without a family history. Symptomatic cases are most
often due to a structural lesion (eg, stroke).
Propriospinal Myoclonus
Propriospinal myoclonus was described by Brown and colleagues9 in 1991. It
arises from a subcortical-nonsegmental physiology within the spinal cord in
which the trigger then travels rostrally and caudally simultaneously via slow
propriospinal pathways. Extension or flexion trunk jerks may occur (CASE 8-6).
Cases may be idiopathic, but lesions are common. Multiple reports exist of
propriospinal myoclonus occurring during transition into sleep, when waking,
or both.39
Orthostatic Myoclonus
Orthostatic myoclonus manifests as leg shaking when the patient is standing,
similar to orthostatic tremor. In contrast to orthostatic tremor, orthostatic
myoclonus produces much more difficulty with gait and has a much higher
prevalence of associated neurologic problems.40 It can be difficult to distinguish
between these two orthostatic movement disorders because of significant clinical
overlap on history and examination. However, the surface EMG pattern in the
lower extremities appears to be different between orthostatic tremor and
orthostatic myoclonus. Instead of the rhythmic 13-Hz to 16-Hz surface EMG
discharges that are seen with orthostatic tremor, orthostatic myoclonus shows
lower frequency and more discharge duration variability with many brief
CONTINUUMJOURNAL.COM 1073
Cortical Tremor
The myoclonus type known as cortical tremor was first described in 1990.41
It has the classic features of cortical physiology, including enlarged cortical
somatosensory evoked potentials. However, the myoclonus EMG discharges
occur rhythmically or semirhythmically at 6 Hz to 9 Hz, which mimics a
nonspecific muscle activation tremor. Cortical tremor can occur from different
etiologies. A particular syndrome, familial autosomal dominant cortical tremor,
myoclonus, and epilepsy, is now a defined syndrome.42
Minipolymyoclonus
Minipolymyoclonus is a form of multifocal myoclonus and is characterized by
small jerks in different locations.43 This term has also been used to describe
small-amplitude jerks in patients with spinal muscular atrophy. Hence, the
CASE 8-5 A 33-year-old woman presented because of “an obnoxious sound” in her
ears. When it began 5 to 7 years ago, she thought that it was simply
“ringing in her ears” from listening to loud music and just a nuisance.
Lately, the sounds were distracting her. She had no other symptoms. Her
past medical, surgical, and family history were unremarkable.
Examination revealed audible clicking from both ears. Inspection of
the soft palate showed that the roof of the soft palate was rhythmically
moving. She had no other movements of the mouth, face, or elsewhere.
The rest of her neurologic examination was normal. Laboratory testing
was unremarkable. Brain MRI was normal. She was diagnosed with
essential palatal myoclonus (tremor) presenting as essential myoclonus
with a segmental physiology. Treatment with clonazepam was started but
did not change the clicks. Botulinum toxin injection into the tensor veli
palatini muscle eliminated the clicks.
COMMENT The clinical course in this patient, without other signs and symptoms and
the relative slow progression, was most consistent with the clinical
presentation of essential myoclonus. Symptomatic myoclonus begins
more abruptly but may be delayed after an acute lesion, nevertheless. In
addition, symptomatic palatal myoclonus often involves other areas of the
mouth or face. Another distinction is that cases of essential palatal
myoclonus have the click as the primary symptom, whereas cases of
symptomatic cases mostly have other symptoms from the lesion, such as
dysphagia, numbness, and ataxia. Essential palatal myoclonus/tremor
usually shows a rhythmic movement that resembles tremor, justifying the
label of essential palatal tremor. Evidence in the literature supports
botulinum toxin as a viable treatment option.38 However, the potential risks
must be clearly delineated. In many centers, the injection is performed by
an otolaryngologist for safety reasons.
TREATMENT OF MYOCLONUS
The etiology of the patient’s disorder should be the most important consideration
when deciding on treatment because symptomatic treatment has limitations.
If treatment of the etiology is not possible or is delayed, then symptomatic
treatment can be considered. Some treatments of myoclonus may worsen
cognitive status or movement coordination; therefore, the treatment of
myoclonus must be weighed against likely side effects. Moreover, if
A 72-year-old woman presented for evaluation of lower thoracic jerking CASE 8-6
that had slowly increased over many months. These jerks caused her to
arch her back and were emanating from her lower thoracic spine. She
stated that this caused a very uncomfortable sensation that would run up
and down her spine; this sensation was just as unsettling as, if not more
than, the movement itself. She had a long history of aching back and neck
pain. Her past medical history included hypertension and esophageal
reflux.
Examination showed trunk extension myoclonus that was worse when
she was sitting than when recumbent or standing. Sensitivity to tactile
stimulation in the lower thoracic spine was present but inconsistent. There
were no upper motor neuron signs.
Brain and cervical spine MRI and EEG were normal. MRI of the thoracic
spine showed a herniated disc at T10 which caused mild edema at that level
of the spinal cord.
She was diagnosed with a T10 central herniated disk producing
propriospinal myoclonus presenting as symptomatic myoclonus with a
subcortical-nonsegmental physiology. The thoracic disk herniation was
repaired surgically, and the myoclonus was not present a few weeks after
the operation.
CONTINUUMJOURNAL.COM 1075
Cortical Myoclonus
Levetiracetam is usually the first-line treatment of cortical myoclonus. This
drug is chemically related to piracetam, an older drug used previously for
posthypoxic myoclonus in countries where it was available. Levetiracetam
(1000 mg/d to 3000 mg/d divided into 2 daily doses) has shown effectiveness in
a few small studies where cortical physiology is present.44–47 Clonazepam
(1 mg/d to 3 mg/d divided into 2 or 3 daily doses) or valproic acid (500 mg/d to
2000 mg/d divided into 2 or 3 daily doses) may be used if levetiracetam is
discontinued for any reason or in combinational therapy for myoclonus.48–50
Sodium oxybate has shown some efficacy in case reports.51 Brivaracetam, a drug
with structural similarity to levetiracetam, has not shown consistent efficacy for
cortical myoclonus.52 Perampanel has shown efficacy in cortical myoclonus,
including in a study with 12 patients with Unverricht-Lundborg disease.53
Initially, deep brain stimulation was met with some skepticism for cortical
myoclonus. However, more reported cases with positive results suggest a
controlled trial is needed to determine case-based efficacy and the best site
for stimulation.54
Cortical-Subcortical Myoclonus
The primary generalized epilepsy syndromes that produce myoclonus (eg,
juvenile myoclonic epilepsy, absence syndromes) usually receive valproic acid
(500 mg/d to 2000 mg/d divided into 2 or 3 daily doses) as the initial antiseizure
CONTINUUMJOURNAL.COM 1077
CONCLUSION
The many types and etiologies of myoclonus may at first seem confusing. An
efficient and pragmatic evaluation of myoclonus that uses the clinical
presentation classification scheme offers the best approach to define the etiology.
Further evaluation that results in physiologic classification can lead to further
understanding of the myoclonus. Combining the information from the etiology
with the determined or presumed physiology is used to develop the best
treatment strategy. Treatment is often unsatisfactory, and more research into
treatment and controlled clinical studies are desperately needed. Rather than
presuming that myoclonus arises from a single area, it may be that a network
abnormality produces myoclonus in some syndromes. Further research on
myoclonus generation mechanisms should shed light on future treatment
possibilities.
REFERENCES
1 Marsden CD, Hallett M, Fahn S. The nosology and 8 Genton P, Thomas P, Kasteleijn-Noist Trenité DG,
pathophysiology of myoclonus. In: Marsden CD, et al. Clinical aspects of juvenile myoclonic
Fahn S, editors. Movement disorders. London, epilepsy. Epilepsy Behav 2013;28(suppl 1):S8–S14.
United Kingdom: Butterworths, 1982:196–248. doi:10.1016/j.yebeh.2012.10.034.
2 van der Salm SM, de Haan RJ, Cath DC, et al. The 9 Brown P, Thompson PD, Rothwell JC, et al. Axial
eye of the beholder: inter-rater agreement myoclonus of propriospinal origin. Brain 1991;
among experts on psychogenic jerky movement 114(pt 1A):197–214. doi:10.1093/oxfordjournals.
disorders. J Neurol Neurosurg Psychiatry 2013; brain.a101857.
84(7):742–747. doi:10.1136/jnnp-2012-304113.
10 Roze E, Apartis E, Clot F, et al. Myoclonus-
3 Caviness JN, Alving LI, Maraganore DM, et al. The dystonia: clinical and electrophysiologic pattern
incidence and prevalence of myoclonus in related to SGCE mutations. Neurology 2008;
Olmsted County, Minnesota. Mayo Clin Proc 70(13):1010–1016. doi:10.1212/01.wnl.0000297516.
1999;74(6):565–569. doi:10.4065/74.6.565. 98574.c0.
4 Caviness JN. Treatment of myoclonus. 11 Caviness JN. Segmental Myoclonus. In: Albanese
Neurotherapeutics 2014;11(1):188–200. doi: A, Jankovic J, editors. Hyperkinetic movement
10.1007/s13311-013-0216-3. disorders. West Sussex, United Kingdom:
Wiley-Blackwell, 2012:221–236.
5 Caviness JN. Clinical neurophysiology of myoclonus.
In: Hallett M, editor. Movement disorders. 12 Deuschl G, Mischke G, Schenck E, et al.
Handbook of clinical neurophysiology. Vol 1. Symptomatic and essential rhythmic palatal
Amsterdam, the Netherlands: Elsevier, 2003: myoclonus. Brain 1990;113(pt 6):1645–1672.
521–548. doi:10.1093/brain/113.6.1645.
6 Avanzini G, Shibasaki H, Rubboli G, et al. 13 Bono F, Salvino D, Sturniolo M, et al. Botulinum
Neurophysiology of myoclonus and progressive toxin is effective in myoclonus secondary to
myoclonus epilepsies. Epileptic Disord 2016; peripheral nerve injury. Eur J Neurol 2012;19:
18(S2):11–27. doi:10.1684/epd.2016.0835. e92–e93. doi:10.1111/j.1468-1331.2012.03780.x.
7 Caviness JN. Epileptic myoclonus. In: Sirven JI, 14 Rose Russell AL, Knight W, Oware A, Fuller GN.
Stern JM, editors. Atlas of video-EEG monitoring. A unique case of peripheral myoclonus. J Neurol
New York, NY: McGraw-Hill Medical, 2011: Neurosurg Psychiatry 2012;84(1):117–118. doi:
309–328. 10.1136/jnnp-2012-303795.
CONTINUUMJOURNAL.COM 1079
41 Ikeda A, Kakigi R, Funai N, et al. Cortical tremor: a 52 Kälviäinen R, Genton P, Andermann E, et al.
variant of cortical reflex myoclonus. Neurology Brivaracetam in Unverricht-Lundborg disease
1990;40(10):1561–1565. doi:10.1212/WNL.40.10.1561. (EPM1): results from two randomized,
double-blind, placebo-controlled studies.
42 Coppola A, Santulli L, Del Gaudio L, et al. Natural
Epilepsia 2016;57(2):210–221. doi:10.1111/epi.13275.
history and long-term evolution in families with
autosomal dominant cortical tremor, myoclonus, 53 Crespel A, Gelisse P, Tang NP, Genton P.
and epilepsy. Epilepsia 2011;52(7):1245–1250. Perampanel in 12 patients with Unverricht-
doi:10.1111/j.1528-1167.2011.03017.x. Lundborg disease. Epilepsia 2017;58(4):543–547.
doi:10.1111/epi.13662.
43 Wilkins DE, Hallett M, Erba G. Primary generalized
epileptic myoclonus: a frequent manifestation of 54 Ramdhani RA, Frucht SJ, Kopell BH. Improvement
minipolymyoclonus of central origin. J Neurol of post-hypoxic myoclonus with bilateral pallidal
Neurosurg Psychiatry 1985;48(6):506–516. deep brain stimulation: a case report and review
of the literature. Tremor Other Hyperkinet Mov
44 Genton P, Gélisse P. Antimyoclonic effect of
(N Y) 2017;7:461. doi:10.7916/D8NZ8DXP.
levetiracetam. Epileptic Disord 2000;2(4):
209–212. 55 Nevitt SJ, Sudell M, Weston J, et al. Antiepileptic
drug monotherapy for epilepsy: a network
45 Striano P, Manganelli F, Boccella P, et al.
meta-analysis of individual participant data.
Levetiracetam in patients with cortical myoclonus:
Cochrane Database of Systematic Reviews
a clinical and electrophysiological study.
2017;(12):CD011412. doi:10.1002/14651858.CD011412.
Mov Disord 2005;20(12):1610–1614. doi:10.1002/
pub3.
mds.20530.
56 Buchanan N. The use of lamotrigine in juvenile
46 Frucht SJ, Louis ED, Chuang C, Fahn S. A pilot
myoclonic epilepsy. Seizure 1996;5:149–151.
tolerability and efficacy study of levetiracetam in
doi:10.1016/S1059-1311(96)80110-5.
patients with chronic myoclonus. Neurology
2001;57(6):1112–1114. doi:10.1212/WNL.57.6.1112. 57 Steinig I, von Podewils F, Möddel G, et al.
Postmarketing experience with brivaracetam in
47 Magaudda A, Gelisse P, Genton P. Antimyoclonic
the treatment of epilepsies: a multicenter cohort
effect of levetiracetam in 13 patients with
study from Germany. Epilepsia 2017;58(7):
Unverricht-Lundborg disease: clinical
1208–1216. doi:10.1111/epi.13768.
observations. Epilepsia 2004;45(6):678–681.
doi:10.1111/j.0013-9580.2004.56902.x. 58 Caviness JN, Forsyth PA, Layton DD, McPhee TJ.
The movement disorder of adult opsoclonus.
48 Goldberb MA, Dorman JD. Intention myoclonus:
Mov Disord 1995;10(1):22–27. doi:10.1002/
successful treatment with clonazepam. Neurology
mds.870100106.
1976;26(1):24–26. doi:10.1212/WNL.26.1.24.
59 Rocha H, Linhares P, Chamadoira C, et al. Early
49 Fahn S. Posthypoxic action myoclonus: review of
deep brain stimulation in patients with
the literature and report of two new cases with
myoclonus-dystonia syndrome. J Clin Neurosci
response to valproate and estrogen. Adv Neurol
2016;27:17–21. doi:10.1016/j.jocn.2015.08.034.
1979;26:49–84.
60 Rath JJ, Tavy DL, Snoeck-Streef I, Contarino MF.
50 Obeso JA, Artieda J, Rothwell JC, et al. The
Sustained remission of segmental myoclonus
treatment of severe action myoclonus. Brain
due to peripheral nerve injury after treatment
1989;112(pt 4):765–777. doi:10.1093/brain/
with onabolinumtoxinA. Parkinsonism Relat
112.3.765.
Disord 2015;21(9):1111–1112. doi:10.1016/j.
51 Arpesella R, Dallocchio C, Arbasino C. A patient parkreldis.2015.07.001.
with intractable posthypoxic myoclonus
61 Chaudhry N, Srivastava A, Joshi L. Hemifacial
(Lance–Adams syndrome) treated with sodium
spasm: the past, present and future. J Neurol Sci
oxybate. Anaesth Intensive Care 2009;37:
2015;356(1–2):27–31. doi:10.1016/j.jns.2015.06.032.
314–318.
By Joseph H. Friedman, MD, FAAN, FANA C O N T I N U UM A U D I O
I NT E R V I E W A V A I L AB L E
ONLINE
ABSTRACT VIDEO CONTENT
PURPOSE OF REVIEW: This
article reviews the history, nosology, clinical A V AI L A B L E O N L I N E
T
Institutes of Health.
he first antipsychotic drug, chlorpromazine, was introduced in France
in 1952 and in the United States in 1954. Acute and subacute motor UNLABELED USE OF
side effects were recognized early, but the first report of a tardive PRODUCTS/INVESTIGATIONAL
USE DISCLOSURE:
syndrome was first published in 1957.1,2 More cases came to light, and Dr Friedman discusses the
the condition, initially thought rare or nonexistent, was recognized as unlabeled/investigational use of
a significant problem.1,2 The term tardive dyskinesia, referring to the late onset, α-methyl-para-tyrosine,
botulinum toxin, clozapine,
was coined in 1964 and caught on.3 deep brain stimulation,
The reasons for doubting that a tardive syndrome was the result of a drug reserpine, and tetrabenazine for
the treatment of tardive
effect were much more understandable at the time than they seem in
syndromes.
retrospect. A wide spectrum of abnormal movements had been associated with
psychiatric disorders by psychiatrists of the early 20th century,4 raising the question
of whether the movements were part of the disease. In that era, little clarity for © 2019 American Academy
diagnoses existed, and a large percentage of people in psychiatric asylums had of Neurology.
CONTINUUMJOURNAL.COM 1081
tertiary syphilis and other disorders, including forms of autism and other behavioral
disorders that could be associated with movement disorders.5 The second
observation was that the purported side effects, usually oral-buccal-lingual
dyskinesias, improved when the offending drug dose was increased and worsened
when the drug was decreased. These observations conflicted with canonical
thinking about drug side effects, which assumed that higher doses of a drug
that causes a side effect should result in more, not fewer, side effects. A third
observation was the variable nature of the movements, the variable time to
onset, and the lack of an apparent dose-effect relationship. It should be noted that
the relationship between drugs and tardive disorders rests on epidemiology, as
no mechanism has yet been elucidated (refer to the following section on
pathophysiology).
CLINICAL ASPECTS
The tardive syndromes may include overlapping signs, but there are often very
clear discriminating features that allow for distinct categorization. The signs,
symptoms, and nosology are important because treatments may be different for
each tardive syndrome.
