You are on page 1of 11

REVIEW

Stiff person syndrome


Shahrzad Hadavi,1 Alastair J Noyce,1 R David Leslie,2 Gavin Giovannoni1

1
Department of Neuroscience Abstract agents can also attenuate the aberrant
and Trauma, Blizard Institute
of Cell and Molecular Science,
Stiff person syndrome (SPS) is a rare disorder, immune process.
Barts and The London School of characterised by fluctuating rigidity and
stiffness of the axial and proximal lower
Background
Medicine and Dentistry, Queen
Mary University of London, limb muscles, with superimposed painful Moersch and Woltman described ‘stiff
London, UK
2
spasms and continuous motor unit activity man syndrome’ in 1956 in 14 patients
Centre for Diabetes, Blizard
Institute of Cell and Molecular on electromyography. Although rare in with tightness of the back, abdominal
Science, Barts and The London general neurology practice, once observed it and thigh muscles.2 Their observa-
School of Medicine and is unforgettable. The general neurologist may tions spanned a 32-year period and
Dentistry, Queen Mary University
of London, London, UK
see only one or two cases during his or her they detailed progressive fluctuating
career and as such it remains underdiagnosed. rigidity and painful spasms, leading to
Correspondence to Left untreated, SPS symptoms can progress
Dr Shahrzad Hadavi, Department a characteristic ‘wooden man’ appear-
to cause significant disability. Patients have
of Neuroscience and Trauma, ance, with postural instability and
Blizard Institute of Cell and a poor quality of life and an excess rate of
Molecular Science, Barts and
falls. Diabetes mellitus (DM) accom-
comorbidity and mortality. The severity of
The London School of Medicine
symptoms and lack of public awareness of
panied a handful of cases.
and Dentistry, Queen Mary Eleven years later, Gordon et al set
University of London, 4 Newark the condition create anxiety and uncertainty
Street, London E1 2AT, UK; for people with the disease. This review aims out clinical criteria to address the need
shahrzadhadavi@doctors.org.uk to raise awareness of SPS and to improve the for improved diagnostic certainty.3
Accepted 3 August 2011 likelihood of its earlier diagnosis and treatment. These were modified by Lorish et al in
19894 and by Dalakas in 2009.1 These
Introduction latest criteria are currently in use.
Stiff person syndrome (SPS) is rare, Moersch and Woltman described the
although its true frequency has not electromyographic findings of motor
been fully ascertained. The British unit activity resembling ‘that which
Neurological Surveillance Unit iden- accompanies contraction of volun-
tified 119 cases among the UK pop- tary muscle’.2 Gordon and colleagues
ulation over 5 years (2000–2005), (1967) used electromyography and
implying a prevalence of 1–2 cases per muscle relaxants, describing ‘persist-
million. ent tonic contraction reflected in con-
The natural history of SPS has stant firing, even at rest’.3
yet to be completely described. The In 1988, Solimena et al described
symptoms range from mild to severe the major breakthrough in the patho-
and can progress, resulting in sig- genesis of SPS in a patient presenting
nificant disability. Perhaps 65% of with diabetic ketoacidosis. Through
patients cannot independently per- elegant experiments, they identified
form normal activities of daily living an autoimmune link between SPS and
due to rigidity and stiffness, phobias, DM. They proposed that autoantibod-
unpredictable spasms and frequent ies against GAD, an enzyme found in
falls.1 both the central nervous system and in
Almost all SPS has an autoimmune pancreatic islets of Langerhans, were
basis; most have the glutamic acid important to both diseases.5 The GAD
decarboxylase (GAD) autoantibody enzyme was already known to syn-
but there are also paraneoplastic thesise the inhibitory neurotransmit-
varieties. Main line therapies include ter, GABA; their findings provided a
γ-aminobutyric acid (GABA)-ergic and plausible disease mechanism. Of inter-
other antispasmodic agents to alleviate est, Howard had reported previously
symptoms, but immunomodulatory that diazepam significantly reduced