Nosology
The term tardive dyskinesia is commonly used in two ways. It is often used
as an umbrella term to include all the tardive syndromes as well as the
most commonly recognized syndrome, oral-buccal-lingual dyskinesia. In this
article the term tardive syndrome6 will be used as the inclusive umbrella term,
and tardive dyskinesia will refer to the various choreoathetoid or stereotypic
movements, including oral-buccal-lingual dyskinesias. The American Psychiatric
Association’s Diagnostic and Statistical Manual of Mental Disorders, Fifth
Edition (DSM-5) defines tardive dyskinesia as: “involuntary athetoid or
choreiform movements (lasting at least a few weeks)… developing in association
with the use of a neuroleptic medication for at least a few months. Symptoms
may develop after a shorter period of medication use in older persons.”7 It
is a modification of the definition from the Diagnostic and Statistical Manual
of Mental Disorders, Fourth Edition, Text Revision (DSM-IV-TR), which
required continuous exposure to a neuroleptic for at least 3 months for those
younger than 60 years of age and continuous exposure for 1 month or more
for those older than 60 years of age, and the movement disorder needed
to develop either while the patient was on the drug or within 4 weeks of
stopping it. Rare cases caused by much shorter exposure have been described.8
The diagnoses of tardive dystonia and tardive akathisia are also listed in
the DSM-5.
Although some authors have embraced the idea that drugs that do not
block dopamine receptors may also cause clinical syndromes identical to a tardive
syndrome, this is uncertain.9 Almost all cases of tardive syndromes have occurred
in patients who developed symptoms after another psychoactive drug was added
to a dopamine receptor–blocking drug, in patients who had a history of dopamine
receptor–blocking drug use, or in patients whose history had not specifically
excluded exposure to dopamine receptor–blocking drugs. In many cases, the
movement disorder lasted only a brief time after the non–dopamine receptor–
blocking drug was discontinued and therefore did not meet clinical criteria for a
tardive syndrome. In a review of this topic, D’Abreu and colleagues9 asserted
CONTINUUMJOURNAL.COM 1083
CASE 9-1 A 77-year-old man presented for a neurologic evaluation for symptoms
of parkinsonism consisting of resting tremor, soft speech, slower
movements, and imbalance. He had been taking aripiprazole 5 mg/d for
3 years for bipolar disorder. It was the only neuroleptic he had ever taken.
A previous trial off aripiprazole had lasted only 2 weeks because of
worsened depression. His tremors had improved during that time.
His initial neurologic examination was normal aside from parkinsonism,
manifest by masked facial expression, resting tremor, bradykinesia,
and typical gait. He had a Unified Parkinson’s Disease Rating Scale,
part III, score of 36 and a Hoehn and Yahr stage of 2.5. His Abnormal
Involuntary Movement Scale (AIMS) score was 0. A dopamine transporter
scan was normal.
Aripiprazole was discontinued 11 days before his next follow-up
4 months later. Off his neuroleptic, his AIMS score was 4 but he was not
bothered by his symptoms.
Eight months later he returned for follow-up (VIDEO 9-1, links.lww.com/
CONT/A362). He and his wife reported that involuntary mouth
movements had worsened 1 month prior. The patient had continuous
chewing movements and tongue writhing. His jaw moved up and down
but also laterally. His lips appeared to not be involved but because of the
continuous tongue movements, it was difficult to be certain. The rest of
his face was not involved. He had stereotypic rubbing movements of his
fingers and dyskinetic movements of both legs, left greater than right.
The mouth movements were unlikely to be due to his being edentulous
because he was not rubbing his gums together and had the other
movements that are typical of oral, buccal, and lingual tardive dyskinesia.
His AIMS score was 13 with a severity rating of 4 (severe), and his Unified
Parkinson’s Disease Rating Scale motor score was 13.
COMMENT Several weeks after aripiprazole was stopped, this patient’s parkinsonism
improved but involuntary movements in his mouth developed. He was
edentulous, which may have contributed to the movements. The
emergence of a tardive syndrome when the offending drug is stopped is
not uncommon. When evaluating any patient on a neuroleptic, the
physician must evaluate both for parkinsonism and for a tardive syndrome.
Any patient on a neuroleptic is supposed to have an AIMS evaluation on a
regular basis, but the possibility of a tardive syndrome being masked by the
neuroleptic must always be kept in mind. It is also important to recognize
that second-generation antipsychotics may cause both parkinsonism and
tardive syndrome.
The patient was not treated for the movements because of medical and
psychiatric issues so the “natural history” of the disorder (ie, 19 months off
dopamine-blocking drugs) was evident, with improved parkinsonism and
minimal change in the tardive syndrome.
This patient was treated with tetrabenazine before the drugs approved by COMMENT
the US Food and Drug Administration (FDA) were available. Tetrabenazine
was initially very helpful but required a high dose, which eventually caused
severe parkinsonism. As her parkinsonism lessened, with the reduction in
tetrabenazine then the trials of deutetrabenazine and valbenazine, her
tardive dyskinesia reemerged. Having failed all three drugs, she was
referred for globus pallidus internus deep brain stimulation.
CONTINUUMJOURNAL.COM 1085
TARDIVE TICS. Vocal and motor tics may mimic Tourette syndrome. Unlike
Tourette syndrome, the syndrome of tardive tics is late in onset and develops
only after chronic exposure to dopamine receptor–blocking drugs. It is rare, and
at least one case with coprolalia has been reported (CASE 9-4).
CASE 9-3 A 45-year-old woman was intially referred for evaluation of severe
restlessness. She had been diagnosed with schizoaffective disorder at
age 30 and was treated with several first-generation antipsychotic drugs,
including haloperidol, chlorpromazine, thioridazine, and others. She
reported being extremely uncomfortable due to her restlessness and
could not keep from moving. She denied feeling anxious. She stated that
this had been present for about 4 years. It did not vary significantly during
the day, but resolved during sleep. She denied paresthesia or abnormal
sensations in her legs. Her husband did not report leg movements during
sleep. Although her antipsychotics had been altered during that time, her
movements had persisted. She had not improved with benztropine or
amantadine. She had no history of thyroid disease, substance abuse, or
medical problems. Her psychotic symptoms were controlled but she felt
helpless and depressed.
At the time of her initial neurologic evaluation, she was taking
haloperidol 2 mg/d and nortriptyline 100 mg/d. Other than the findings
shown in the video (VIDEO 9-4, links.lww.com/CONT/A365), her neurologic
examination was normal except for mild oral, buccal, and lingual
dyskinesias. She did not have parkinsonian signs.
She was diagnosed with akathisia. At the time of her initial evaluation,
the only drugs available to treat this were reserpine and α-methyl-para-
tyrosine. Tetrabenazine and its derivatives were not then available. It was
recommended that she be treated with clozapine once it became available
6 years later, which replaced her haloperidol. Her akathisia resolved
completely when the haloperidol was replaced by the clozapine, but the
dyskinetic mouth movements were unchanged.
VIDEO 9-5 (links.lww.com/CONT/A366) shows the same patient on
follow-up 33 years later. She has continuous writhing movements of her
tongue as well as continuous lip and jaw movements.
CONTINUUMJOURNAL.COM 1087
CASE 9-4 A 61-year-old woman with schizoaffective disorder was referred for
management of a movement disorder. She was unaware of her
movements, which her family noticed only within the past year. Once
pointed out to her, they did not bother or concern her. She did not
think that they attracted attention.
She had been treated with neuroleptics for 13 years, including
haloperidol, perphenazine, risperidone, aripiprazole, ziprasidone, and
brexpiprazole and was currently taking lurasidone. She liked being on
lurasidone because it controlled her auditory hallucinations and made
her feel better than she had on any of the other antipsychotics. She and
her family denied her having tics as a child and she had no family
history of tics or other movement disorders. She did not have
compulsive behaviors. Her identical twin had no tics or other
movement disorders. She had no history of other neurologic problems.
She was treated for hypertension, hypothyroidism, and increased
cholesterol. Her thyroid hormone levels had been therapeutic. Her
medications included lurasidone 40 mg/d.
Her neurologic examination showed mild parkinsonism, with mild facial
masking, a mildly stooped posture, and absent arm swing; her tics
manifested by sudden extension or flexion of either knee while sitting
VIDEO 9-7 (links.lww.com/CONT/A368), and she exhibited facial tics.
COMMENT This patient’s movements were tics. They primarily involved her legs, right
more than left. She also had facial tics, more commonly seen on the right,
affecting both the upper and lower face. It is unusual that she did not
perceive the movements, as most tics are associated with an urge to move.
Her lack of concern for attracting attention may reflect her social isolation,
her psychiatric disorder, or her personality. The standard approach to
treating the tardive tics would be replace her lurasidone with quetiapine
and then treat the tics with either valbenazine or deutetrabenazine. If the
quetiapine did not adequately control her psychotic symptoms, then
clozapine would be instituted. However, both drugs are associated with
weight gain, which was a major concern for this patient. She spontaneously
remarked how happy she had been on lurasidone, in contrast to the many
drugs she’s failed, and was very reluctant to change. In addition, she was
not aware of her movements, other than for being told about them, and did
not care about them. Since the natural history of these movements was
unknown, and may not worsen, the recommendation was to not alter
medications and simply follow her. Her movements, had they been
bothersome, could have been treated by adding valbenazine or
deutetrabenazine to her regimen of lurasidone.
Tardive dyskinesia Choreoathetoid or stereotypic movements, which can affect any body part, but most
commonly the oral, buccal, lingual region
Tardive dystonia Dystonia, a sustained, involuntary muscle contraction, often writhing in nature, producing
an abnormal posture; this may cause torticollis, blepharospasm, jaw opening or closing
dystonia, or affect other body parts
Tardive akathisia An uncomfortable feeling of restlessness causing movements such as marching in place,
pacing, rocking in place while seated, and rubbing hands
Pseudoakathisia Overlaps with or may be considered stereotypy: patients are in constant motion as if
restless but deny the feeling of restlessness
Tardive stereotypy Repetitive, purposeless movements, such as rocking, crossing legs, rubbing hands when
not feeling restless
Tardive tremor and tardive A debated syndrome since many authorities believe that cases reported as tardive tremor
parkinsonism or tardive parkinsonism really had essential tremor or idiopathic Parkinson disease.
Tardive pain A pain disorder associated with one of the above disorders that is not directly caused by
the movements themselves
Tardive myoclonus Myoclonic lightninglike jerk affecting isolated muscles causing an obvious limb or trunk
jerk or vocalization
CONTINUUMJOURNAL.COM 1089
Tardive Dyskinesia
◆ Idiopathic oral facial dyskinesias of the elderly
◆ Stereotypic chewing in the edentulous
◆ Stereotypies in patients with autism
◆ Teeth, gum, tongue, mouth disorders (loose dentures)
◆ Chorea (inherited, metabolic, inflammatory, structural)
◆ Drug toxicity (eg, phenytoin, lithium)
◆ Levodopa-induced dyskinesias in a patient with Parkinson disease
◆ Psychogenic
◆ Tics
Tardive Stereotypies
◆ Behaviors unrelated to drugs
◆ Sensory neuropathies (“piano playing fingers”)
◆ Tactile hallucinations
◆ Anxiety
◆ Obsessive-compulsive disorder
◆ Tics
Tardive Akathisia
◆ Anxiety
◆ Drug withdrawal/drug intoxication
◆ Psychotic/other psychogenic internal stimuli
◆ Chorea
Tardive Dystonia
◆ Inherited or brain injury
◆ Peripheral injury
◆ Psychogenic
◆ Presenting feature of Parkinson disease or other neurodegenerative disorder
Tardive Myoclonus
◆ Tics
◆ Toxic or metabolic disorders
Tardive Tics
◆ Tourette syndrome
a
This table lists disorders that may be confused with the tardive syndromes.
Evaluation
The Abnormal Involuntary Movement Scale (AIMS) has become the standard
evaluation tool for tardive syndrome studies. The AIMS is focused on
choreoathetoid movements and stereotypies. It does not capture akathisia,
which is usually measured with the Barnes Akathisia Rating Scale.18 The
usual clinical research criteria for diagnosing tardive dyskinesia are the
Schooler-Kane19 criteria, which require scores of 2 (mild) or greater in at
least two body parts or one score of 3 (moderate) or greater in one body part,
while meeting the contemporary DSM-5 criteria for onset and duration.
TABLE 9-2 lists the differential diagnosis of the tardive syndromes.
CONTINUUMJOURNAL.COM 1091
PATHOPHYSIOLOGY
The pathophysiology of tardive syndrome is unknown. The most “popular” and
clearly the most heuristically appealing theory of tardive syndrome is the dopamine
receptor supersensitivity, or “up-regulation” theory.8,11,26 The hypothesis presumes
that, in some patients, long-term exposure to dopamine receptor–blocking drugs
causes an increase in D2 receptors or in their sensitivity. This is based largely on the
observation that dopamine receptor–blocking drugs or drugs that reduce dopamine
stimulation reduce the movements. The theory is further supported by rodent
studies showing that dopamine receptor–blocking drugs do cause receptor
hypersensitivity, but human studies have produced mixed results,27,28 and
long-term exposure of dopamine receptor–blocking drugs in rodents also do not
support this hypothesis. Alternative hypotheses include structural changes in the
synapse26,29 or the presynaptic dopamine-secreting neuron29 due to direct toxic
effects of the drugs, or indirectly via oxidative stress, genetic anomalies affecting
dopamine receptors, and alterations of RNA production affecting other
neurotransmitters induced by dopamine receptor–blocking drugs.
The synaptic plasticity hypothesis rests on the observation that synapses in the
human neocortex are altered by dopamine receptor–blocking drugs, as well as
in the striatum of rodents, and that abnormal plasticity is found in animal models
of hyperkinetic human disorders.26 The theory posits that the movements are
generated by abnormal signal processing.
Another hypothesis proposes that dopamine receptor–blocking drugs cause a loss
of dopamine-secreting cells in the substantia nigra, which, in turn, causes
“denervation supersensitivity and tardive dyskinesia.”30 However, data on loss of
neurons in the human substantia nigra in patients on chronic antipsychotics are
conflicting.31,32 An oxidative stress theory proposes an increase in hydrogen
peroxide and free radicals as a result of increased dopamine turnover resulting from
dopamine receptor blockade. Many human genetic studies reveal correlations
between tardive syndromes and a large number of dopamine, serotonin, and other
neurotransmitter receptors, suggesting the possibility that the genetics of a number
of different neuroreceptor subtypes may explain both the risk of developing a
syndrome and the type and severity of the disorder. Animal studies have revealed
changes in gene expression in the brains after chronic dopamine receptor–blocking
drug exposure, raising a question as to whether this may occur in humans as well.
EPIDEMIOLOGY
In American nursing homes, 20% of residents take antipsychotics. In addition,
some neuroleptics are also approved in the United States for treating depression
and are among the best-selling drugs in the country. As the elderly are at greatest
risk for the development of a tardive syndrome, having a fivefold greater risk
than younger patients,33 it is likely that many instances of tardive dyskinesia go
unnoticed in nursing homes.
The development of second-generation antipsychotics was thought to herald
the beginning of the end of tardive syndromes. Clozapine, the first “atypical”
antipsychotic, has not been linked to a tardive disorder except when there has
RISK FACTORS
Most reports on risk factors for tardive syndromes have not been confirmed.
Older age as a risk factor has the most support. In cohorts of older versus
CONTINUUMJOURNAL.COM 1093
younger patients exposed to neuroleptics for the same period of time, the older
patients are more likely to develop a tardive syndrome. Other cited factors are
not so reliable. Presumed racial differences may be explained by different
prescribing and diagnosing patterns. African Americans are thought to have a
higher risk than white Americans, but this is confounded by the observation that
African Americans generally received higher doses of antipsychotics, and racial
distinctions may not be scientifically accurate. It could also be argued that this
higher dosing might actually mask the syndrome, reducing the perceived risk. A
history of early-onset extrapyramidal side effects such as acute dystonic
reactions, akathisia, or parkinsonism has been suggested but not confirmed as
risk factors for later onset of a tardive syndrome. Brain damage has also been
suggested as a risk factor, but this is a difficult risk factor to quantify. The
presence of an affective disorder has been proposed to increase the risk. Female
gender, duration of antipsychotic use, dose of the antipsychotic, dementia,
diabetes mellitus, HIV infection, intellectual disability, brain damage, and
anticholinergic use all have soft support for being risk factors.40
TREATMENT
Anticholinergic drugs were commonly used when neuroleptics were initiated
to reduce the acute extrapyramidal side effects, particularly acute dystonic
reactions, and they were effective for this purpose. They were also commonly
used to treat parkinsonian side effects, with much less clear benefit. This
common use was expanded so that they have frequently been used to treat
tardive syndromes, particularly dyskinetic ones, although no evidence
supports their utility, and it is likely that they worsen the tardive syndromes,
as anticholinergic drugs worsen choreic movements in general; additionally,
for patients with oral, buccal, lingual movements, the dry mouth caused by
anticholinergics increases the discomfort and the movements themselves.
Only two treatments approved by the US Food and Drug Administration
(FDA) exist for tardive syndromes: valbenazine and deutetrabenazine, both
of which are tetrabenazine-related drugs. They are similar to the previous
standard therapies, reserpine, tetrabenazine, and α-methyl-para-tyrosine,
in that they reduce dopamine stimulation. They, like tetrabenazine, are
selective vesicular monoamine transporter inhibitors type 2, which reduce the
incorporation of dopamine (as well as histamine, serotonin, and norepinephrine)
into vesicles, hence reducing dopamine stimulation. Deutetrabenazine has a
deuterated hydrogen atom, while valbenazine is a prodrug of the most active isomer
of tetrabenazine, both of which have significantly longer serum half-lives than
tetrabenazine, leading to a more sustained pharmacokinetic profile, possibly
explaining their much reduced side effects, avoiding depression and parkinsonism41
in particular. Unfortunately, these two drugs cost between $65,000 and $100,000
each year.
Four Class I trials of both drugs41 have shown significant reductions in
AIMS scores, the primary outcome variable, and no depression or parkinsonism
within the 6 to 12 week study and open-label extensions. The studies did not
distinguish the various types of tardive syndromes, however. The drugs
probably are more effective for dyskinesias, stereotypies, and akathisia than
dystonia or myoclonus. Interestingly, patients were less impressed by their
improvement than the blinded raters. These drugs have not been compared head
to head either with each other or with the older remedies.
CONTINUUMJOURNAL.COM 1095
focal tardive dystonias, and deep brain stimulation, generally targeting the
globus pallidus internus segment, for uncontrolled dyskinesias and dystonia.