272 Practical Neurology 2011;11:272–282. doi:10.1136/practneurol-2011-000071


REVIEW

Figure 1 The timeline of significant dates in the history of SPS.2 5–12 GABARAP, γ-aminobutyric acid (A) receptor-associated protein; GAD, glutamic acid
decarboxylase; SPS, stiff person syndrome

muscle stiffness and rigidity in SPS, suggesting GAD autoantibodies


involvement of GABA-related inhibitory path- About 60–80% of SPS cases have autoantibodies
ways (figure 1).6 against GAD, a rate-limiting cytoplasmic enzyme
responsible for synthesising the inhibitory neuro-
Pathophysiology transmitter GABA in the brain and spinal cord.1 16
The exact pathogenic role of autoantibodies in GAD is synthesised mainly in presynaptic GABA-
SPS remains unclear. All SPS autoantigens iden- ergic neurons in both the central nervous sys-
tified to date are synaptic proteins involved in tem and in the islet of Langerhans β-cells of the
inhibitory synaptic transmission. The presyn- pancreas (figure 2).16 The mechanism by which
aptic autoantigens are GAD and amphiphysin, autoantibodies recognise intracellular GAD is
and the postsynaptic autoantigens are GABA (A) unclear.
receptor-associated protein and gephyrin (fig- There are two GAD isoforms—GAD65 and
ure 2).5 10–12 GAD67—but it is GAD65 that is the main target
The SPS autoantibodies probably mediate their for GAD autoantibodies in both SPS and DM.17
effect through pharmacological blockade of their In SPS, GAD autoantibodies recognise both lin-
target autoantigens rather than by causing struc- ear and conformational epitopes, whereas in
tural changes in GABA-ergic neurons. This is type 1 DM they recognise only conformational
supported by the lack of abnormal neurological epitopes.18 An epitope is the specific region of an
signs other than increased muscle tone, and the antigen recognised by the immune system. A lin-
improvement of symptoms with immunotherapy.13 ear epitope has a continuous sequence of amino
The fact that most cases have normal postmortem acids and it is this primary structure that the
findings supports this theory. Some cases show autoantibody recognises. In contrast, a confor-
non-specific neuronal and interneuronal loss in the mational epitope has a three-dimensional shape
spinal cord.14 with discontinuous sequence of amino acids and
Interestingly, a report of stiff horse syndrome interacts with autoantibodies based on this terti-
described a stiff gait and painful muscles to palpa- ary structure.
tion. The clinical examination, electromyographic The GAD autoantibodies in type 1 DM recognise
and serological investigations were strikingly sim- the carboxy-terminal end or the centre of the GAD
ilar to SPS.15 molecule, while in SPS, the GAD autoantibodies

Practical Neurology 2011;11:272–282. doi:10.1136/practneurol-2011-000071 273


REVIEW

Figure 2 A summary diagram of the γ-aminobutyric acid (GABA) metabolic pathway and the molecular biology of GABA neurotransmission.5 10–12 19
(A) GABA biochemistry: glutamate is converted to GABA via two isoforms of glutamic acid decarboxylase (GAD65 and GAD67). GABA is then packaged into
presynaptic vesicles to allow exocytosis into the GABA synapse. Excess GABA is metabolised to succinate, which then enters the tricarboxylic acid cycle.
(B) The GABA synapse: GABA accumulates in the presynaptic vesicle. Fusion of vesicles with the presynaptic membrane enables exocytosis of GABA into the
synapse. A protein complex is formed to allow fusion and is composed of synaptosomal-associated protein 25 (SNAP-25), synaptobrevin and syntaxin. The
component proteins in this complex are anchored by SNAP receptor (SNARE) proteins; v-SNARE is vesicle-associated and t-SNARE is ‘target’ or presynaptic
membrane-associated. Synaptotagmin is involved in docking of the vesicle with the presynaptic membrane and calcium-mediated fusion. Synaptophysin
is a ubiquitous synaptic vesicle glycoprotein. Amphiphysin is involved in endocytosis and cycling of synaptic vesicle membrane, following GABA exocytosis.
Once GABA has entered the synapse, it binds to GABA receptors. Gephyrin is thought to be involved with clustering of GABA receptors at the postsynaptic
membrane, as is the GABA (A) receptor-associated protein (GABARAP). Autoantibodies against GAD, amphiphysin, gephyrin and GABARAP have all been
identified in patients diagnosed with stiff person syndrome.