CONCLUSION
Tardive dyskinesia reduces quality of life and remains a major public health
issue.44 Second-generation antipsychotics have not been shown to decrease the
risk of tardive syndromes, and their increasing use for nonpsychotic indications
is likely to increase the prevalence. Our understanding of tardive syndrome
pathophysiology is not significantly better than it was decades ago. Two bright
spots have been the emergence of effective and safe symptomatic treatment for
tardive syndrome and the possibility that antipsychotic drugs that do not involve
dopamine are being developed.45 Meanwhile, there is a continued need for
vigilance in our use of dopamine receptor–blocking drugs and a need for
better treatments.
VIDEO LEGENDS
VIDEO 9-1 VIDEO 9-5
Tardive dyskinesia and stereotypy. Video Classic tardive dyskinesia. Video shows a
shows the 78-year-old man discussed in CASE 9-1 78-year-old woman with symptoms of classic
exhibiting involuntary mouth movements indicative tardive dyskinesia, involving oral, buccal, and lingual
of tardive dyskinesia and stereotypy. Exposure to a movements. She is the same patient discussed in
second-generation antipsychotic drug at usual CASE 9-3, 33 years later. She has continuous writhing
doses may result in tardive syndromes in a patient movements of her tongue as well as continuous lip
with no risk factors other than age. and jaw movements.
http://links.lww.com/CONT/A362 http://links.lww.com/CONT/A366
© 2019 American Academy of Neurology. © 2019 American Academy of Neurology.
1 Friedman JH. Historical perspective on 16 Shenker JI, Wylie SA, Fuchs K, et al. On-line
movement disorders. J Clin Psychiatry 2004; anosognosia: unawareness for chorea in real
65(suppl 9):3–8. time but not on videotape delay. Neurology
2004;63(1):159–160. doi:10.1212/01.WNL.
2 Caroff SN, Ungvari GS, Cunningham Owens DG.
0000131901.46303.65.
Historical perspectives on tardive dyskinesia.
J Neurol Sci 2018;389:4–9. doi:10.1016/j. 17 Vitale C, Pellecchia MT, Grossi D, et al.
jns.2018.02.015. Unawareness of dyskinesias in Parkinson’s and
Huntington’s diseases. Neurol Sci 2001;22(1):
3 Faurbye A, Rasch PJ, Petersen PB, et al.
105–106. doi:10.1007/s100720170066.
Neurological symptoms in pharmacotherapy of
psychoses. Acta Psychiatr Scand 1964;40(1): 18 Barnes TR. A rating scale for drug-induced
10–27. doi:10.1111/j.1600-0447.1964.tb05731.x. akathisia. Br J Psychiatry 1989;154:672–676.
doi:10.1192/bjp.154.5.672Pu.
4 Fenton WS. Prevalence of spontaneous
dyskinesia in schizophrenia. J Clin Psychiatry 19 Schooler NR, Kane JM. Research diagnoses for
2000;61(suppl 4):10–14. tardive dyskinesia. Arch Gen Psychiatry 1982;
39(4):486–487. doi:10.1001/archpsyc.1982.
5 Shah BB, Lang AE. Acquired neurosyphilis
04290040080014.
presenting as movement disorders. Mov Disord
2012;27(6):690–695. doi:10.1002/mds.24950. 20 Jeste DV, Wyatt RJ. Therapeutic strategies against
tardive dyskinesia. Two decades of experience.
6 Frei K, Truong DD, Fahn S, et al. The nosology of
Arch Gen Psychiatry 1982;39(7):803–816.
tardive syndromes. J Neurol Sci 2018;389:10–16.
doi:10.1001/archpsyc.1982.04290070037008.
doi:10.1016/j.jns.2018.02.008.
21 Zutshi D, Cloud LJ, Factor SA. Tardive syndromes
7 American Psychiatric Association. Diagnostic and
are rarely reversible after discontinuing
statistical manual. 5th ed. Washington, DC:
dopamine receptor blocking agents: experience
American Psychiatric Association, 2013.
from a university-base movement disorder clinic.
8 Marsden CD, Jenner P. The pathophysiology of Tremor Other Hyperkinet Mov (N Y) 2014;4:282.
extrapyramidal side-effects of neuroleptic doi:10.7916/D8MS3R8C.
drugs. Psychol Med 1980;10(1):55–72. doi:10.1017/
22 Quitkin F, Rifkin A, Gochfeld L, Klein DF. Tardive
S003329170003960XPu.
dyskinesia: are first signs reversible? Am J
9 D'Abreu A, Akbar U, Friedman JH. Tardive Psychiatry 1977;134(1):84–87. doi:10.1176/
dyskinesia: epidemiology. J Neurol Sci 2018;389: ajp.134.1.84.
17–20. doi:10.1016/j.jns.2018.02.007.
23 Glazer WM, Morgenstern H, Schooler N, et al.
10 Fishbain DA, Dominguez M, Goldberg M. Predictors of improvement in tardive dyskinesia
Dyskinesia associated with fluoxetine use: case following discontinuation of neuroleptic
report. Neuropsychiatry Neuropsychol Behav medication. Br J Psychiatry 1990;157:585–592.
Neurol 1992;5(2):97–100. doi:10.1192/bjp.157.4.585.
11 Aquino CC, Lang AE. Tardive dyskinesia 24 Bergman H, Rathbone J, Agarwal V, Soares-
syndromes: current concepts. Parkinsonism Weiser K. Antipsychotic reduction and/or
Relat Disord 2014;20(suppl 1):S113–S117. cessation and antipsychotics as specific
doi:10.1016/S1353-8020(13)70028-2. treatments for tardive dyskinesia. Cochrane
Database Syst Rev 2018;2:CD000459.
12 Ford B, Greene P, Fahn S. Oral and genital tardive
doi:10.1002/14651858.CD000459.pub3.
pain syndromes. Neurology 1994;44(11):2115–2119.
doi:10.1212/WNL.44.11.2115. 25 Fernandez HH, Krupp B, Friedman JH. The course
of tardive dyskinesia and parkinsonism in
13 Weiden PJ, Mann JJ, Haas GE, et al. Clinical
psychiatric inpatients: 14-year follow-up.
nonrecognition of neuroleptic-induced
Neurology 2001;56(6):805–807. doi:10.1212/
movement disorders: a cautionary study. Am J
WNL.56.6.805.
Psychiatry 1987;144(9):1148–1153. doi:10.1176/
ajp.144.9.1148. 26 Teo JT, Edwards MJ, Bhatia K. Tardive dyskinesia
is caused by maladaptive synaptic plasticity: a
14 Hansen TE, Brown WL, Weigel RM, Casey DE.
hypothesis. Mov Disord 2012;27(10):1205–1215.
Under recognition of tardive dyskinesia and
doi:10.1002/mds.25107.
drug-induced-parkinsonism by psychiatry
residents. Gen Hosp Psychiatry 1992;14(5): 27 Adler CM, Malhotra AK, Elman I, et al.
340–344. Amphetamine-induced dopamine release
and post-synaptic specific binding in
15 Correll CU, Schenk EM. Tardive dyskinesia and
patients with mild tardive dyskinesia.
new antipsychotics. Curr Opin Psychiatry 2008:
Neuropsychopharmacology 2002;26(3):295–300.
21(2);151–1516. doi:10.1097/YCO.0b013e3282f53132.
doi:10.1016/S0893-133X(01)00309-8.
CONTINUUMJOURNAL.COM 1097
28 Silvestri S, Seeman MV, Negrete JC, et al. 37 Woods SW, Morgenstern H, Saksa JR, et al.
Increased dopamine D2 receptor binding after Incidence of tardive dyskinesia with atypical and
long-term treatment with antipsychotics in conventional antipsychotic medications: a
humans: a clinical PET study. Psychopharmacology prospective cohort study. J Clin Psychiatry 2010;
(Berl) 2000;152(2):174–180. doi:10.1007/ 71(4):463–473. doi:10.4088/JCP.07m03890yel.
s002130000532.
38 Glazer WM, Morgenstern H, Doucette JT.
29 Seeman P, Tinazzi M. Loss of dopamine neuron Predicting the long-term risk of tardive dyskinesia
terminals in antipsychotic-treated schizophrenia; in outpatients maintained on neuroleptic
relation to tardive dyskinesia. Prog medications. J Clin Psychiatry 1993;54(4):133–139.
Neuropsychopharmacol Biol Psychiatry 2013;44:
39 Carbon M, Hsieh C-H, Kne JM, et al. Tardive
178–183. doi:10.1016/j.pnpbp.2013.02.011.
dyskinesia prevalence in the period of
30 Chen S, Seeman P, Liu F. Antipsychotic drug second-generation antipsychotic use: a
binding in the substantia nigra: an examination of meta-analysis. J Clin Psychiatry 2017;78(3):
high metoclopramide binding in the brains of e264–e278. doi:10.4088/JCP.16r10832.
normal, Alzheimer’s disease, Huntington’s
40 Solmi M, Pigato G, Kane JM, Correll CU. Clinical
disease, and multiple sclerosis patients, and its
risk factors for the development of tardive
relation to tardive dyskinesia. Synapse 2011;65(2):
dyskinesia. J Neurol Sci 2018;389:21–27.
119–24. doi:10.1002/syn.20825.
doi:10.1016/j.jns.2018.02.012.
31 Shuaib UA, Rajput AH, Robinson CA, Rajput A.
41 Bhidayasiri R, Jitkritsadakul O, Friedman JH, Fahn
Neuroleptic-induced parkinsonism:
S. Updating the recommendations for treatment
clinicopathological study. Mov Disord 2016;31(3):
of tardive syndromes: a systematic review of
360–365. doi:10.1002/mds.26467.
new evidence and practical treatment algorithm.
32 Christensen E, Moller JE, Faurbye A. J Neurol Sci 2018;389:67–75. doi:10.1016/j.
Neuropathological investigation of 28 brains jns.2018.02.010.
from patients with dyskinesia. Acta Psychiatr
42 Bergman H, Rathbone J, Agarwal V, Soares-
Scan 1970;46(1):14–23. doi:10.1111/j.1600-0447.1970.
Weiser K. Antipsychotic reduction and/or
tb02097.x.
cessation and antipsychotics as specific
33 Kane J, Woerner P, Weinhold P, Wegner J. Results treatments for tardive dyskinesia. Cochrane
from a prospective study of tardive dyskinesia Database Syst Rev 2018;2:CD000459.
development: preliminary findings over a 2-year doi:0.1002/14651858.CD000459.pub3.
period. Psychopharmacol Bull 1982;18:82–83.
43 Mentzel CL, Bakker PR, van Os J, et al. Effect of
34 Swartz MS, Stroup TS, McEvoy JP, et al. What antipsychotic type and dose changes on tardive
CATIE found: results from the Schizophrenia dyskinesia and parkinsonism severity in patients
Trial. Psychiatr Serv 2008;59(5):500–506. with a serious mental illness: the Curaçao
doi:10.1176/ps.2008.59.5.500. extrapyramidal syndromes study XII. J Clin
Psychiatry 2017;78(3):e279–e285. doi:10.4088/
35 Leucht S, Wahlbeck K, Hamann J, Kissling W. New
JCP.16m11049.
generation antipsychotics versus low-potency
conventional antipsychotics: a systematic review 44 Browne S, Roe M, Lane A, et al. Quality of life in
and meta-analysis. Lancet 2003;361(9369): schizophrenia: relationship to sociodemographic
1581–1589. doi:10.1016/S0140-6736(03)13306-5. factors, symptomatology and tardive dyskinesia.
Acta Psychiatr Scand 1996;94(2):118–124.
36 Correll CU, Leucht S, Kane JM. Lower risk for
doi:10.1111/j.1600-0447.1996.tb09835.x.
tardive dyskinesia associated with second-
generation antipsychotics: a systematic review 45 Cummings J, Isaacson S, Mills R, et al.
of 1-year studies. Am J Psychiatry 2004;161(3): Pimavanserin for patients with Parkinson’s
414–425. doi:10.1176/appi.ajp.161.3.414. disease psychosis: a randomised, placebo-
controlled phase 3 trial. Lancet 2014;383(9916):
533–540. doi:10.1016/S0140-6736(13)62106-6.
in Children C O N T I N U UM AUDIO
INTERVIEW AVAILABLE
ONLINE
By Toni S. Pearson, MBBS; Roser Pons, MD
ABSTRACT
PURPOSE OF REVIEW: This article provides an overview of the clinical features
and disorders associated with movement disorders in childhood. This
article discusses movement disorder phenomena and their clinical
presentation in infants and children and presents a diagnostic approach to
Downloaded from http://journals.lww.com/continuum by BhDMf5ePHKbH4TTImqenVI1NGeaZoDmOz04fr30kzVnjjmqlmokk+z4PbuF9jR/5lE+EiScBcMU= on 08/03/2019
RELATIONSHIP DISCLOSURE:
Dr Pearson has received
research/grant support from
the National Institutes of
INTRODUCTION Health/National Institute of
M
ovement disorders in childhood encompass a range of Neurological Disorders and
neurologic syndromes that are characterized by abnormalities Stroke. Dr Pons has received
personal compensation for
of tone, posture, the initiation or control of voluntary serving as a consultant for and
movements, or unwanted involuntary movements. Movement on the scientific advisory board
disorders are conventionally divided into two main categories: of Biogen, for serving as a
consultant for Guidepoint on a
hyperkinetic (involuntary movements such as dystonia, chorea, myoclonus, and rare metabolic disorder, and for
tremor) and hypokinetic (parkinsonism). In contrast to adults, children often serving as a speaker for Biogen
and for PTC Therapeutics.
present with mixed movement disorders rather than pure syndromes. Also,
symptoms may evolve over time as brain development interacts with the UNLABELED USE OF
underlying disease process so that different motor symptoms emerge at different PRODUCTS/INVESTIGATIONAL
USE DISCLOSURE:
ages along the course of a given disease. Drs Pearson and Pons report
This article describes the approach to clinical evaluation and diagnosis of no disclosures.
movement disorders that cause dystonia, chorea, ataxia, myoclonus, and
parkinsonism in infancy and childhood. This article focuses on genetic disorders © 2019 American Academy
and highlights rare, treatable disorders that present in infancy. For information of Neurology.
CONTINUUMJOURNAL.COM 1099
on tics and stereotypies, refer to the article “Tics and Tourette Syndrome” by
Harvey S. Singer, MD, FAAN,1 in this issue of Continuum.
Dystonia
Dystonia is one of the most common movement disorders in children and is
defined as abnormal often repetitive movements or postures that are caused by
sustained or intermittent involuntary muscle contractions. Numerous conditions
are associated with dystonia in children. Dystonic cerebral palsy associated with
brain injury due to complications of prematurity, stroke, or hypoxic ischemic
encephalopathy is the most common cause. Dystonia is also a feature of many
genetic disorders and may occur either in isolation or as part of a complex
neurologic syndrome. In contrast to dystonia in adults, isolated dystonia in
children is more likely to progress to generalized or multifocal dystonia than
remain focal.
Chorea
Chorea, ballism, and athetosis are hyperkinetic movement disorders that often
coexist in the same patients and are viewed as part of a continuum. Chorea is
characterized by an ongoing random-appearing sequence of one or more discrete
involuntary movements or movement fragments.2 Chorea is frequently
associated with athetosis (choreoathetosis), which is characterized by slow,
continuous, involuntary distally predominant writhing movements that prevent
maintenance of a stable posture. Ballism is defined as chorea that affects
proximal joints such as shoulder or hip joints and leads to large-amplitude
movements of the limbs. In children, acute chorea due to postinfectious/
autoimmune conditions is the most frequent cause of chorea, while dyskinetic
cerebral palsy is the most frequent cause of chronic chorea. In general, genetic
chorea is chronic, develops gradually, and tends to be generalized and
symmetric, whereas acquired chorea often manifests acutely or subacutely and,
when related to brain injury, can be asymmetric or unilateral.
Ataxia
Ataxia refers to impaired muscle control or coordination of voluntary
movements that cannot be attributed to weakness or involuntary movements
(eg, dystonia, chorea, or myoclonus).3 On examination, children with ataxia may
exhibit a variety of clinical signs, depending on the underlying pathology.
Signs include eye movement abnormalities (saccade dysmetria, nystagmus,
oculomotor apraxia), slow speech with impaired articulation (cerebellar
dysarthria), poor accuracy and coordination of voluntary reaching movements
(dysmetria, intention tremor), poor stability of head and trunk position during
sitting (titubation), and an unsteady, broad-based, or veering gait (gait ataxia).
Young children with ataxia may habitually walk quickly or run to compensate
for their balance difficulties and minimize falls.
Myoclonus
Myoclonus is defined as the “sequence of repeated, often nonrhythmic, brief
shock-like jerks due to sudden involuntary contraction or relaxation of one or
more muscles.”2 Myoclonus is associated with abnormal neuronal excitability of
cortical or subcortical gray matter; it can be classified based on distribution of
the movements (focal, multifocal, segmental, generalized) or on the etiologic
location (cortical, subcortical, spinal). While myoclonus in adults is often a
benign sign associated with metabolic disturbances, myoclonus in children is
often an ominous manifestation that can be caused by tumors, metabolic
diseases, neurodegenerative disease, or encephalitis and is often associated with
seizures and encephalopathy. Benign forms of myoclonus can occur in children
as well.2 Myoclonus can be physiologic (hypnic myoclonus) or it can be the
manifestation of a broad range of systemic disorders and metabolic
derangements, the adverse effect of multiple drugs, and the symptom of a broad
range of neurologic disorders. In pediatric patients, neurologic conditions
manifesting with myoclonus include inflammatory/autoimmune disorders (ie,
opsoclonus-myoclonus-ataxia syndrome), severe hypoxic injury, encephalitis,
and focal mass lesions or dysplasias (often manifesting as epilepsia partialis
continua).4,5
Parkinsonism
Parkinsonism is characterized by the combination of bradykinesia and one or
more of the following cardinal signs: rigidity, resting tremor, and postural
instability. Parkinsonism is rare in children, especially in infancy, and the clinical
manifestations in children differ from adults in several important ways. First,
tremor is often, but not always, absent. Second, dystonia is a common
accompanying feature; hence, childhood-onset parkinsonism is frequently
referred to as parkinsonism-dystonia.6 Third, infants with parkinsonism typically
have marked hypotonia as the primary baseline disturbance of tone.