mostly recognise the amino-terminal fragment of Other associated autoantibodies


GAD.17 The differential recognition of the GAD See table 1 and figure 2.
molecule is reflected in T cell immunoreactivity Clinical features
to it: T cells from type 1 DM patients recognise The symptom onset is typically insidious. Patients
epitopes of GAD at the carboxy-terminal while may report intermittent aching and tightness in
those from SPS recognise epitopes of GAD at the the neck, paraspinal and abdominal muscles. The
amino-terminal.17 GAD autoantibodies in type 1 rigidity spreads slowly through the proximal mus-
DM tend to be restricted to the immunoglobulin cles and is often asymmetrical.1 Over time, activi-
G1 (IgG1) isotype, but in SPS the isotype profile ties of daily living become severely impaired and
is much broader, including IgG4 and IgM; these patients report difficulty dressing, walking and
differences may either be disease-related or may bending forward, for example, when putting on
reflect the much higher GAD titre in SPS.17 GAD shoes.
autoantibody titres in SPS are up to 50 times ele- The age of onset varies considerably but is
vated but only 10 times in DM,1 although not all usually in adulthood, at an average age of 41.2
SPS patients have high GAD autoantibody titres.17 years (range 29–59 years).20 SPS affects twice as
Of note, the GAD autoantibody titre in serum or many women as it does men,20 in line with other
cerebrospinal fluid (CSF) does not correlate with adult-onset autoimmune diseases: this observa-
symptom severity,1 and so titre monitoring is tion led to a nomenclature change from stiff man
unnecessary. syndrome to SPS. Several clinical features given
GAD autoantibodies are not specific to SPS and below are consistent mainly with GAD autoanti-
DM, and occur also in cerebellar ataxia, myo- body positive cases.
clonus, epilepsy and several other neurological
disorders (see Differential Diagnosis section). Rigidity and stiffness
This spectrum of clinical entities results from dif- Rigidity and stiffness of the trunk muscles are
ferences in epitope recognition.1 the earliest symptoms and result from constant

274 Practical Neurology 2011;11:272–282. doi:10.1136/practneurol-2011-000071


REVIEW

Table 1 Other autoantibodies associated with stiff person syndrome (SPS)10–12


Gephyrin Present in a very small proportion of the paraneoplastic variant. It is a postsynaptic cellular protein responsible for clustering of two
inhibitory neurotransmitters: (1) glycine receptors in the spinal cord and (2) GABA-A receptors in the brain.
Amphiphysin Present in 5% of the paraneoplastic variant. It is a synaptic vesicle protein responsible for endocytosis of the vesicle membrane after
exocytosis of GABA from the axonal terminals.
GABA (A) receptor- Present in 70% of SPS patients. It is a postsynaptic protein at GABA-ergic synapses, responsible for the stability and surface expression
associated protein of GABA-A receptor and organises the clustering of GABA (A)-receptor by linking them to gephyrin.
GABA, γ-aminobutyric acid.

Figure 3 Postural abnormalities and examination findings in stiff person syndrome. (A) and (B) Hyperlordosis. (C) Coexistent contraction of abdominal
muscles. (D) Note the skin creases in the lumber region of the back, hinting at exaggerated lordosis.

contraction of both lumbar and abdominal mus- Importantly, lumbar hyperlordosis persists even
cles (figure 3). The abdominal and lumbar par- when the patient lies flat on their back, but usu-
aspinal rigidity begins insidiously and fluctuates ally alleviates in sleep.21
at first. As it progresses, patients develop a fixed The rigidity progresses slowly from the trunk
posture, giving the characteristic lumbar spine to the proximal lower limb muscles and can cause
‘hyperlordosis’, a diagnostic hallmark (figure 3). walking difficulty. The gait may be bizarre, but

Practical Neurology 2011;11:272–282. doi:10.1136/practneurol-2011-000071 275


REVIEW

BOX 1 WHAT TO LOOK FOR DURING


EXAMINATION
■ Increased tone in the axial/truncal muscle groups

■ Increased tone in the legs (symmetric or

asymmetric)
■ Normal power in the upper and lower limbs,

unless at an advanced stage of the disease


■ Possible hyper-reflexia, without plantar
extension
■ Normal sensory function and coordination

■ Hyperlordosis of the lumbar spine (resulting

from cocontraction of abdominal and paraspinal


muscles)
■ ‘Woody’ feel on palpation of the muscles, due

to spasms
■ Slow, wide and cautious gait

■ Intact cognitive function

■ Normal sphincter function

typically is slow and wide, in an effort to improve


balance.1 Patients with severe and untreated SPS
ultimately become bedridden through progres-
sion of the stiffness.
Facial muscle involvement is rare, but can give
an ‘emotionless mask’ appearance.13 If the stiff-
ness affects the thoracic muscles, there may be Figure 4 Possible additional ophthalmic signs. (A) Primary position: left
restriction of chest expansion and breathing dif- eye deviated down and out. (B) and (C) Rightward and leftward gaze: left
ficulty13; this can be catastrophic if treatment is eye cannot fully adduct. (D) Upward gaze: left eye fails to fully elevate.
withdrawn abruptly. The hands or feet may be Cranial nerve palsies can be observed very occasionally in stiff person
syndrome (SPS) and are more common in the SPS plus variant, progressive
involved in up to 25% of cases22; typically, these encephalitis with rigidity (see Differential Diagnosis section). However, this
cases are GAD autoantibody-negative. The arms, patient has Graves’ ophthalmopathy with coexistent autoimmune thyroid
if involved, may have a flexed posture.3 disease.

Superimposed spasms
as task-specific phobias, depression and gener-
Muscle spasms, superimposed on muscle rigidity,
alised anxiety disorders. Major psychological
are initially intermittent and precipitated by star-
features can dominate the clinical picture and
tle (particularly sudden auditory or tactile stimu-
lead to the misdiagnosis of a psychogenic move-
lation), by psychological factors and by passive
ment disorder.13 However, SPS patients do not
or active movement of the affected or unaffected
typically have premorbid phobias or anxiety and
muscles. Spasms can be extremely painful and
often have realistic and appropriate fears of cer-
disabling. They are usually short lasting (minutes)
tain situations because of their stiffness, spasms
and disappear gradually on removal of the trig-
and falls.23 Anxiety-provoking examples include
gering stimulus. Spasms may also occur in bouts,
crossing roads, initiating walking without sup-
resembling tetanus,21 or may lead to a ‘shock-like’
port or descending stairs without a banister.20 The
clinical presentation, with sweating, tachycardia
anxiety encountered in SPS therefore results more
and restlessness.3 Falls from severe spasms are
from the primary neurological disorder than from
very common, sometimes even with long bone
a primary phobic disorder, although the reduc-
fractures and joint dislocations.3 The fear of fall-
tion in GABA levels may predispose then to an
ing may prompt patients to use mobility aids
exaggerated anxiety response.23 These associated
(canes or wheelchairs), even when the severity of
features can complicate the diagnosis and lead to
rigidity does not make this necessary.
suspicions of malingering in undiagnosed cases,
Psychological features often reinforced by the response to diazepam
Clinicians treating patients with SPS must be aware and the pain relief with morphine. As with other
of frequent coexisting psychiatric symptoms such chronic disorders, depression commonly coexists