CONTINUUMJOURNAL.COM 1101
Tremor
Tremor refers to oscillating, rhythmic movements about a fixed point, usually a
joint, producing a regular back-and-forth movement. Common causes of acute
tremor in children include drug-induced tremor and psychogenic tremor.
Essential tremor, characterized by a chronic, slowly progressive, isolated postural
and action tremor, may begin in childhood. Many secondary tremors occur in
conjunction with other movement disorders, such as dystonia and ataxia.
Parkinsonian resting tremor is uncommon in children.
Movement
Disorder Etiologies Comments
Ataxia Postinfectious cerebellar ataxia May follow infectious illness or vaccination; onset is usually very
acute, 90% recover completely over period of 2 to 3 months
Autoimmune encephalitis Rare; associated with inflammatory lesions of the basal ganglia
Tremor, dystonia, Psychogenic movement disorder Clinical clues: sudden onset of dystonia with fixed posture, episodic
gait disturbance tremor; treatment may include physical therapy and management
of comorbid mood symptoms with medication or psychotherapy
CONTINUUMJOURNAL.COM 1103
While no single etiology is specified, the term cerebral palsy usually implies an
underlying acquired etiology, such as brain lesions associated with complications
of prematurity, neonatal hypoxic ischemic encephalopathy, perinatal stroke, or
chronic sequelae of meningoencephalitis. Dyskinetic cerebral palsy is the
phenotypic classification applied to the 10% to 15% of children with cerebral
palsy whose motor syndrome is predominated by involuntary movements.
Children with dyskinetic cerebral palsy typically have a combination of dystonia
and athetosis, often accompanied by axial hypotonia with or without spasticity
in the limbs.
Dyskinetic cerebral palsy is classically associated with injury to the basal
ganglia. In practice, dystonia is a relatively common finding in children with
other cerebral palsy phenotypes, including spastic hemiplegia, diplegia, and
quadriplegia, who have injury to brain structures other than the basal ganglia.
Normal brain imaging has been reported in approximately one-third of patients
with a diagnosis of dyskinetic cerebral palsy,10 suggesting that etiologies other
than brain injury may underlie the diagnosis of cerebral palsy in a significant
proportion of children with a diagnosis of dyskinetic cerebral palsy.
CONTINUUMJOURNAL.COM 1105
Monoamine GCH, TH, PTS, Infancy, Hypotonia, dystonia, oculogyric crises, ptosis, autonomic
neurotransmitter disorders QDPR, SPR, childhood dysfunction (dopa-responsive dystonia: dystonia, spastic
DDC, DNAJC12 paraplegia with diurnal variation)
Glucose transporter type 1 SLC2A1 Infancy, Ataxia, spasticity, dystonia, paroxysmal exertional
(GLUT1) deficiency childhood dyskinesia, seizures, intellectual disability
syndrome
Cerebral folate deficiency FOLR1a Infancy, Hypotonia, developmental delay, irritability, ataxia,
childhood spasticity, chorea, dystonia, seizures
Pyruvate dehydrogenase PDHA1, DLAT, Infancy, Ataxia (may be intermittent in milder cases), hypotonia,
complex deficiency others childhood intellectual disability, seizures, paroxysmal exertional
dyskinesia (rare)
Coenzyme Q10 deficiency COQ8A, Infancy to Ataxia, may have encephalopathy, spasticity, seizures,
PDSS2, others adulthood myopathy, intellectual disability, sensorineural deafness
Ataxia with vitamin E TTPA Childhood, Ataxia, dystonia, head tremor, may have peripheral
deficiency adolescence neuropathy, decreased vibration sense
Usually normal Low CSF glucose, normal serum glucose, Ketogenic diet
low-normal CSF lactate
T2 hyperintensity in basal ganglia, High CSF lactate, abnormal urine organic Thiamine, biotin supplementation
brainstem acids profile
Ventriculomegaly, corpus callosum High lactate, pyruvate (plasma, CSF) normal Ketogenic diet, thiamine
dysgenesis, T2 hyperintensity in lactate to pyruvate ratio
basal ganglia, brainstem
Cerebral volume loss, white High serum ammonia, high lactate, may Biotin
matter T2-hyperintensity have abnormal urine organic acids profile;
low serum biotinidase activity
May have cerebellar atrophy High plasma lactate, low coenzyme Q10 Coenzyme Q10 (ubiquinone)
T2 hyperintensity in globus High GAA, low creatine (urine and plasma) Arginine restriction, creatine and ornithine
pallidus; may be normal supplements
T1 hyperintensity of basal ganglia, High whole-blood manganese Chelation with disodium calcium edetate;
sparing thalamus iron
CONTINUUMJOURNAL.COM 1107
CASE 10-1 A 6-month-old girl was brought by her parents for evaluation of
hypotonia and developmental delay. She had been born full term after an
uneventful pregnancy, delivery, and neonatal period. Her parents had
begun to be concerned at around the age of 4 months because of the lack
of motor development and the occurrence of daily episodes of tonic
upward eye deviation of several minutes duration that resolved with
sleeping. She often had nasal congestion and sweated profusely.
On examination she was alert and interactive but was easily distressed.
She showed poor facial expression and minimal spontaneous movements.
She had dystonic posturing of all limbs and dystonic tremor of the upper
limbs. She had prominent axial hypotonia. Appendicular tone was decreased
when relaxed and increased when manipulated or distressed. Tendon
reflexes were brisk, and her toes were spontaneously up (striatal toes).
Brain MRI was normal. CSF analysis of biogenic amines disclosed
decreased concentration of the dopamine metabolite homovanillic acid.
Molecular analysis of the gene encoding tyrosine hydroxylase revealed a
homozygous pathogenic mutation (c.707T>C). These findings were
consistent with tyrosine hydroxylase deficiency.
She was started on treatment with levodopa 0.5 mg/kg/d that was
followed by gradual psychomotor development. The oculogyric crises
decreased in frequency and gradually disappeared. Her hyperhidrosis and
nasal congestion resolved completely.
COMMENT This case illustrates the typical motor and autonomic symptoms of infantile
dystonia-parkinsonism associated with congenital disorders of monoamine
neurotransmitter synthesis: oculogyric crises, hypokinesia, hypotonia, and
dystonia, in conjunction with excessive sweating and nasal congestion.
CSF examination readily detected a low concentration of the dopamine
metabolite homovanillic acid, and diagnosis was confirmed with molecular
genetic testing. The patient had an excellent response to treatment with
levodopa.
CONTINUUMJOURNAL.COM 1109
CASE 10-2 A 5-year-old boy presented for evaluation following his first seizure. He
had a history of unsteady gait. He had been a healthy neonate and had
been born at term without perinatal complications. His parents had
become concerned about his motor development when he was 12 months
old. He often fell when trying to stand. He walked independently at age
16 months, but his gait always appeared unsteady. His parents noticed
that he was much stiffer and less steady in the mornings immediately
after waking, with subsequent improvement by midmorning after eating
breakfast. His balance also worsened whenever he was sick.
At 4 years of age he had several brief episodes, typically 10 to
15 minutes in duration, of involuntary stiffening and jerking movements of
both legs following periods of running or active play. At age 5 years, he
presented to the hospital following a single, brief generalized tonic-
clonic seizure.
On examination, he was observed to be alert and cooperative but
distractible. His speech was dysarthric. He had moderate lower limb
spasticity and abnormally brisk lower limb reflexes with several beats of
bilateral ankle clonus. He had mild upper limb dysmetria and a spastic-
ataxic gait pattern characterized by a narrow but variable base, toe
walking, veering, unsteadiness, and multiple falls.
His brain MRI was normal. A lumbar puncture was performed, and his
CSF glucose was 35 mg/dL (serum glucose 92 mg/dL, with a CSF to serum
ratio of 0.38). A diagnosis of glucose transporter type 1 (GLUT1) deficiency
syndrome was confirmed with the finding of a heterozygous pathogenic
missense variant in SLC2A1.
He was started on the ketogenic diet, and on follow-up 6 months
later, he had had no further seizures or episodes of involuntary leg
movements, showed marked improvement in his ataxia, and had
improved attention at school.
Epileptic-Dyskinetic Encephalopathies
A recently described group of complex neurologic disorders that may manifest
with a hyperkinetic movement disorder early in life are the epileptic-dyskinetic
encephalopathies. They are a genetically and clinically heterogeneous group of
disorders characterized by a spectrum of manifestations ranging from isolated
movement disorders (most frequently chorea, but also dystonia and
stereotypies) to severe infantile epileptic encephalopathy.13 Mutations in FOXG1
cause a developmental encephalopathy manifesting in infancy or early childhood
with severe developmental delay, acquired microcephaly, profound intellectual
disability, epilepsy, and a hyperkinetic movement disorder emerging within
the first year of life that includes various combinations of chorea,
orolingual/facial dyskinesias, dystonia, myoclonus, and hand stereotypies.
Neuroimaging may show corpus callosum hypoplasia or aplasia, delayed
myelination, simplified gyration, and frontotemporal abnormalities.14 Mutations
in GNAO1 can cause a severe infantile epileptic encephalopathy or a static
encephalopathy with associated hyperkinetic movement disorder (chorea,
ballismus, dystonia, and orofaciolingual dyskinesia) (CASE 10-3). The initial
motor phenotype is often nonspecific with hypotonia, dystonia, and motor
developmental delay. Characteristic episodes of severe chorea and ballismus in
combination with autonomic dysfunction, triggered by infections or other
stressors, can last hours or days.13 Other genes associated with epileptic-
dyskinetic encephalopathies include GRIN1, SCN8A, FRRS1L, GPR88,
UNC13A, and SYT1.15 The list is likely to expand further with the discovery of
new genes.
Dystonia
Dystonia is a feature of many childhood-onset genetic conditions, divided into
three main categories: (1) isolated (pure dystonia), (2) combined (dystonia
accompanied by myoclonus or parkinsonism), and (3) complex (dystonia as one
feature of a complex neurologic syndrome).
CONTINUUMJOURNAL.COM 1111
CASE 10-3 An 11-year-old girl presented to the emergency department with acute,
severe chorea and ballismus. She had been born at term following an
uncomplicated pregnancy and delivery. As a neonate she had hypotonia
and feeding difficulties, and at age 9 months she was diagnosed with
developmental delay when she was unable to sit independently. She
eventually sat at 18 months and walked with a walker at age 3 years.
On examination at age 3 years she had generalized hypotonia,
bradykinesia, dystonia in the upper and lower limbs, and slightly brisk
lower limb reflexes. Her brain imaging was normal, and a diagnosis of
atypical cerebral palsy was made.
At age 9 years she had an episode of mild chorea in the context of an
acute viral respiratory illness, which resolved spontaneously within a
few weeks.
At age 11, in the setting of another viral upper respiratory tract illness,
she developed severe acute chorea complicated by rhabdomyolysis.
This led to a prolonged intensive care unit admission. Whole exome
sequencing performed during this admission revealed a de novo
heterozygous pathogenic missense variant mutation in GNAO1.
Treatment with tetrabenazine 400 mg/d controlled the involuntary
movements. Six months later, bilateral globus pallidus internus deep
brain stimulation leads were placed, and over the next 18 months, the
tetrabenazine was tapered off without any recurrence of chorea. Her
motor function remained significantly impaired compared to baseline;
she could no longer sit independently or walk and was very hypokinetic.
COMMENT This case illustrates the characteristic clinical features of the predominant
motor phenotype of GNAO1 encephalopathy: a static neurodevelopmental
disorder consisting of hypotonia and developmental delay, with recurrent
episodes of acute chorea triggered by illness that can be severe and life-
threatening. As in this case, the hyperkinetic movement disorder has been
reported to be responsive to deep brain stimulation in several patients.
CONTINUUMJOURNAL.COM 1113
Ataxia
Numerous genes are associated with chronic progressive ataxia, which is usually
subdivided broadly into autosomal dominant, autosomal recessive, and
spastic-ataxia subgroups.24 The autosomal recessive ataxias are collectively the
most common in childhood and include Friedreich ataxia (the most prevalent
inherited ataxia), ataxia-telangiectasia, ataxia with oculomotor apraxia types 1
and 2, and ataxia with vitamin E deficiency.25 Several genes that are more
commonly associated with complex dystonia syndromes may also present with a
predominantly ataxic phenotype, including PLA2G6 and ATP1A3. The autosomal
◆ Delineate the clinical syndrome, including any accompanying involuntary movements, and
associated neurologic features and systemic features. If a distinctive clinical syndrome,
such as Friedreich ataxia, is apparent, consider proceeding directly to single-gene
genetic testing.
◆ Examine the brain MRI to determine whether cerebellar atrophy is present (eg, ataxia-
telangiectasia, ataxia with oculomotor apraxia types 1 and 2) or absent (eg, Friedreich ataxia,
ataxia with vitamin E deficiency) and search for associated abnormalities including cerebral
or brainstem volume loss, white matter lesions, or basal ganglia lesions.
◆ Perform selected routine laboratory investigations to screen for treatable disorders (plasma
vitamin E, coenzyme Q10), and identify abnormal biochemical markers that may assist in
the diagnosis of a nontreatable ataxia syndrome (ataxia-telangiectasia and ataxia with
oculomotor apraxia types 1 and 2), including serum α-fetoprotein, IgG subclasses, albumin,
creatine kinase, and cholesterol panel.
◆ Perform molecular genetic testing: In cases where the clinical, biochemical, and
radiologic phenotype suggests a specific syndrome, single-gene testing may be appropriate.
Increasingly, multigene next-generation sequencing panels or whole exome sequencing
are used early in the testing process. It is important to be aware that next-generation
sequencing methods will not detect triplet repeat expansions, which is an important
consideration for a number of the genetic ataxias (selected SCAs, dentatorubral-
pallidoluysian atrophy, and Friedreich ataxia).
Myoclonus
Genetic conditions manifesting with nonprogressive myoclonus include
hereditary hyperekplexia and myoclonus-dystonia, while progressive myoclonus
is characteristic of the progressive myoclonic epilepsies. Other neurodegenerative
CONTINUUMJOURNAL.COM 1115
Parkinsonism
Several degenerative conditions resemble Parkinson disease with recessive
modes of inheritance and onset of symptoms before 21 years of age.30 In these
CONTINUUMJOURNAL.COM 1117
the precise motor phenomena that occur during the episode itself. Review of the
episodes on home video remains a valuable tool to distinguish the attacks from
seizures or another episode type. The three classic paroxysmal dyskinesia
syndromes are paroxysmal kinesigenic dyskinesia, paroxysmal exertional
dyskinesia, and paroxysmal nonkinesigenic dyskinesia.
Children with paroxysmal kinesigenic dyskinesia and paroxysmal nonkinesigenic
dyskinesia typically have a normal interictal neurologic examination. Paroxysmal
kinesigenic dyskinesia episodes are usually seconds in duration, while paroxysmal
nonkinesigenic dyskinesia episodes typically last minutes to hours. Paroxysmal
kinesigenic dyskinesia episodes are often triggered by sudden movement or the
intent to move, while paroxysmal nonkinesigenic dyskinesia episodes may be
triggered by caffeine, alcohol, and stress. Both disorders are caused by monoallelic
mutations in their respective genes, PRRT2 (for paroxysmal kinesigenic
dyskinesia) and PNKD (for paroxysmal nonkinesigenic dyskinesia), and may be
either sporadic or inherited in an autosomal dominant pattern. Paroxysmal
kinesigenic dyskinesia episodes are typically well controlled with low-dose
anticonvulsant medication, most commonly carbamazepine, while paroxysmal
nonkinesigenic dyskinesia is treated with benzodiazepines.
Episodes of paroxysmal exertional dyskinesia are triggered by sustained
exercise. Disorders that cause paroxysmal exertional dyskinesia usually have other
interictal neurologic features, including intellectual disability and persistent motor
abnormalities such as ataxia or dystonia. GLUT1 deficiency syndrome, the
prototypical cause of paroxysmal exertional dyskinesia, is one example and is an
important diagnosis to recognize as it is treatable with the ketogenic diet
(discussed in the section on GLUT1 deficiency syndrome). Paroxysmal
exertional dyskinesia has also been rarely reported as a manifestation of pyruvate
dehydrogenase deficiency, which may respond to treatment with thiamine.34
Other genetic disorders in which paroxysmal dyskinesia occurs in the context
of a complex neurologic syndrome include ADCY5-related dyskinesia (as
previously discussed) and alternating hemiplegia of childhood due to mutations
in ATP1A3. In ADCY5-related dyskinesia, a distinctive feature of the paroxysmal
attacks is that they may occur at night during drowsiness.35 Infants with
ATP1A3-related alternating hemiplegia of childhood may have episodes of
paroxysmal dystonia, typically accompanied by other classic signs including
monocular nystagmus, episodic hemiplegia or quadriplegia, and persistent
neurologic deficits, including hypotonia, ataxia, dystonia, chorea, and bulbar
dysfunction.36
CONCLUSION
Movement disorders in infants and children are associated with a large number
of acquired and genetic disorders. Clinical characterization of (1) the course and
timing of neurologic symptoms in the context of the child’s development, and (2)
the movement disorder phenomena and associated neurologic and systemic
findings on examination are the key to accurate diagnosis. Disease-modifying
treatments exist for many movement disorders that present acutely and for
selected genetic, metabolic conditions that present with chronic symptoms. In
the current context of technologic advances in relation to genetic diagnosis and to
treatment, such as neuromodulation and gene therapy, the diagnosis and
treatment of childhood movement disorders is poised to evolve rapidly.
REFERENCES
1 Singer HS. Tics and Tourette syndrome. Continuum 9 Dale RC, Singh H, Troedson C, et al. A prospective
(Minneap Minn) 2019;25(4, Movement Disorders): study of acute movement disorders in children.
936–958. Dev Med Child Neurol 2010;52(8):739–748.
doi:10.1111/j.1469-8749.2009.03598.x.
2 Sanger TD, Chen D, Fehlings DL, et al. Definition
and classification of hyperkinetic movements 10 Krgeloh-Mann I, Cans C. Cerebral palsy update.
in childhood. Mov Disord 2010;25(11):1538–1549. Brain Dev 2009;31(7):537–544. doi:10.1016/j.
doi:10.1002/mds.23088. braindev.2009.03.009.
3 Sanger TD, Chen D, Delgado MR, et al. Definition 11 Kurian MA, Gissen P, Smith M, et al. The monoamine
and classification of negative motor signs in neurotransmitter disorders: an expanding range of
childhood. Pediatrics 2006;118(5):2159–2167. neurological syndromes. Lancet Neurol 2011;10(8):
doi:10.1542/peds.2005-3016. 721–733. doi:10.1016/S1474-4422(11)70141-7.
4 Sanger TD. Pathophysiology of pediatric movement 12 Hoffman GF, Blau N. Signs and symptoms of
disorders. J Child Neurol 2003;18(suppl 1):S9–S24. neurotransmitter disorders: approach to diagnosis
doi:10.1177/0883073803018001S0401. congenital neurotransmitter disorders a clinical
approach. New York, NY: Nova Science Publishers
5 Eberhardt O, Topka H. Myoclonic disorders. Brain
Inc, 2014:1–15.
Sci 2017;7(8):pii: E103. doi:10.3390/brainsci7080103.
13 Carecchio M, Mencacci NE. Emerging monogenic
6 García-Cazorla A, Ortez C, Pérez-Dueñas B, et al.
complex hyperkinetic disorders. Curr Neurol
Hypokinetic-rigid syndrome in children and
Neurosci Rep 2017;17(12):97. doi:10.1007/
inborn errors of metabolism. Eur J Paediatr Neurol
s11910-017-0806-2.
2011;15(4):295–302. doi:10.1016/j.ejpn.2011.04.013.
14 Mencacci NE, Carecchio M. Recent advances in
7 Bonnet C, Roubertie A, Doummar D, et al.
genetics of chorea. Curr Opin Neurol 2016;29(4):
Developmental and benign movement disorders
486–495. doi:10.1097/WCO.0000000000000352.
in childhood. Mov Disord 2010;25(10):1317–1334.
doi:10.1002/mds.22944. 15 de Gusmao CM, Waugh JL. Inherited and
acquired choreas. Semin Pediatr Neurol 2018;
8 Fernández-Alvarez E. Transient benign paroxysmal
25:42–53. doi:10.1016/j.spen.2018.01.002.
movement disorders in infancy. Eur J Paediatr
Neurol 2018;22(2):230–237. doi:10.1016/
j.ejpn.2018.01.003.
CONTINUUMJOURNAL.COM 1119
16 Mencacci NE, Isaias IU, Reich MM, et al. 27 Roze E, Lang AE, Vidailhet M. Myoclonus-
Parkinson's disease in GTP cyclohydrolase dystonia: classification, phenomenology,
1 mutation carriers. Brain 2014;137(pt 9): pathogenesis, and treatment. Curr Opin Neurol
2480–2492. doi:10.1093/brain/awu179. 2018;31(4):484–490. doi:10.1097/WCO.
0000000000000577.
17 Ozelius L, Lubarr N. DYT1 early-onset isolated
dystonia. In: Adam MP, Ardinger HH, Pagon RA, 28 Grabowski M, Zimprich A, Lorenz-Depiereux B,
et al, editors. Seattle, WA: GeneReviews, 1999. et al. The epsilon-sarcoglycan gene (SGCE),
mutated in myoclonus-dystonia syndrome, is
18 Meyer E, Carss KJ, Rankin J, et al. Mutations in the
maternally imprinted. Eur J Hum Genet 2003;11(2):
histone methyltransferase gene KMT2B cause
138–144. doi:10.1038/sj.ejhg.5200938.
complex early-onset dystonia. Nat Genet 2017;
49(2):223–237. doi:10.1038/ng.3740. 29 Kälviäinen R. Progressive myoclonus epilepsies.
Semin Neurol 2015;35(3):293–299. doi:10.1055/
19 Zech M, Boesch S, Maier EM, et al.
s-0035-1552620.
Haploinsufficiency of KMT2B, encoding the
lysine-specific histone methyltransferase 2B, 30 Puschmann A. Monogenic Parkinson's disease
results in early-onset generalized dystonia. Am J and parkinsonism: clinical phenotypes and
Hum Genet 2016;99(6):1377–1387. doi:10.1016/ frequencies of known mutations. Parkinsonism
j.ajhg.2016.10.010. Relat Disord 2013;19(4):407–415. doi:10.1016/
j.parkreldis.2013.01.020.
20 Gorman KM, Meyer E, Kurian MA. Review of the
phenotype of early-onset generalised progressive 31 Paviour DC, Surtees RA, Lees AJ. Diagnostic
dystonia due to mutations in KMT2B. Eur J considerations in juvenile parkinsonism.
Paediatr Neurol 2018;22(2):245–256. doi:10.1016/ Mov Disord 2004;19(2):123–135. doi:10.1002/
j.ejpn.2017.11.009. mds.10644.
21 Klein C, Lohmann K, Marras C, Munchau A. 32 Al-Rumayyan A, Klein C, Alfadhel M. Early-onset
Hereditary dystonia overview. In: Adam MP, parkinsonism: case report and review of the
Ardinger HH, Pagon RA, et al, editors. Seattle, literature. Pediatr Neurol 2017;67:102.e1–106.e1.
WA: GeneReviews, 2003. doi:10.1016/j.pediatrneurol.2015.06.005.
22 Kurian MA, Dale RC. Movement disorders 33 Mink JW. Dopa-responsive dystonia in children.
presenting in childhood. Continuum (Minneap Curr Treat Options Neurol 2003;5(4):279–282.
Minn) 2016;22(4 Movement Disorders):1159–1185. doi:10.1007/s11940-003-0033-9.
doi:10.1212/CON.0000000000000367.
34 Castiglioni C, Verrigni D, Okuma C, et al.
23 Meijer IA, Pearson TS. The twists of pediatric Pyruvate dehydrogenase deficiency presenting
dystonia: phenomenology, classification, and as isolated paroxysmal exercise induced
genetics. Semin Pediatr Neurol 2018;25:65–74. dystonia successfully reversed with thiamine
doi:10.1016/j.spen.2018.02.001. supplementation. Case report and mini-review.
Eur J Paediatr Neurol 2015;19(5):497–503.
24 Bird TD. Hereditary ataxia overview. In: Adam
doi:10.1016/j.ejpn.2015.04.008.
MP, Ardinger HH, Pagon RA, et al., editors.
Seattle, WA: GeneReviews, 1998. 35 Chang FC, Westenberger A, Dale RC, et al.
Phenotypic insights into ADCY5-associated
25 Anheim M, Tranchant C, Koenig M. The autosomal
disease. Mov Disord 2016;31(7):1033–1040.
recessive cerebellar ataxias. N Engl J Med 2012;
doi:10.1002/mds.26598.
366(7):636–646. doi:10.1056/NEJMra1006610.
36 Silveira-Moriyama L, Kovac S, Kurian MA, et al.
26 Nowak-Wegrzyn A, Crawford TO, Winkelstein JA,
Phenotypes, genotypes, and the management
et al. Immunodeficiency and infections in
of paroxysmal movement disorders. Dev Med
ataxia-telangiectasia. J Pediatr 2004;144(4):
Child Neurol 2018;60(6):559–565. doi:10.1111/
505–511. doi:10.1016/j.jpeds.2003.12.046.
dmcn.13744.
37 Jen JC, Wan J. Episodic ataxias. Handb Clin
Neurol 2018;155:205–215. doi:10.1016/B978-
0-444-64189-2.00013-5.
Movement Disorders C O N T I N U UM A U D I O
I NT E R V I E W A V A I L AB L E
ONLINE
By Mary Ann Thenganatt, MD; Joseph Jankovic, MD, FAAN
VIDEO CONTENT
A V AI L A B L E O N L I N E
ABSTRACT
PURPOSE OF REVIEW: This article reviews a practical approach to psychogenic
movement disorders to help neurologists identify and manage this
complex group of disorders.
CITE AS:
RECENT FINDINGS: Psychogenic movement disorders, also referred to as CONTINUUM (MINNEAP MINN)
Downloaded from http://journals.lww.com/continuum by BhDMf5ePHKbH4TTImqenVJsWiCZ2rfmURyvd+GhSb/tCduNFOQnx1h7K2U0aUH/H on 08/03/2019
P
sychogenic movement disorders have been described as a “crisis for Science Group, and Hodder
neurology”1 as they represent a major public health and economic Arnold. Dr Jankovic has received
research/grant support from
problem, with an estimated annual incidence of 4 to 12 cases per Allergan, CHDI Foundation,
100,000.2–4 Debate exists about the most appropriate name for this Dystonia Coalition, F.
group of disorders. The term functional movement disorders is preferred Hoffman-La Roche AG,
Huntington Study Group,
by some who argue that this term is free of stigma and does not imply a Michael J. Fox Foundation for
psychological etiology of the disorder.5 Others prefer the term psychogenic Parkinson’s Research, and the
National Institutes of Health.
movement disorders, finding the term functional vague and confusing to patients
who often see themselves as “dysfunctional” rather than “functional.”3 UNLABELED USE OF
Psychogenic movement disorders are among the most challenging movement PRODUCTS/INVESTIGATIONAL
USE DISCLOSURE:
disorders to treat as they are often difficult to diagnose, the presentation is Drs Thenganatt and Jankovic
variable in terms of phenomenology and course, the pathophysiology is poorly report no disclosures.
understood, and there is no consensus of the best therapeutic approach.6 Over
the past several decades, there has been greater awareness of this disorder with © 2019 American Academy
increased efforts into understanding and treating this common neurologic of Neurology.
CONTINUUMJOURNAL.COM 1121
DIAGNOSTIC CRITERIA
The initial and most widely recognized diagnostic criteria for psychogenic
movement disorders are the Fahn-Williams13 criteria, proposed in 1988. This set
of criteria categorizes patients into four categories: documented, clinically
established, probable, and possible. For a classification of documented
psychogenic movement disorders, persistent resolution of symptoms occurs
after psychotherapy, placebo, or suggestion or when the patient is witnessed
without the abnormal movements when observed unknowingly. The clinically
established psychogenic movement disorders category is based on inconsistency
of movements or incongruence with organic movement disorders, as well as one
additional finding of other psychogenic signs, psychological disturbances, or
multiple somatizations. The probable psychogenic movement disorders category
is based on either the presence of movements inconsistent/incongruent with
organic movement disorders or on the presence of multiple psychogenic signs or
somatizations. The possible psychogenic movement disorders category only
requires the presence of an emotional disturbance.
HISTORY
Obtaining details regarding the onset and evolution of symptoms is important as
psychogenic movement disorders usually begin suddenly and rapidly progress to
severe disability. Although the course often fluctuates, spontaneous, albeit
transient, remissions may occur. Patients often associate symptoms with a
preceding injury or illness. In one study, 80% of patients reported a physical
event prior to the onset of their symptoms.18 In a study of 43 patients with motor
conversion disorder, 56% reported a stressful life event in the month preceding
symptom onset, compared to only 21% in patients with depression and 18% in
controls.19 These events were not identified on routine assessment and needed
more in-depth evaluation.
Symptoms are often episodic, occurring for varying periods of time and then
self-resolving. Aside from paroxysmal dyskinesias, discrete episodes of
symptoms are not typical in organic movement disorders and should suggest the
possibility of a psychogenic movement disorder. These episodes in psychogenic
movement disorders are variable either in duration, phenomenology, or body
location. The movements may be limited to certain situations such as only when
seated or lying down but not interrupting walking.
In addition to the chief complaint, patients often have other transient
symptoms such as intermittent speech disturbances, weakness, gait and balance
problems, or tunnel vision.20,21 Their level of disability is often discordant with
their physical deficits; patients may use assistive devices such as canes, walkers,
or wheelchairs or be dependent on family members for total care.
Past medical history typically includes a long list of diagnoses and surgical
procedures, often with multiple somatoform symptoms. While anxiety and
depression are common in neurologic disease in general, studies have shown
more severe depression, anxiety, and a greater number of somatizations in
patients with psychogenic movement disorders compared to those with organic
disease.22 Those with psychogenic movement disorders also report higher rates of
childhood traumas (emotional, sexual, and physical) compared to those with
organic movement disorders and healthy controls.23 Family history may reveal
other family members with similar disorders or other chronic neurologic
CONTINUUMJOURNAL.COM 1123
History
◆ Sudden onset
◆ Rapid progression
◆ Preceding illness or injury
◆ Episodic symptoms
◆ Multiple somatizations
◆ Occupation in health care profession
General Examination
◆ La belle indifference
◆ Convergence spasm
◆ Giveway weakness
◆ Nonanatomic sensory signs
◆ Extreme effort and pain during testing
◆ Excessive slowness
◆ Excessive startle
Core Clinical Signs
◆ Variability of
◇ Movement frequency
◇ Direction
◇ Phenomenology
◇ Body location
◆ Distractibility
◇ Decrease or complete cessation of involuntary movements when engaged in mental
tasks or voluntary movements with an unaffected limb
◆ Entrainability
◇ Involuntary movement adopts the same frequency or a harmonic of the frequency of
voluntary repetitive movements performed with an unaffected limb
◆ Suggestibility
◇ Involuntary movement increases or decreases with the power of suggestion, such as the
application of a vibrating tuning fork
◆ Other
◇ Sudden onset
◇ Spontaneous remissions
◇ Dual-task interference
◇ Transient arrest of tremor by contralateral volitional ballistic movement
◇ Whack-a-mole sign
CONTINUUMJOURNAL.COM 1125
Psychogenic Tremor
Psychogenic tremor is the most common psychogenic movement disorder,
accounting for 50% of this group of disorders.8 Variability of tremor frequency
and direction is common, such as changing from a pronation/supination tremor
to a flexion/extension tremor about the wrist,28 as well as suggestibility,
distractibility, and entrainability (VIDEO 11-1, links.lww.com/CONT/A369). A
blinded rater evaluated videos of 33 subjects with essential tremor and 12 with
psychogenic tremor; the patients with psychogenic tremor were significantly
more likely to relay a history of sudden onset (P=.03), spontaneous remissions
(P=.03), shorter duration of tremor (P=.001), greater degree of distraction with
alternate finger tapping (P=.01) and with mental concentration on serial 7s
(P=.01), and more suggestibility with a tuning fork (P=.04) compared to those
with essential tremor.29
Variability of tremor amplitude is not specific for psychogenic tremor as
organic tremor can have variable amplitudes as well,30 often increasing with
stress or anxiety. Irregular tremor of the head and limbs is also typical of dystonic
tremor but distinguishes itself from psychogenic by features such as task
specificity, a stereotyped abnormal posture, and the presence of a null point.
Psychogenic tremor may be equally present in all states (rest, posture, kinetic),
which is not typical for organic tremors such as in Parkinson disease, where
resting tremor is typical and decreases with action. One exception is Holmes
Psychogenic Dystonia
Psychogenic dystonia is characterized by a sudden onset, rapid progression to a
fixed dystonia, and early local pain.37 This is in contrast to organic dystonia,
which is usually mobile and action induced at onset with a fixed posture
associated with pain later on in the disease course. In psychogenic dystonia,
patients usually do not have associated sensory tricks and minimal or no
exacerbation with action. The most common phenotype is plantar flexion and
inversion of the foot, and associated weakness in the dystonic limb may be seen.
On examination, active resistance to passive range of motion often occurs
(CASE 11-2).When occurring in the hands, the typical pattern is a clenched fist
with flexion of digits two through five with sparing of the thumb or the index
finger, preserving the pincer grasp. When affecting the cervical region, the
typical phenotype is a fixed laterocollis with ipsilateral shoulder elevation and
contralateral shoulder depression, and the latter is not typically seen in organic
cervical dystonia.
A mobile phenotype of psychogenic dystonia has also been described, which
has a more jerky quality.38 In one study, patients with mobile cervical dystonia
were on average 10 years older than the fixed group, which had an average age of
CONTINUUMJOURNAL.COM 1127
onset in their thirties.38 The mobile group was more likely to have dystonic
movements limited to the neck area, while the patients with fixed dystonia had
fixed postures in the limbs as well. The mobile group was also more likely to have
periods of remission with subsequent relapses.
A controversial entity referred to as peripherally induced dystonia has been
described, which is preceded by a mild injury and results in a fixed dystonia often
associated with signs of complex regional pain syndrome.39 In one review of
patients with peripherally induced movement disorders, more than one-third
had associated complex regional pain syndrome, and nearly 15% were diagnosed
with a psychogenic movement disorder. The patients were also more likely to
have fixed dystonia and were less likely to respond to treatment with botulinum
toxin injections.40 A review of reported cases of peripherally induced dystonia
found that many, but not all, patients have clinical features consistent with
psychogenic dystonia.41
Psychogenic Myoclonus
Psychogenic myoclonus often presents as jerking movements of the limbs,
head, or trunk that, unlike organic myoclonus, is often associated with facial
CONTINUUMJOURNAL.COM 1129
A370). Providing minimal support by the examiner with one finger often results in
dramatic improvement in balance.
Patients may have an exaggerated response to the pull test, with arms waving
in the air, reeling backward, or a simple collapse in the examiner’s hands
(CASE 11-3). A “fear of falling” gait, typically seen in elderly women after a fall, is
not considered a psychogenic gait disorder.47 This gait disorder is characterized
by sliding feet along the ground, as if walking on ice, holding onto walls and
furniture. Dramatic improvement occurs with suggestion and encouragement.
This gait disorder is often confused with a parkinsonian gait.48,49
One study evaluated a “chair test” in nine patients with a psychogenic gait
compared to those with an organic gait disorder.50 Eight patients with a psychogenic
gait disorder were able to propel themselves forward in a swivel chair when seated,
whereas those with an organic gait disorder had equal impairments when seated or
walking. It is important to distinguish a psychogenic gait disorder from organic
disease with a complex gait pattern. For example, a dystonic gait may be task
specific, only occurring in certain states such as walking forward, backward, or
running. Patients with organic diseases such as Huntington disease and
spinocerebellar ataxia and patients with levodopa-induced or paroxysmal
dyskinesia may have a complex, bizarre gait disorder with a combination of
choreic, dystonic, and ataxic features that may be misdiagnosed as psychogenic.51
CASE 11-3 A 54-year-old woman presented for evaluation of an abnormal gait that
had started 6 months ago. She described her gait as unsteady, and she
had started to use a walker. She had difficulty climbing up stairs but
would climb up her front porch when no one was there to help her. She
fell frequently but fortunately had had no major injuries.