276 Practical Neurology 2011;11:272–282. doi:10.1136/practneurol-2011-000071


REVIEW

disabling rigidity and stiffness, task-specific or non-


BOX 2 THE DALAKAS CRITERIA FOR THE
specific phobias, the unpredictability of dangerous
DIAGNOSIS OF TYPICAL STIFF PERSON
SYNDROME1 muscle spasms, or frequent falls.1 Even SPS patients
■ Stiffness in the axial muscles, prominently in the receiving treatment can have markedly reduced
abdominal and thoracolumbar paraspinal muscle quality of life, owing to their problems in social
leading to a fixed deformity (hyperlordosis) and physical functioning.24 Stiffness severity can
■ Superimposed painful spasms precipitated by have a major effect on quality of life: for example,
unexpected noises, emotional stress, tactile severe cervical rigidity may limit head rotation, and
stimuli cause problems when driving.4 Coexisting depres-
■ Confirmation of the continuous motor unit
sion can further diminish quality of life.
activity in agonist and antagonist muscles by
electromyography Diagnosis and diagnostic criteria
■ Absence of neurological or cognitive impairments SPS is largely a clinical diagnosis, facilitated by
that could explain the stiffness a high degree of suspicion; there are no specific
■ Positive serology for GAD65 (or neurological signs or laboratory tests.20 The vari-
amphiphysin) autoantibodies, assessed by ability in clinical presentation and recognition of
immunocytochemistry, western blot or SPS variants further increase the diagnostic uncer-
radioimmunoassay tainty.27 The delay in diagnosis ranges from 1 to
■ Response to diazepam*
18 years, with a mean of 6.2 years.20
Not part of Dalakas’ criteria, but commonly included in the The Dalakas criteria1 20 (see box 2) are used
diagnostic criteria. worldwide to diagnose SPS. Patients not meeting
*GAD, glutamic acid decarboxylase.
these criteria (eg, without the classical axial dis-
tribution of stiffness and rigidity) are described as
in SPS patients and should be sought and treated.24 atypical.
The psychological/psychiatric symptoms may be Autoimmune and other associations
managed in partnership with a psychiatrist once SPS is strongly associated with other autoim-
the SPS diagnosis is secure. mune diseases: about 35% of SPS patients also
Pain have type 1 DM.1 Importantly, most patients with
Pain is common in SPS. The first symptom may adult-onset type 1 DM do not require treatment
be a persistent, progressively worsening ache, with insulin, whether they have SPS or not.28
localised to the area of rigidity. The pain is typi- About 5–10% of patients also have autoim-
cally chronic but worsens acutely with muscle mune thyroid disease, Graves’ disease, perni-
spasms. The clinician must therefore specifically cious anaemia or vitiligo.16 Ten per cent of GAD
ask about pain since it significantly impacts on autoantibody-positive SPS patient have epilepsy,
quality of life. possibly from functional impairment of GABA-
ergic neurons,16 and a further 10% have ataxia.1
Examination findings
The head retraction reflex occurs in many SPS Investigations
patients (see box 1).25 This is a non-specific abnor- Details of routine and additional investigations
mal cutaneo-muscular brainstem reflex, elicited are listed in table 2.
by tapping the nasal ridge, upper lip, glabella or Differential diagnosis
chin, provoking a backward jerk of the head or The differential diagnosis of SPS is summarised
truncal retropulsion. in box 3.
Eye movement disturbances may occur in SPS Patients are often initially suspected of having
but are not sufficiently frequent to be a diagnostic a more common neurological, medical or psychi-
guide. These present as gaze palsies suggestive of atric disorder. They may be referred to numerous
third, fourth or sixth nerve palsies, supranuclear specialists before SPS is diagnosed, for example to
gaze palsies or nystagmus.26 An eye movement orthopaedic surgeons or rheumatologists for back
abnormality should prompt consideration of one pain and spinal stiffness, or psychiatrists for pho-
of the ‘SPS plus’ variants or additional pathology bias and anxiety.
(figure 4). SPS variants
Social implications Barker et al divided SPS into three subcategories:
Up to 65% of SPS patients cannot independently the SPS, the stiff limb syndrome and progressive
undertake normal activities of daily living,1 due to encephalomyelitis with rigidity.27

Practical Neurology 2011;11:272–282. doi:10.1136/practneurol-2011-000071 277


REVIEW

Table 2
Tests Essential Occasionally needed
Blood tests Full blood count, electrolytes, liver function test, Protein electrophoresis, serum B12, folate, HTLV-1, treponemal
thyroid function tests, fasting glucose, creatine kinase, agglutination, rheumatoid factor
erythrocyte sedimentation rate, C reactive protein
Oral glucose tolerance test If the patient does not have DM, a glucose tolerance test, together with a
C-peptide curve, can usefully assess pancreatic β-cell reserve in incipient
DM.
Serology Anti-GAD In GAD-negative patients, antigephyrin and anti-amphiphysin (part of
the screen for paraneoplastic SPS). Anti-GABARAP and anti-Ri might be
considered in patient with atypical presentations
Antiparietal cell, anti-tissue transglutaminase, anti-intrinsic factor,
antithyroid microsomal autoantibodies, antinuclear, extractable nucleic
antigens (to identify coexisting autoimmune disease)
Cerebrospinal fluid Cell count (normal), protein (normal), glucose (normal),
locally synthesised oligoclonal IgG bands (positive)
Imaging MRI scans of brain and spinal cord (pathology here Chest x-ray, spinal x-ray, CT scan of chest/abdomen/pelvis, colonoscopy/
can present with muscle rigidity) gastroscopy, mammography/breast ultrasound scan, thyroid ultrasound
scan/nuclear imaging, lymph node biopsy, whole body FDG-PET scan (to
identify a possible primary tumour)
Electromyography Continuous muscle fibre activity (pathognomic of SPS)
Genetics HLA-DRB1 and HLA-DQB1 genes (associated with SPS). Differential
diagnosis includes hereditary spastic paraparesis (eg, SPG11) and primary
dystonia (DYT1),
Histopathology Muscle biopsy (usually normal)
Postmortem: often normal: inconsistent and non-specific findings such as
neuronal loss in the spinal cord.
DM, diabetes mellitus; FDG-PET, fluorodeoxyglucose positron emission tomography; GABARAP, γ-aminobutyric acid (A) receptor-associated protein;
GAD, glutamic acid decarboxylase; HTLV-1, human T cell lymphotropic virus type 1; IgG, immunoglobulin G; SPS, stiff person syndrome.