Her examination showed no evidence for spasticity, neuropathy, or
parkinsonism. She had giveway weakness in her proximal leg muscles.
Upon standing, she developed a bouncing of the trunk, and her feet shook
with each step. She had difficulty with tandem gait, with her body
contorting from side to side while walking heel to toe. This improved
markedly with the assistance of the examiner’s pinky finger. On the pull
test, she reeled back, her arms flailing, and fell into the examiner’s hands.
Review of her brain MRI showed no abnormalities. On further discussion
with the patient, she was the primary caregiver for her mother with
Parkinson disease, who had passed away 6 months prior to the patient’s
symptom onset. The patient was concerned that she was developing
Parkinson disease.
CONTINUUMJOURNAL.COM 1131
PSYCHOGENIC TICS. Psychogenic tics are uncommon, with one study identifying
psychogenic tics in 4.8% of 184 patients with psychogenic movement disorders.58
Psychogenic tics can be difficult to distinguish from organic tics, which also
have sudden onset and distractibility. When compared to patients with Tourette
syndrome, those with psychogenic tics were more likely to be female and
had an older age at presentation (36.3 years versus 18.7 years of age).58 Patients
with psychogenic tics lacked a family history or a childhood history of tics. A
reemergence of childhood tics is the most common cause of tics in adulthood.
Patients with psychogenic tics lack an urge/premonitory sensation and are
unable to suppress their movements. They are less likely to report a history of
obsessive-compulsive disorder or attention deficit hyperactivity disorder,
which are common in patients with organic tics.59 A lack of a rostrocaudal
progression has been shown in psychogenic tics, as opposed to organic tics that
often start in the face and then spread. Blocking tics, which interfere with normal
movement, have been demonstrated in patients with psychogenic tics and are
rare in patients with organic tics. Patients with psychogenic tics also commonly
had manifestations of other psychogenic movement disorders or psychogenic
nonepileptic seizures.
TREATMENT
At this time, there is no consensus on the optimal treatment strategy for
psychogenic movement disorders. While a multidisciplinary approach is usually
CONTINUUMJOURNAL.COM 1133
advocated,64 access to centers that provide this support is often limited because
of the paucity of centers that specialize in psychogenic movement disorders
and frequent denials of insurance coverage. The treatment plan is coordinated
by the neurologist, who should provide continued follow-up and support to
the patient.
Psychiatrists may play an important role in the treatment rather than in the
diagnosis of patients with psychogenic movement disorders. Patients may
benefit from psychotherapy to identify underlying stressors, understand
potential psychodynamic factors, and develop coping mechanisms. Patients also
may benefit from psychotropic medications if they have underlying depression
or anxiety.65 The type of psychotherapy implemented is varied and can include
psychodynamic therapy, cognitive-behavioral therapy, and hypnosis. A
retrospective study of psychodynamic therapy for patients with psychogenic
movement disorders demonstrated that out of 30 patients, 60% showed
improvement.66 Cognitive-behavioral therapy is also used to identify cognitive
distortions and negative beliefs and modify one’s response to stressors. A pilot
study of cognitive-behavioral therapy for 21 patients with psychogenic
movement disorders demonstrated an improvement in motor symptoms and a
reduction in anxiety and depression.67 A 2016 study demonstrated the value of
qualitative interviews with open-ended conversations, rather than standardized
questionnaires.68 Out of 36 patients with psychogenic movement disorders,
28 were identified as having an additional diagnosis of a psychiatric disorder.
Such interviews provided insight to the patient’s experience of their symptoms.
Patients with psychogenic movement disorders have poor insight and
demonstrate dysfunctional emotional processing.
Physical therapy, focused on motor retraining, has been shown to be helpful in
most patients. Motor retraining focuses on regaining control over movement
by demonstrating that normal movements are possible. Distraction techniques
may be used to limit self-focus and allow for automatic movements.69 Avoiding
the use of canes or walkers helps decrease a patient’s reliance on such assistive
devices and dependency. A systematic review of physical therapy studies
involving 373 patients with psychogenic movement disorders demonstrated
improvement in 60% to 70% of patients.70
In 2015, a group of neurologists, neuropsychiatrists, and physical and
occupational therapists developed consensus recommendations for physical
therapy in psychogenic movement disorders.69 They advocated for specialized
physical therapy focused on a biopsychosocial approach. This involved
patient education about their illness, motor retraining focusing on normal
movements, and diverting attention away from their impairments. Other helpful
techniques include self-observation with mirrors and videos to change their
self-perception, using visualization techniques to focus on normal movements
while performing exercises, and keeping a journal to reinforce treatment
strategies and monitor progress.
Another possible rehabilitation strategy is using the feature of entrainment
or suppressibility in psychogenic tremor as a biofeedback tool to retrain
movements. A small study of 10 patients demonstrated the feasibility of this
approach; patients were asked to move their wrists at varying frequencies in
response to a stimulus, resulting in improvement of tremor at 6 months.71
Inpatient programs with a combination of physical therapy and
psychotherapy may have additional benefits over outpatient therapy,
Pathophysiology
Research efforts are increasingly dedicated to elucidating the underlying
mechanism of psychogenic movement disorders.6 Impaired self-agency (a sense
of control over voluntary actions) has been associated with psychogenic
movement disorders, with the temporoparietal junction identified as a key player
in modulating the neural circuit for self-agency.76 Functional MRI (fMRI) studies
have demonstrated decreased connectivity between the right temporoparietal
junction and bilateral sensorimotor regions in patients with psychogenic
movement disorders, supporting this concept.76 An impaired self-agency may
help explain why movements that are physiologically produced through
voluntary circuits are perceived by patients as involuntary.
Dysfunctional emotional processing has also been associated with
psychogenic movement disorders. Alexithymia, impaired emotional processing,
is manifested as the inability to identify and describe one’s emotion. A
cross-sectional study demonstrated that those with psychogenic motor disorders
had a higher proportion of alexithymia (34.5%) compared to patients with
organic movement disorders (9.1%) and healthy controls (5.9%).77 This suggests
that patients with psychogenic movement disorders may have difficulty
identifying physical symptoms such as tremor/weakness as stress-related
autonomic arousal.
fMRI studies have demonstrated abnormal functional connectivity in emotional
processing circuits in patients with psychogenic tremor and psychogenic
dystonia.78,79 When patients with psychogenic dystonia were presented with
CONTINUUMJOURNAL.COM 1135
emotional stimuli, decreased activation was seen in select motor and sensory
areas compared to subjects with organic dystonia or healthy controls.79
Overactivity of the limbic system has been demonstrated in patients with
psychogenic movement disorders. When presented with fearful and sad faces,
patients with psychogenic motor symptoms had increased activity of the
amygdala compared to healthy controls.80 This activity increased over time, with
repeated exposure to fearful faces in particular, in contrast to the healthy
controls that did not demonstrate this heightened arousal. Studies have also
demonstrated increased connectivity between the amygdala and motor
preparatory systems during states of emotional arousal,81 suggesting a
connection between this hyperarousal and movements.
Physiologic studies have tried to identify biological markers of stress in
patients with psychogenic movement disorders.6 A 2016 study evaluating the
autonomic nervous system found decreased resting parasympathetic activity
in psychogenic movement disorders cases versus controls.82 This lower vagal
tone may explain a vulnerability to stressful events in patients with psychogenic
movement disorders. Conversely, another study failed to demonstrate a
difference in circulating levels of cortisol (a marker of stress) between
patients with psychogenic movement disorders and healthy controls.83 Thus,
it may be the patient’s response to stress rather than the amount of stress
that is impaired in psychogenic movement disorders.
CONCLUSION
Psychogenic movement disorders is a burgeoning field of neurology with
increasing clinical reports and research focusing on underlying pathophysiology
and treatment approaches. Increasing familiarity with this group of disorders,
including physician training and public awareness, will help facilitate diagnosis
and treatment. As health care professionals become more knowledgeable and
comfortable treating this group of disorders, multidisciplinary treatment
programs will hopefully become more accessible to patients. Further
understanding of the various risk factors and underlying disease processes may
identify biomarkers that help with diagnosis and disease-course monitoring
and may lead to the development of more effective therapies.
USEFUL WEBSITES
FND HOPE FUNCTIONAL NEUROLOGICAL DISORDER SOCIETY (FNDS)
FND Hope is a patient advocacy organization for The Functional Neurological Disorder Society
those with functional neurologic symptoms. focuses on promoting education, research, and
Website content is provided by patients, caregivers, collaboration among health care professionals
physicians, and researchers. interested in functional neurologic disorders.
fndhope.org fndsociety.org
VIDEO 11-2
Psychogenic chorea and gait disorder. Video shows
a 47-year-old woman with psychogenic chorea and
gait disorder exhibiting jerky intermittent tremors
and choreiform movements in both of her hands,
which are distractible, variable, suggestible, and
entrainable. The gait shows the classic features of
astasia-abasia. The patient experienced a sudden
onset of tremors and imbalance after a revision
cervical spine surgery with a waxing and waning
course and periods of complete remission. The
episodes are triggered by stressful situations such
as getting assessed for job fitness.
links.lww.com/CONT/A370
REFERENCES
1 Hallett M. Psychogenic movement disorders: a 8 Factor SA, Podskalny GD, Molho ES. Psychogenic
crisis for neurology. Curr Neurol Neurosci Rep movement disorders: frequency, clinical profile,
2006;6(4):269–271. doi:10.1007/s11910-006-0015-x. and characteristics. J Neurol Neurosurg Psychiatry
1995;59(4):406–412. doi:10.1136/jnnp.59.4.406.
2 Fahn S, Olanow CW. “Psychogenic movement
disorders”: they are what they are. Mov Disord 9 Ferrara J, Jankovic J. Psychogenic movement
2014;29(7):853–856. doi:10.1002/mds.25899. disorders in children. Mov Disord 2008;23(13):
1875–1881. doi:10.1002/mds.22220.
3 Jankovic J. “Psychogenic” versus “functional”
movement disorders? That is the question. 10 Batla A, Stamelou M, Edwards MJ, et al. Functional
Mov Disord 2014;29(13):1697–1698. doi:10.1002/ movement disorders are not uncommon in the
mds.26040. elderly. Mov Disord 2013;28(4):540–543.
doi:10.1002/mds.25350.
4 Carson A, Lehn A. Epidemiology. Handb Clin
Neurol 2016;139:47–60. doi:10.1016/B978-0-12- 11 Carson A, Stone J, Hibberd C, et al. Disability,
801772-2.00005-9. distress and unemployment in neurology
outpatients with symptoms ‘unexplained by
5 Edwards MJ, Stone J, Lang AE. From psychogenic
organic disease’. J Neurol Neurosurg Psychiatry
movement disorder to functional movement
2011;82(7):810–813. doi:10.1136/jnnp.2010.
disorder: it's time to change the name.
220640.
Mov Disord 2014;29(7):849–852. doi:10.1002/
mds.25562. 12 Gelauff J, Stone J. Prognosis of functional
neurologic disorders. Handb Clin Neurol 2016;
6 Baizabal-Carvallo JF, Hallett M, Jankovic J.
139:523–541. doi:10.1016/B978-0-12-801772-
Pathogenesis and pathophysiology of functional
2.00043-6.
(psychogenic) movement disorders. Neurobiol
Dis 2019;127:32–34. doi:10.1016/j.nbd.2019.02.013. 13 Fahn S, Williams DT. Psychogenic dystonia.
Adv Neurol 1988;50:431–455.
7 Stone J, Carson A, Duncan R, et al. Who is
referred to neurology clinics?–the diagnoses 14 Shill H, Gerber P. Evaluation of clinical diagnostic
made in 3781 new patients. Clin Neurol Neurosurg criteria for psychogenic movement disorders.
2010;112(9):747–751. doi:10.1016/j.clineuro. Mov Disord 2006;21(8):1163–1168. doi:10.1002/
2010.05.011. mds.20921.
CONTINUUMJOURNAL.COM 1137
15 Gupta A, Lang AE. Psychogenic movement 29 Kenney C, Diamond A, Mejia N, et al. Distinguishing
disorders. Curr Opin Neurol 2009;22(4):430–436. psychogenic and essential tremor. J Neurol Sci
doi:10.1097/WCO.0b013e32832dc169. 2007;263(1–2):94–89. doi:10.1016/j.jns.2007.06.008.
16 American Psychiatric Association. Diagnostic and 30 Mostile G, Fekete R, Giuffrida JP, et al. Amplitude
statistical manual of mental disorders, 5th ed fluctuations in essential tremor. Parkinsonism
(DSM-5). Washington, DC: American Psychiatric Relat Disord 2012;18(7):859–863. doi:10.1016/j.
Association, 2013. parkreldis.2012.04.019.
17 Levenson JL, Sharpe M. The classification of 31 Park JE, Maurer CW, Hallett M. The “whack-
conversion disorder (functional neurologic a-mole” sign in functional movement disorders.
symptom disorder) in ICD and DSM. Handb Clin Mov Disord Clin Pract 2015. doi:10.1002/
Neurol 2016;139:189–192. doi:10.1016/B978-0-12- mdc3.12177.
801772-2.00016-3.
32 Kamble NL, Pal PK. Electrophysiological
18 Pareés I, Kojovic M, Pires C, et al. Physical evaluation of psychogenic movement disorders.
precipitating factors in functional movement Parkinsonism Relat Disord 2016;22(suppl 1):
disorders. J Neurol Sci 2014;338:174–177. S153–S158. doi:10.1016/j.parkreldis.2015.09.016.
doi:10.1016/j.jns.2013.12.046.
33 Apartis E. Clinical neurophysiology of
19 Nicholson TR, Aybek S, Craig T, et al. Life events psychogenic movement disorders: how to
and escape in conversion disorder. Psychol diagnose psychogenic tremor and myoclonus.
Med 2016;46(12):2617–2626. doi:10.1017/ Neurophysiol Clin 2014;44(4):417–424. doi:10.1016/
S0033291716000714. j.neucli.2013.08.014.
20 Baizabal-Carvallo JF, Jankovic J. Psychogenic 34 Kumru H, Begeman M, Tolosa E, Valls-Sole J.
ophthalmologic movement disorders. Dual task interference in psychogenic tremor.
J Neuropsychiatry Clin Neurosci 2016;28(3): Mov Disord 2007;22(14):2077–2082. doi:10.1002/
195–198. doi:10.1176/appi.neuropsych.15050104. mds.21670.
21 Baizabal-Carvallo JF, Jankovic J. Speech and 35 Kumru H, Valls-Solé J, Valldeoriola F, et al.
voice disorders in patients with psychogenic Transient arrest of psychogenic tremor induced
movement disorders. J Neurol 2015;262(11): by contralateral ballistic movements. Neurosci
2420–2424. doi:10.1007/s00415-015-7856-7. Lett 2004;370(2–3):135–139. doi:10.1016/j.
neulet.2004.08.009.
22 Defazio G, Pastore A, Pellicciari R, et al.
Personality disorders and somatization in 36 Schwingenschuh P, Saifee TA, Katschnig-Winter P,
functional and organic movement disorders. et al. Validation of “laboratory-supported” criteria
Psychiatry Res 2017;257:227–229. doi:10.1016/j. for functional (psychogenic) tremor. Mov Disord
psychres.2017.07.068. 2016;31(4):555–562. doi:10.1002/mds.26525.
23 Kranick S, Ekanayake V, Martinez V, et al. 37 Schmerler DA, Espay AJ. Functional dystonia.
Psychopathology and psychogenic movement Handb Clin Neurol 2016;139:235–245. doi:10.1016/
disorders. Mov Disord 2011;26(10):1844–1850. B978-0-12-801772-2.00020-5.
doi:10.1002/mds.23830.
38 Petrović IN, Tomić A, Vončina MM, et al.
24 Fekete R, Baizabal-Carvallo JF, Ha AD, et al. Characteristics of two distinct clinical
Convergence spasm in conversion disorders: phenotypes of functional (psychogenic) dystonia:
prevalence in psychogenic and other movement follow-up study. J Neurol 2018;265(1):82–88.
disorders compared with controls. J Neurol doi:10.1007/s00415-017-8667-9.
Neurosurg Psychiatry 2012;83(2):202–204.
39 Jankovic J. Peripherally induced movement
doi:10.1136/jnnp-2011-300733. doi:10.1176/appi.
disorders. Neurol Clin 2009;27(3):821–32, vii.
neuropsych.15050104.
doi:10.1016/j.ncl.2009.04.005.
25 Espay AJ, Aybek S, Carson A, et al. Current
40 van Rooijen DE, Geraedts EJ, Marinus J, et al.
concepts in diagnosis and treatment of
Peripheral trauma and movement disorders: a
functional neurological disorders. JAMA Neurol
systematic review of reported cases. J Neurol
2018;75(9):1132–1141. doi:10.1001/jamaneurol.
Neurosurg Psychiatry 2011;82(8):892–898.
2018.1264.
doi:10.1136/jnnp.2010.232504.
26 Thenganatt MA, Jankovic J. Psychogenic
41 Ganos C, Edwards MJ, Bhatia KP. Posttraumatic
movement disorders. Neurol Clin 2015;33(1):
functional movement disorders. Handb Clin
205–224. doi:10.1016/j.ncl.2014.09.013.
Neurol 2016;139:499–507. doi:10.1016/B978-0-12-
27 Baizabal-Carvallo JF, Jankovic J. Examiner 801772-2.00041-2.
manoeuvres 'sensory tricks' in functional
42 Erro R, Bhatia KP, Edwards MJ, et al. Clinical
(psychogenic) movement disorders. J Neurol
diagnosis of propriospinal myoclonus is unreliable:
Neurosurg Psychiatry 2017;88(5):453–455.
an electrophysiologic study. Mov Disord 2013;
doi:10.1136/jnnp-2016-315120.
28(13):1868–1873. doi:10.1002/mds.25627.
28 Thenganatt MA, Jankovic J. Psychogenic tremor:
43 van der Salm SM, Erro R, Cordivari C, et al.
a video guide to its distinguishing features.
Propriospinal myoclonus: clinical reappraisal and
Tremor Other Hyperkinet Mov (N Y) 2014;4:253.
review of literature. Neurology 2014;83(20):
doi:10.7916/D8FJ2F0Q.
1862–1870. doi:10.1212/WNL.0000000000000982.
CONTINUUMJOURNAL.COM 1139
72 Williams DT, Lafaver K, Carson A, Fahn S. 78 Espay AJ, Maloney T, Vannest J, et al. Impaired
Inpatient treatment for functional neurologic emotion processing in functional (psychogenic)
disorders. Handb Clin Neurol 2016;139:631–641. tremor: a functional magnetic resonance imaging
doi:10.1016/B978-0-12-801772-2.00051-5. study. Neuroimage Clin 2017;17:179–187.
doi:10.1016/j.nicl.2017.10.020.
73 Jacob AE, Kaelin DL, Roach AR, et al. Motor
retraining (MoRe) for functional movement 79 Espay AJ, Maloney T, Vannest J, et al. Dysfunction
disorders: outcomes from a 1-week in emotion processing underlies functional
multidisciplinary rehabilitation program. PM R (psychogenic) dystonia. Mov Disord 2018;33(1):
2018;10(11):1164–1172. doi:10.1016/j.pmrj.2018.05.011. 136–145. doi:10.1002/mds.27217.
74 Ferrara J, Stamey W, Strutt AM, et al. 80 Aybek S, Nicholson TR, O'Daly O, et al.
Transcutaneous electrical stimulation (TENS) Emotion–motion interactions in conversion
for psychogenic movement disorders. disorder: an FMRI study. PLoS One 2015;10(4):
J Neuropsychiatry Clin Neurosci 2011;23(2): e0123273. doi:10.1371/journal.pone.0123273.
141–148. doi:10.1176/appi.neuropsych.23.2.141.
81 Voon V, Brezing C, Gallea C, et al. Emotional stimuli
75 Pollak TA, Nicholson TR, Edwards MJ, David AS. and motor conversion disorder. Brain 2010;133(pt 5):
A systematic review of transcranial magnetic 1526–1536. doi:10.1093/brain/awq054.
stimulation in the treatment of functional
82 Maurer CW, Liu VD, LaFaver K, et al. Impaired
(conversion) neurological symptoms. J Neurol
resting vagal tone in patients with functional
Neurosurg Psychiatry 2014;22(suppl 1):S153–S158.
movement disorders. Parkinsonism Relat Disord
doi:10.1136/jnnp-2012-304181.
2016;30:18–22. doi:10.1016/j.parkreldis.2016.06.009.
76 Maurer CW, LaFaver K, Ameli R, et al. Impaired
83 Maurer CW, LaFaver K, Ameli R, et al. A biological
self-agency in functional movement disorders:
measure of stress levels in patients with
a resting-state fMRI study. Neurology 2016;87(6):
functional movement disorders. Parkinsonism
564–570. doi:10.1212/WNL.0000000000002940.
Relat Disord 2015;21(9):1072–1075. doi:10.1016/j.
77 Demartini B, Petrochilos P, Ricciardi L, et al. The parkreldis.2015.06.017.
role of alexithymia in the development of
functional motor symptoms (conversion
disorder). J Neurol Neurosurg Psychiatry 2014;
85(10):1132–1137. doi:10.1136/jnnp-2013-307203.
Warn a Patient C O N T I N U UM A U D I O
I NT E R V I E W A V A I L AB L E
ONLINE
By Joseph S. Kass, MD, JD, FAAN; Rachel V. Rose, JD, MBA
ABSTRACT
This medicolegal article examines a physician’s liability when he or she has
knowledge of adverse effects associated with a prescription medication
and suggests ways to mitigate that liability risk. The article also discusses
the circumstances under which pharmaceutical companies face liability
Downloaded from http://journals.lww.com/continuum by BhDMf5ePHKbH4TTImqenVChdHhRBUo+1QvvE9Pg71eVHGqr1Dx2I54OCdtggYgCWOEsfYTEOFrQ= on 08/03/2019
CASE
Note: This is a hypothetical case.
A 38-year-old woman presented to discuss her migraine treatment with
a new neurologist after moving to a new city. The patient had a history of
chronic migraines, took topiramate daily for migraine prophylaxis, and
used metoclopramide and sumatriptan to abort her acute migraine attacks.
CITE AS:
The patient asked the neurologist to prescribe this same effective
CONTINUUM (MINNEAP MINN)
treatment regimen. 2019;25(4, MOVEMENT DISORDERS):
The neurologist was concerned about this patient's risk of developing 1141–1144.
T
his article examines a physician’s liability in relation to the following Dr Kass serves as associate
editor of medicolegal issues for
issues presented in this case: Under what circumstances have physicians Continuum, as an associate
faced liability for causing metoclopramide-associated tardive dyskinesia, editor for Continuum Audio, as a
neurology section editor of
and under what circumstances have pharmaceutical companies been Ferri’s Clinical Advisor for
found liable for causing tardive dyskinesia? The article then provides Elsevier, and as co-editor of
suggestions to mitigate the risk of liability when a physician has knowledge of Neurology Secrets, Sixth Edition.
Dr Kass has received personal
adverse effects associated with a prescription medication. compensation for CME lectures
from Pri-Med LLC. Ms Rose
Malpractice Basics serves on the editorial board of
BC Advantage and receives book
The most common legal underpinning for lawsuits against physicians and royalties from the American Bar
pharmaceutical companies is an allegation of negligence. In legal terms, Association.
negligence is defined as the following:
UNLABELED USE OF
PRODUCTS/INVESTIGATIONAL
The omission to do something which a reasonable [person], guided by USE DISCLOSURE:
those considerations which ordinarily regulate the conduct of human affairs, Dr Kass and Ms Rose report no
disclosures.
would do[,] or doing something which a prudent and reasonable [person]
would not do. It must be determined in all cases by reference to the situation © 2019 American Academy
and knowledge of the parties and all the attendant circumstances.1 of Neurology.
CONTINUUMJOURNAL.COM 1141
The February 2009 US Food and Drug Administration (FDA) boxed warning
about metoclopramide-induced tardive dyskinesia resulted in an uptick in
malpractice cases filed related to this adverse effect.5 These cases related directly
to physicians’ failure to disclose the risk of tardive dyskinesia, obtain informed
consent, and record the informed consent discussion in the patient chart.
The duty to obtain informed consent resides not with the pharmaceutical
company but rather with the provider. Although physicians who fail to obtain
adequate informed consent are vulnerable to a malpractice lawsuit,
pharmaceutical companies have a duty to disclose potential medication
adverse effects as soon as these adverse effects come to light. Although the nature
of the duty of care is different for the physician and pharmaceutical company,
both parties can be found liable on different theories of negligence
(malpractice due to inadequate informed consent for the physician and failure
CASE CONTINUED
The neurologist explained to the patient why she was reluctant to
prescribe metoclopramide, describing the risks of tardive dyskinesia,
extrapyramidal side effects, and neuroleptic malignant syndrome, and
warned her of the types of symptoms that should prompt the patient to
seek immediate medical attention. Because the patient found that this
medication, when taken in combination with sumatriptan, was the only
drug regimen that aborted severe migraines, the patient was willing to
accept these risks. The neurologist, however, was judicious in her
prescribing of metoclopramide, limiting the prescription to severe migraine
attacks only. She also documented the informed consent discussion in the
medical record.
CONCLUSION
While both physicians and pharmaceutical companies may be sued for failing to
disclose known side effects, physicians can take specific steps to mitigate their
risk of a successful lawsuit against them. First, physicians must inform their
patients about a therapy’s common side effects as well as its rare but serious
side effects, such as tardive dyskinesia. The physician should document the
informed consent discussion in the medical record, which ideally includes a
review of the risks of the medication, the rationale for prescribing the medicine,
the patient’s understanding and acceptance of the risks of treatment, and
symptoms that should prompt the patient to seek urgent medical evaluation.
Second, if a physician is an early adopter of a drug (ie, using a therapy
before a robust literature exists to support its use), the physician should
disclose this situation to the patient and include an explanation of why the
therapy is medically appropriate despite lack of strong evidence to support
its use. Of course, this discussion should also be documented in the
medical record.
CONTINUUMJOURNAL.COM 1143
REFERENCES
1 Black’s Law Dictionary. The law dictionary. What 4 Informed consent: code of medical ethics
is negligence? thelawdictionary.org/ opinion 2.1.1. American Medical Association.
negligence/. Accessed June 11, 2019. ama-assn.org/delivering-care/ethics/informed-
consent. Accessed June 11, 2019.
2 Rodriguez-Escobar v Goss, 392 S.W.3d 109, 113
(Tex 2013). 5 Ehrenpreis ED, Krishnan A, Alexoff A, et al. A
survey of lawsuits filed for the complaint of
3 Reglan (metoclopramide) tablets: highlights of
tardive dyskinesia following treatment with
prescribing information. United States Food and
metoclopramide. Clin Pharmacol Biopharm 2014;
Drug Administration. accessdata.fda.gov/
4(1). doi:10.4172/2167-065X.100013.1.
drugsatfda_docs/label/2017/017854s062lbl.pdf.
Accessed June 11, 2019. 6 Joint and several liability. Cornell Law School.
law.cornell.edu/wex/joint_and_several_liability.
Accessed June 11, 2019.
CONTINUUMJOURNAL.COM 1175
Movement Disorders
Article 1: Parkinson Disease
Theresa A. Zesiewicz, MD, FAAN. Continuum (Minneap Minn). 2019; 25 (4 Movement
Disorders):896–918.
ABSTRACT
PURPOSE OF REVIEW:
Parkinson disease is a common neurodegenerative disorder that affects millions of people
worldwide. Important advances in the treatment, etiology, and the pathogenesis of Parkinson
disease have been made in the past 50 years. This article provides a review of thecurrent
understanding of Parkinson disease, including the epidemiology, phenomenology, and
treatment options.
RECENT FINDINGS:
Parkinson disease is nowrecognized to be a heterogeneouscondition marked by both motor and
nonmotor symptoms. It is composed of preclinical, prodromal, and clinical phases. New
medications with improved ease of administration have been approved for its treatment.
Innovative surgical therapies for Parkinson disease may be used when motor symptoms persist
despite optimal medical management.
SUMMARY:
Parkinson disease is a complex, heterogeneous neurodegenerativedisorder. Considerable
progress has been made in its treatment modalities, both pharmacologic and surgical. While its
cure remains elusive, exciting new research advances are on the horizon.
KEY POINTS
• A renaissance of therapeutic options for Parkinson disease have occurred in the last 50 years. Levodopa
remains the gold standard for treatment of Parkinson disease, but dopamine agonists, monoamine oxidase
type B inhibitors, catechol-O-methyltransferase inhibitors, and surgical procedures have greatly expanded
the therapeuticoptions.
• Parkinson disease affects millions of people worldwide, and its prevalence increasesgreatly with advancing
age.
• Clinical features of Parkinson disease include tremor, rigidity, akinesia (or bradykinesia), and
posturalinstability. Nonmotor symptoms are commonly experienced by patients and often negatively impact
quality of life. Premotor symptoms include constipation, anosmia, rapid eye movement sleep disorder,
and depression.
• The diagnosis of Parkinson disease is made clinically. Red flags for atypical parkinsonism include severe
dysautonomia, early-onset hallucinations and dementia, freezing, postural instability, and lack of response
ABSTRACT
PURPOSE OF REVIEW:
Patients who have parkinsonian features, especially without tremor, that are not responsive to
levodopa, usually have one of these three major neurodegenerative disorders rather than
Parkinson disease: progressive supranuclear palsy (PSP), multiple system atrophy (MSA), or
corticobasal degeneration (CBD). Each of these disorders eventually develops signs and
symptoms that distinguish it from idiopathic Parkinson disease, but these may not be present at
disease onset. Although these conditions are not generally treatable, it is still important to
correctly diagnose the condition as soon as possible.
RECENT FINDINGS:
In recent years, it has been increasingly recognized that thesymptoms of these diseases do not
accurately predict the pathology, and the pathology does not accurately predict the clinical
syndrome. Despite this, interest has grown in treating these diseases by targetingmisfolded tau
(in thecase of PSP and CBD) and misfolded α-synuclein (in the case of MSA).
KEY POINTS
• Patients with parkinsonian features who do not improve with levodopa usually do not have idiopathic
Parkinsondisease and often have either progressive supranuclear palsy, multiple system atrophy, or
corticobasal degeneration.
• Progressive supranuclear palsy is a likely diagnosis in patients with parkinsonian features and
earlydevelopment of a supranuclear palsy.
• Some patients with progressive supranuclear palsy do not develop a supranuclear palsy until later in the
course of the disease. Early features that suggest progressive supranuclear palsy are an angry or puzzled
look, growling speech, early development of dysphagia, and a broad-based gait with abducted arms.
• A minority of patients with the pathology of progressive supranuclear palsy may have signs and symptoms
suggesting a variety of conditions, including corticobasal degeneration and, rarely, idiopathic Parkinson
disease, primary progressive aphasia, cerebellar ataxia, frontotemporal dementia, and primary lateral
sclerosis.
• Other pathologies may produce signs and symptoms suggestive of progressive supranuclear palsy, including
Alzheimer disease, some frontotemporal dementias, Whipple disease, Niemann-Pick disease type C, and
Gaucher disease.
• The pathology of progressive supranuclear palsy is characterized by deposits of 4-repeat tau in astrocytes
and oligodendroglia in multiple regions of the basal ganglia and cortex of the brain.
• Preliminary studies of agents that interfere with the formation or spread of misfolded tau are being
conducted with the hope of stopping or slowing the progression of progressive supranuclear palsy.
• The clinical hallmarks of classic corticobasal degeneration are parkinsonism combined with unilateral
dystonia, myoclonus, and cortical deficits such as apraxia, cortical sensory loss, and alien limb phenomenon.
• As in progressive supranuclear palsy, the pathology of corticobasal degeneration also involves widespread
deposition of 4-repeat tau but also includes asymmetric cortical atrophy and neuronal, oligodendroglial, and
astrocytic deposits distinct from the deposits in progressive supranuclear palsy.
• As with progressive supranuclear palsy, multiple pathologies may mimic the signs and symptoms of
corticobasal degeneration, including progressive supranuclear palsy, Alzheimer disease, Pick disease, and
Creutzfeldt-Jakob disease. When this happens, it is known as corticobasal syndrome. Similarly, the
pathology of corticobasal degeneration may present as progressive supranuclear palsy, primary nonfluent
aphasia, Alzheimer disease, and other conditions.
• Middle-aged patients presenting with parkinsonism, autonomic insufficiency, and ataxia usually have
multiple system atrophy. However, many patients with multiple system atrophy may initially only have
symptoms in one or two of these categories, making the correct diagnosis more difficult. The development of
laryngeal stridor is a strong clue that the diagnosis is multiple system atrophy.
• Unlike progressive supranuclear palsy and corticobasal degeneration, multiple system atrophy is a
synucleinopathy, not a tauopathy. This may have implications for future treatments.
• Like progressive supranuclear palsy and corticobasal degeneration, there are widespread pathologic
abnormalities in multiple systematrophy, but the characteristic inclusions contain α-synuclein, not tau. The
first identified abnormality was a glial cytoplasmic inclusion containing α-synuclein, but neuronal inclusions
havealso been identified. Rarely, Lewy bodies are found in multiple system atrophy.
• There has been an intensive search for genetic risk factors for these conditions. Some candidate genes have
been identified, but this has not currently led to any therapeutic innovations. Some genes have been
identified thatoccasionally produce one of these syndromes, but those, so far, have been responsible for
only a small percentage of known cases.
ABSTRACT
PURPOSE OF REVIEW:
The purpose of this article is to present current information on the phenomenology,
epidemiology, comorbidities, and pathophysiology of tic disorders and discuss therapy options.
It is hoped that a greater understanding of each of these components will provide physicians and
caregivers with the necessary information to deliverthoughtful and optimal care to affected
individuals.
RECENT FINDINGS:
Recent advances include the finding that Tourette syndrome is likely due to a combination of
several different genes, both low-effect and larger-effect variants, plus environmental factors.
Pathophysiologically, increasing evidence supports involvement of the cortical–basal
ganglia–thalamocortical circuit; however, the primary location and neurotransmitter remain
controversial. Behavioral therapy is first-line treatment, and pharmacotherapy is based on tic
severity. Several newer therapeutic agents are under investigation (eg, valbenazine,
deutetrabenazine, cannabinoids), and deep brain stimulation is apromising therapy.
SUMMARY:
Tics, defined as sudden, rapid, recurrent, nonrhythmic motormovements or vocalizations, are
essential components of Tourette syndrome. Although some tics may be mild, others can cause
significant psychosocial, physical, and functional difficulties that affect daily activities. In
addition to tics, most affected individuals have coexisting neuropsychological difficulties
(attention deficit hyperactivity disorder, obsessive compulsive disorder, anxiety, mood
disorder, disruptive behaviors, schizotypal traits, suicidal behavior, personality disorder,
antisocial activities, and sleep disorders) that can further impact social and academic activities
or employment.
KEY POINTS
• Tics have several characteristics that are useful in identifying their presence, including precipitating factors,
a waxing and waning pattern, admixture of new and old tics, a premonitory urge thatresolveswhen the tic
is done, reduction when engrossed, and variable severity.
• Tics can be highly variable and fluctuate, and an individual’s tic repertoire evolves over time.
• The diagnosis of a tic disorder is based on historical features and observation of the tics; no
definitivediagnostic laboratory test has yet been established.
• Simple tics are relatively common in childhood, with reports of prevalence (the number of cases in
thepopulation at a given time) being 6% to 12% (range of 4% to 24%).
Article 4: Tremor
Elan D. Louis, MD, MS, FAAN. Continuum (Minneap Minn). August 2019; 25 (4 Movement
Disorders):959–975.