Stiff limb syndrome in classical SPS. However, the symptoms improve


There is a focal onset, usually in one lower limb, dramatically with methylprednisolone, suggesting
with subsequently more widespread symptoms. that the underlying mechanism is autoimmune
The stiffness usually remains most prominent in mediated inflammation.30 31
the limb where symptoms began.1 27 Up to half
of these patients have sphincter disturbance and Paraneoplastic variants
about a third develop brainstem involvement.27 Paraneoplastic SPS, comprising 5% of patients,
Electrophysiological findings are similar to those manifests as stiffness mostly in the neck and arms,
in classical SPS. Most patients are GAD autoan- in contrast to the distribution of typical SPS.1
tibody-negative and only partially respond to Paraneoplastic SPS is associated with malignan-
GABA-ergic treatment.27 cies of the breast, colon, lung, thymus and in
Hodgkin’s lymphoma, occasionally manifesting
Progressive encephalitis with rigidity
before the cancer does. Autoantibodies against
Progressive encephalomyelitis with rigidity can
amphiphysin and gephyrin may occur: this situ-
present in patients already suspected of having
ation should prompt thorough investigation (see
SPS. It presents as a complex progressive illness
table 2) and ongoing vigilance.
dominated by an acute onset of painful rigidity
Less common SPS variants include: persistent
and muscle spasms in the limbs and trunk. It has
focal stiff man or stiff leg syndrome, a cerebellar
a rapid course,29 with brainstem dysfunction (nys-
subtype with truncal ataxia, gait ataxia, dysarthria
tagmus, opsoclonus, ophthalmoparesis, deafness,
and abnormal eye movements, and the ‘jerking
dysarthria, dysphagia) and profound autonomic
stiff man syndrome’.
disturbance.30
The CSF shows a mild lymphocytic pleocytosis Treatment of SPS
with elevated protein and oligoclonal IgG bands. SPS treatments are aimed at symptom relief and/
MRI may show increased signal intensity through- or modulation of the underlying aberrant immune
out the spinal cord and the brainstem.31 The process. The rarity of SPS makes it difficult to
response to diazepam is mild compared with that recruit sufficient patient numbers for good quality

278 Practical Neurology 2011;11:272–282. doi:10.1136/practneurol-2011-000071


REVIEW

relief, sometimes with troublesome side effects.