ABSTRACT
PURPOSE OF REVIEW:
Tremor may be defined as an involuntary movement that is rhythmic (ie, regularly recurrent)
and oscillatory (ie, rotating around a central plane) and may manifest in a variety of ways;
accordingly, tremor has a rich clinical phenomenology. Consequently, the diagnosis of tremor
disorders can be challenging, and misdiagnoses are common. The goal of this article is to provide
the reader with straightforward approaches to the diagnosis and treatment of tremors.
RECENT FINDINGS:
Focused ultrasound thalamotomy of the ventral intermediate nucleus of the thalamus is an
emerging and promising therapy for the treatment of essential tremor.
SUMMARY:
The evaluation should start with a detailed tremor history followed by a focused neurologic
examination, which should attend to the many subtleties of tremor phenomenology. Among
KEY POINTS
• Tremors are involuntary movements that are both rhythmic and oscillatory.
• An initial step in evaluating patients with tremor is to determine whether the tremor is primarily present at
rest or with activity.
• The key feature of essential tremor is kinetic tremor.
• The kinetic tremor of essential tremor is typically slightly asymmetric.
• Approximately one-half of patients with essential tremor exhibit intention tremor during the
fingernose-finger maneuver.
• The postural tremor in essential tremor is generally out of phase; this can create a seesaw effect when
thepatients’ arms are held in the wing-beat position.
• Resting tremor may occur in patients with severe or long-standing essential tremor, but it is restricted to the arms.
• Neck tremor is several times more common in women with essential tremor than in men with essential tremor.
• Neck tremor is always pathologic. It is not a feature of enhanced physiologic tremor.
• Although limb tremor may be present, head tremor should not be a feature of drug-induced action tremor.
• The tremor in dystonia may be neither rhythmic nor oscillatory.
• Dystonic head tremor often persists after the patient lies on his or her back; this is generally not true of
essential tremor.
• Primary writing tremor is a tremor that occurs mainly while writing but not during other tasks that involve
the hands.
• In some cases, orthostatic tremor may be heard when a stethoscope is placed over the affected leg; the
tremormakes a sound like a distant helicopter.
• The clinical phenomenology of tremor of cerebellar origin is heterogeneous, and it extends beyond that
ofintention tremor to include postural tremor, kinetic tremor, resting tremor, and orthostatic tremor.
• Rubral tremor is strikingly asymmetric, and it has resting, postural, and kinetic components.
• Psychogenic tremors often have an abrupt onset.
• Wing-beat tremor is considered a classic tremor in Wilson disease, but it is not the most common type of
tremor in that disease.
• Although intention tremor is common in patients with fragile X tremor-ataxia syndrome, kinetic, postural, and
resting tremorsmay also occur.
• The resting tremor in Parkinson disease is generally asymmetric.
• In contrast to essential tremor, the jaw tremor of Parkinson disease is more often noted when the patient’s
mouth is closed and relaxed rather than while the patient is speaking.
ABSTRACT
PURPOSE OF REVIEW:
This article provides a summary of the state of the art in the diagnosis, classification, etiologies,
and treatment of dystonia.
RECENT FINDINGS:
Although many different clinicalmanifestations of dystoniahave been recognized for decades, it
is only in the past 5 years that a broadly accepted approach has emerged for classifying them
into specific subgroups. The new classification system aids clinical recognition and diagnosis by
focusing on key clinical features that help distinguish the many subtypes. In the past few years,
major advances have been made inthe discovery of new genes as well advances in our
understanding of the biological processes involved. These advances have led to major changes
in strategies for diagnosis of the inherited dystonias. An emerging trend is to move away from
heavy reliance on the phenotype to target diagnostic testing toward a broader approach that
involves large gene panels or whole exome sequencing.
SUMMARY:
The dystonias are a large family of phenotypically andetiologically diverse disorders. The
diagnosis of these disorders depends on clinical recognition of characteristic clinical features.
Symptomatic treatments are useful for all forms of dystonia and include oral medications,
botulinum toxins, and surgical procedures. Determination of etiology is becoming increasingly
important because the number ofdisorders is growing and more specific and sometimes
disease-modifying therapies now exist.
KEY POINTS
• Dystonic movements are not always slow; they can be rapid or jerky, or resemble tremor.
• Dystonic movements tend to be patterned, not random.
• Dystonic movements are often triggered or worsened by voluntary muscle activity.
• Identification of a geste antagoniste (sensory trick) can be a very helpful clue because it is unique to dystonia
and is important to ask patients about.
• The history and examination of patients with dystonia should focus on four areas: body region affected, age
at onset, temporal features, and ancillary neurologic problems.
• For the most common focal dystonias that emerge after 40 years of age, laboratory investigations are usually
not needed.
• For any dystonia that emerges in a child or young adult, laboratory investigations are guided by the history
and examination.
• For almost all classic inherited dystonic disorders in children, late-onset cases or less severe cases are
known to occur in adults.
• Elucidating etiology is important because specific treatments are available for several types of dystonia.
• Isolated dystonia may be the initial manifestation for neurologic disorders typically associated with more
complex syndromes.
• More than 100 known causes for dystonia exist.
• Genetic forms of dystonia should be referred to by the name of the gene, not the DYT locus name.
Article 6: Chorea
Pichet Termsarasab, MD. Continuum (Minneap Minn). August 2019; 25 (4 Movement
Disorders):1001–1035.
ABSTRACT
PURPOSE OF REVIEW:
This article provides an overview of the approach to chorea in clinical practice, beginning with a
discussion of the phenomenologic features of chorea and how to differentiate it from other
movement disorders. The diagnostic approach, clinical features of important acquired and
genetic choreas, and therapeutic principles are also discussed. Practical clinical points and
caveats are included.
RECENT FINDINGS:
C9orf72 disease is the most common Huntington disease phenocopy, according to studies in the
European population. Anti-IgLON5 disease can present with chorea. The role of
immunotherapies in Sydenhamchorea has increased, and further clinical studiesmay be useful.
Benign hereditary chorea is a syndrome or phenotype due to mutations in several genes,
including NKX2-1, ADCY5, GNAO1, PDE10A. New-generation presynaptic dopamine-depleting
agents provide more options for symptomatic treatment of chorea with fewer adverse effects.
Deep brain stimulation has been performed in several choreic disorders, but features other than
chorea and the neurodegenerative nature should be taken intoconsideration. Studies on genetic
interventions for Huntington disease are ongoing.
SUMMARY:
Clinical features remain crucial in guiding the differentialdiagnosis and appropriate
investigations in chorea. Given the complexity of most choreic disorders, treating only the
chorea is not sufficient. A comprehensive and multidisciplinary approach is required.
KEY POINTS
• Randomness is the key phenomenologic feature of chorea.
• Chorea with quick velocities may look jerky, resembling myoclonic jerks.
ABSTRACT
PURPOSE OF REVIEW:
This article reviews the symptoms, laboratory and neuroimaging diagnostic tests, genetics, and
management of cerebellarataxia.
RECENT FINDINGS:
Recent advances in genetics have led to the identificationof novel genetic causes for ataxia and
a more comprehensive understanding of the biological pathways critical for normal cerebellar
function.When these molecular pathways become dysfunctional, patients develop cerebellar
ataxia. In addition, several ongoing clinical trials for Friedreich ataxia and spinocerebellar ataxia
will likely result in novel symptomatic and disease-modifying therapies for ataxia. Antisense
oligonucleotides for spinocerebellar ataxias associated with CAG repeat expansions might be a
promising therapeutic strategy.
SUMMARY:
Cerebellar ataxias include heterogeneous disorders affecting cerebellar function, leading to
ataxic symptoms. Step-by-step diagnosticworkups with genetic investigations are likely to
reveal the underlying causes of ataxia. Some disease-specific therapies for ataxia exist, such as
vitamin E for ataxia with vitamin E deficiency and thiamine for Wernicke encephalopathy,
highlighting the importance of recognizing these forms of ataxia. Finally, genetic diagnosis for
patients with ataxia will accelerate clinical trials for disease-modifying therapy and will have
prognostic value and implications for family planning for these patients.
KEY POINTS
• Determining the etiology of cerebellar ataxia iscomplex; however, step-by-step approaches can streamline
the diagnostic workflow.
• Key questions regarding difficulty running, trouble walking in high heels or barefoot on the beach, andveering
toward one side can be helpful in identifying the subtle gait abnormality associated with ataxia.
• Patients with ataxia can have a variety of eye movement abnormalities, including nystagmus, hypermetric or
hypometric saccades, and ophthalmoplegia.
• The gait abnormality associated with cerebellar ataxia can change over the course of the disease.
• After establishing the signs of cerebellar ataxia, look for other neurologic signs (eg, tremor, dystonia,
parkinsonism, motor neuron signs) as clues to the cause of ataxia.
• Laboratory evaluation can be helpful in identifying nutritional and immunologic causes of cerebellar ataxia.
• Aside from cerebellar atrophy, specific changes on MRI associated with different forms of cerebellar ataxia
can provide important diagnostic clues.
Article 8: Myoclonus
John N. Caviness, MD, FAAN. Continuum (Minneap Minn). August 2019; 25 (4
Movement Disorders):1055–1080.
ABSTRACT
PURPOSE OF REVIEW:
This article offers clinicians a strategic approach for making sense of a symptom complex that
contains myoclonus. The article presents an evaluation strategy that highly leverages the two
major classification schemes of myoclonus. The goal of this article is to link evaluation strategy
with diagnosis and treatment of myoclonus.
RECENT FINDINGS:
The growth of medical literature has helped better define myoclonus etiologies. Physiologic
study of myoclonus types and etiologies with electrophysiologic testing has provided greater
clarity to the pathophysiology of the myoclonus in various diseases. Although studies have been
limited, the role of newer treatment agents and methods has made progress.
SUMMARY:
Myoclonus has hundreds of different etiologies. Classification is necessary to evaluate
myoclonus efficiently and pragmatically. The classification of myoclonus etiology, which is
grouped by different clinical presentations, helps determine the etiology and treatment of the
myoclonus. The classification of myoclonus physiology using electrophysiologic test results
helps determine the pathophysiology of the myoclonus and can be used to strategize
symptomatic treatment approaches. Both basic ancillary testing (including EEG and imaging) and
more comprehensive testing may be necessary. Treatment of the underlying etiology is the ideal
approach. However, if such treatment is not possible or is delayed, symptomatic treatment
guided by the myoclonus physiology should be considered. More controlled study of myoclonus
treatment is needed. Further research on myoclonus generation mechanisms should shed light
on future treatment possibilities.
KEY POINTS
• The brief, lightninglike muscle contraction defines it as myoclonus.
• Myoclonus is a symptom or sign, not a diagnosis. It occurs in multiple diseases and conditions.
• Evaluation for myoclonus begins with a comprehensive history and neurologic examination that allows the
clinical presentation classification into a physiologic, essential, epileptic, or symptomatic category.
• EEG should be the initial electrophysiologic testing for myoclonus without a determined etiology.
ABSTRACT
PURPOSE OF REVIEW:
This article reviews the history, nosology, clinical features, epidemiology, and treatment of
tardive syndromes.
RECENT FINDINGS:
The major advance in the field of tardive syndromes has been the development and US Food and
Drug Administration (FDA) approval of two vesicular monoamine transporter type 2 inhibitors,
valbenazine and deutetrabenazine, for treating tardive syndromes. These medications are
derivatives of tetrabenazine and reduce dyskinetic movements by reducing dopamine
stimulation. Treatment is not curative, and the medications reduce, or “mask,” symptoms but
presumablywithout adding to the long-term risk of increased involuntary movements believed to
KEY POINTS
• The relationship between tardive dyskinesia and antipsychotics took several years to establish and was
initially thought to be rare.
• All definitions of tardive dyskinesia or tardive syndromes require at least several weeks exposure to a drug
preceding the development of a new movement disorder that persists for several weeks while on the drug, or
off the drug, and is not better explained by an alternative etiology.
• There are several different tardive syndromes. Dyskinesia and stereotypies are very similar, while akathisia
and dystonia are very different. The others are rare. Patients may have more than one syndrome. It is
important to note that patients often have more than one tardive syndrome.
• While widely believed to represent dopamine supersensitivity, the pathophysiology of tardive syndromes
remains unknown, and no explanation explains the variety of tardive syndromes.
• Tardive syndromes remain a major problem for patients treated with dopamine receptor–blocking drugs.
While there are data to suggest that second-generation antipsychotics are less likely to cause a tardive
syndrome than first-generation antipsychotics, these data are not convincing, and the largest study
performed to answer this question did not find a difference.
• Deutetrabenazine and valbenazine are approved treatments for tardive syndromes, and probably work best
for nondystonic disorders. Replacing the neuroleptic with clozapine at a dose to treat the psychosis may be
very helpful, especially for dystonic syndromes.
• Botulinumtoxin is likely to be helpful for all focal dystonias, including tardive dystonias. Deep brain
stimulation, with globus pallidus interna as the target, may be helpful for dystonic or choreoathetoid tardive
disorders.
ABSTRACT
PURPOSE OF REVIEW:
This article provides an overview of the clinical features and disorders associated with
movement disorders in childhood. This article discusses movement disorder phenomena and
their clinical presentation in infants and children and presents a diagnostic approach to
suspected genetic disorders with a focus on treatable conditions.
KEY POINTS
• Many causes of childhood ataxia exist that may be broadly divided into acute, intermittent, and chronic
categories.
• Myoclonus can be physiologic (hypnic myoclonus), or it can be the manifestation of a broad range of systemic
disorders and metabolic derangements.
• Parkinsonism in children differs from parkinsonism in adults, often manifesting as bradykinesia/hypokinesia,
dystonia, and axial hypotonia; tremor is often absent.
• Neonates, infants, and toddlers may manifest with a number of benign and transient movement disorders
such as myoclonus, dystonia, or tremor; development is normal, and treatment is not required.
• The most common etiologies underlying acute movement disorders in a previously healthy child are
autoimmune, drug-induced, and psychogenic.
• In a child with a dyskinetic cerebral palsy phenotype, absent risk factors for perinatal brain injury, and normal
brain MRI, investigation for an underlying genetic disorder should be considered. Some genetic disorders
have disease-specific treatment that improves symptoms and developmental outcome.
• The primary monoamine neurotransmitter disorders comprise defects of enzymes, cofactors, and
transporters involved in the metabolism and homeostasis of the catecholamines and serotonin.
• In biogenic amine disorders, neuroimaging is usually normal, and diagnosis is confirmed with the analysis of
monoamine neurotransmitter metabolites and pterins in CSF and with molecular analysis.
• The epileptic-dyskinetic encephalopathies are a heterogeneous group of disorders that are associated with a
spectrum of movement disorders, most frequently chorea, but also dystonia and stereotypies.
• Huntington disease in childhood often presents with an akinetic-rigid syndrome rather than chorea.
• Myoclonus-dystonia is a rare genetic movement disorder characterized by a combination of nonepileptic
myoclonic jerks and dystonia.
• Myoclonus-dystonia is compatible with an active and normal life span; however, some patients have a
progressive course leading to considerable disability. Treatment is usually disappointing.
• Progressive myoclonic epilepsy is characterized by action myoclonus, epileptic seizures, and progressive
neurologic decline. The majority of genes involved in progressive myoclonic epilepsy encode lysosomal
proteins and are inherited in an autosomal recessive pattern. The largest group of progressive myoclonic
epilepsies are the neuronal ceroid lipofuscinosis.
• Juvenile parkinsonism refers to hereditary conditions with onset before the age of 21 years that clinically
resemble Parkinson disease but with different histopathologic characteristics.
• In juvenile parkinsonism disease, progression is slower than in idiopathic Parkinson disease. Patients have a
marked response to levodopa, although dyskinesias and motor fluctuations occur early.
• The classic genetic paroxysmal dyskinesias may be clinically distinguished from one another by the episode
triggers, episode duration, and the presence or absence of interictal neurologic features.
ABSTRACT
PURPOSE OF REVIEW:
This article reviews a practical approach to psychogenic movement disorders to help
neurologists identify and manage this complex group of disorders.
RECENT FINDINGS:
Psychogenic movement disorders, also referred to as functional movement disorders, describe
a group of disorders that includes tremor, dystonia, myoclonus, parkinsonism, speech and
gait disturbances, and other movement disorders that are incongruent with patterns of
pathophysiologic (organic) disease. The diagnosis is based on positive clinical features that
include variability, inconsistency, suggestibility, distractibility, suppressibility, and other
supporting information. While psychogenic movement disorders are often associated with
psychological and physical stressors, the underlying pathophysiology is not fully understood.
Although insight-oriented behavioral and pharmacologic therapies are helpful, a multidisciplinary
approach led by a neurologist, but also including psychiatrists and physical, occupational, and
speech therapists, is needed for optimal outcomes.
SUMMARY:
The diagnosis of psychogenic movement disorders is based on clinical features identified on
neurologic examination, and neurophysiologic and imaging studies can provide supporting
information.
KEY POINTS
• Early diagnosis of a psychogenic movement disorder is important as longer duration of symptoms is
associated with poor outcome.
• Diagnosis of a psychogenic movement disorder is based on positive signs and symptoms and isnot a diagnosis
of exclusion.
• Psychogenic movement disorders are typically sudden in onset and rapidly progress to severe disability.
• In patients with psychogenic tremor, variability of tremor frequency and direction is common, as well as
suggestibility, distractibility, and entrainability.
• Total body tremor is a typical manifestation of psychogenic tremor, including a bobbing of the head and
trunk.
• A rapid-onset fixed dystonia is the typical phenotype of psychogenic dystonia.
• A clinical diagnosis of propriospinal myoclonus is unreliable, and more than 50% of cases are psychogenic.
• Psychogenic gait often presents as slowness and buckling at the knees, dramatic compensatory measures,
and improvement with minimal support.
• Psychogenic parkinsonism often coexists with organic parkinsonism.
• Psychogenic facial spasms are typically tonic contractions of the lower face with ipsilateral platysma
contraction and downward deviation of the lip.
• Psychogenic tics typically lack a premonitory urge, suppressibility, and often lack a family or childhood
history of tics.