BOX 3 DIFFERENTIAL DIAGNOSIS
■ Clinical differential
Some patients require and can tolerate very large
■ Myelopathy: compressive, ischaemic,
doses, but uptitration must be undertaken gradu-
haemorrhagic and inflammatory (including ally. In general, the trend is for addition or sub-
multiple sclerosis and infectious causes) stitution with other therapeutic agents to avoid
■ Myopathy: channelopathies, inflammatory, unwanted effects.
myotonic dystrophy, paramyotonia Baclofen
■ Neuropathic: neuromyotonia,
Baclofen is a GABA-B agonist, frequently used
Isaac’s syndrome
■ Parkinson’s disease or Parkinson-plus
to treat spasticity, along with benzodiazepines.
Twenty years ago, it was proposed as a treatment
syndromes (eg, progressive supranuclear
palsy, multiple system atrophy) for SPS. Most patients are maintained on oral
■ Primary lateral sclerosis baclofen. Sometimes high doses are needed, which
■ Dystonia (generalised and focal) may cause disabling cognitive side effects. Due to
■ Ankylosing spondylitis poor CSF bioavailability, intrathecal baclofen is
■ Neuroleptic malignant syndrome, malignant used for severe spasticity and can significantly
hyperthermia and serotonin syndrome improve the features of both classical SPS and its
■ Tetanus variant, progressive encephalitis with rigidity and
■ Psychogenic
myoclonus.32
■ Hereditary spastic paraparesis
Silbert et al undertook a double-blind placebo-
■ Leukodystrophies
controlled trial of intrathecal baclofen in three
■ Drug-induced and toxicity: monoamine
patients with SPS. While only one patient reported
oxidase inhibitors, phenothiazines,
subjective improvement, all three patients
amphetamines, 5,6-methylenedioxy-N-
methyl-2-aminoindane, 1-methyl-4-phenyl- showed significant electrophysiological evidence
1,2,3,6-tetrahydropyridine, carbon monoxide of improvement and a trend towards improved
■ Spinal interneuronitis with rigidity muscle stiffness on objective scales.33
■ Diseases associated with positive Intrathecal baclofen is typically prescribed to
GAD autoantibodies patients requiring high-dose benzodiazepines but
■ Cerebellar ataxia experiencing intolerable side effects. Clinicians
■ Epilepsy must be cautious when using intrathecal baclofen
■ Limbic encephalitis
since interruption in drug delivery can lead to
■ Myasthenia gravis
severe symptomatic withdrawal state and even
■ Myoclonus
death from autonomic failure.34 Catheter mal-
■ Neuromyotonia

■ Batten’s disease
functions occurs in up to 40% of patients requir-
ing intrathecal infusion devices for spasticity;
GAD, glutamic acid decarboxylase.
there is a useful protocol for systematic checks if
this is suspected.35
clinical drug trials, hence limiting the quality of
Other options
treatment guidance. The last 30 years have pro-
Dantrolene and tizanidine have been tradition-
vided some insight into the various options avail-
ally used to manage conditions with spasticity,
able and are the foundation upon which current
including SPS. These are commonly combined
practice is based (see table 3). There remains an
with other muscle relaxants. Tiagabine, gabap-
important role for a multidisciplinary approach
entin, valproate and carbamazepine may all help
to management, including physiotherapy and
SPS symptoms; vigabatrin is rarely used because
occupational therapy input.
of its potential for visual field constriction.
Improving symptoms: muscle relaxants and other agents Levetiracetam has been the subject of a single-
Benzodiazepines blind placebo-controlled trial in three patients
Benzodiazepines augment GABA-dependent and benefited both symptoms and electrophysi-
pathways and are both anticonvulsant and anxi- ological findings.36 Propofol helped the stiff limb
olytic. They are also profound muscle relaxants variant of SPS in a single case refractory to other
and have long been a mainstay treatment of SPS. therapeutic strategies. Intramuscular botulinum
Drugs such as diazepam remain preferred agents toxin A can significantly improve muscle tone and
for symptom management in SPS. Over time, spasms, ambulation and pain in SPS. Analgesics
patients often need increasing doses for symptom remain an important part of SPS treatment.

Practical Neurology 2011;11:272–282. doi:10.1136/practneurol-2011-000071 279


REVIEW

Table 3 Main treatment options in stiff person syndrome


Agent Action Daily doses Side effects
Drugs treating symptoms
Benzodiazepines, eg, diazepam, clonazepam GABA-A agonist Diazepam 5–100 mg (divided doses) Drowsiness, vertigo, dysarthria,
Clonazepam 1–6 mg (divided doses) respiratory depression
(though often doses are far higher)
Baclofen GABA-B agonist Oral 5–60 mg (divided doses) Drowsiness, vertigo
Intrathecal 50–800 μg/day
Antiepileptics, eg, levetiracetam, gabapentin GABA-ergic and other Levetiracetam 2000 mg Variable
actions Gabapentin 3600 mg
Other options: tizanidine, dantrolene, botulinum toxin
Drugs that modulate the immune process
IVIG Incompletely understood 2 g/kg Infusion reactions including
anaphylaxis, thrombotic events,
headaches, aseptic meningitis
Plasma exchange Incompletely understood 5–6 Exchanges Hypotension, bleeding, allergic
reaction, severe immune
suppression
Rituximab B cell depletion 2 g (Divided doses) Breathing problems, arrhythmia
and rarely skin reactions
(Stevens–Johnson syndrome)
and progressive multifocal
leucoencephalopathy

Other options: corticosteroids, mycophenolate mofetil, cyclophosphamide, cyclosporine, tacrolimus, sirolimus.


Other treatments: analgesics, physiotherapy, occupational therapy.
GABA, γ-aminobutyric acid; IVIG, intravenous immunoglobulin.

However, clinicians should be aware that opi- While IVIG is generally considered safe, neurol-
ate analgesics, while reducing the pain of rigidity ogists should be aware of its common and impor-
and spasms may, on rare occasions, worsen these tant adverse effects. These include immediate
symptoms; follow-up monitoring is advisable infusion reactions (mild to severe) with a small
when commencing or uptitrating these drugs. but potentially risk of fatal anaphylaxis. This
Disease modifying immunomodulation/immunosuppression occurs typically in IgA-deficient patients, which
Intravenous immunoglobulin is therefore a relative contraindication for IVIG.
Intravenous immunoglobulin (IVIG) is the best There may also be skin reactions, headaches, asep-
second-line treatment for patients with severe or tic meningitis and renal tubular acidosis. Venous
refractory SPS. The evidence for this came origi- thromboembolic disease is a significant risk, par-
nally from several case reports where there were ticularly in those with limited mobility. Arterial
significant improvements in stiffness, startle, thrombus formation may lead to stroke, myocar-
functional status and clinical examination find- dial infarction, pulmonary embolism or ischaemia
ings, as well as most showing radiographic and affecting other tissue beds.
serological improvements. IVIG has subsequently Cost remains a major factor: treatment deci-
been shown to improve quality of life in SPS and sions in the UK are made on a case-by-case basis.
also to improve symptoms in the GAD-positive However, the rarity of SPS means that the over-
stiff limb variant. all cost of treating this patient group is much less
A randomised, double-blinded, placebo-con- than for a more common disease such as chronic
trolled, crossover trial of monthly IVIG demon- inflammatory demyelinating polyradiculoneu-
strated a significant decrease in stiffness, which ropathy. There have been regular updates in the
stabilised during washout and increased again published guidance on the use of IVIG in immune-
on switching to placebo. IVIG-treated patients mediated neuromuscular diseases in recent
reported improvement of symptoms and abil- years. The European Federation of Neurological
ity to undertake activities of daily living, lasting Sciences recommends IVIG for patients with SPS
between 6 weeks and 1 year. The GAD autoanti- who respond incompletely to diazepam and/or
body titre also fell after IVIG.37 baclofen and who have a significant disability

280 Practical Neurology 2011;11:272–282. doi:10.1136/practneurol-2011-000071


REVIEW

requiring a cane or walker due to truncal stiffness


PRACTICE POINTS
and frequent falls.38 The recommended dose is 2 ■ Stiff person syndrome (SPS) is underdiagnosed
g per kg over 2–5 days.
and often misdiagnosed.
Plasma exchange ■ The most prominent clinical finding is
The evidence supporting plasma exchange for hyperlordosis due to lumbar paraspinal
SPS is less well established than for IVIG and and abdominal muscle cocontraction with
there have been several conflicting results. superimposed spasms.
■ Most cases have a clear autoimmune basis,
Plasma exchange was first used successfully over
characterised by autoantibodies against
20 years ago. In anecdotal reports, some patients
glutamic acid decarboxylase and γ-aminobutyric
enjoyed improvements in symptoms and serologi- acid (A) receptor-associated protein. Rarer
cal and electrophysiological markers; however, paraneoplastic variants occur, with antibodies
equal numbers had no benefit. Patients showing against gephyrin and amphiphysin.
improvement tended to be on beneficial con- ■ In all, 30–40% of SPS patients have, or develop,

comitant medications. To date, there has been no diabetes mellitus.


reported randomised placebo-controlled study of ■ The diagnosis of SPS should be based
plasma exchange in SPS. on established clinical, laboratory and
electromyography criteria. Cases that do not fit
Rituximab within these criteria should be labelled atypical
As with many autoimmune disorders, the disease or an alternative diagnosis sought.
course should, in theory, be altered by depletion ■ Psychiatric disorders are common in SPS;
of mature B cells using rituximab, the chimeric referral to a psychiatrist may be necessary as
anti-CD20 monoclonal autoantibody. Rituximab part of the long-term management strategy.
was recently shown to give symptomatic and ■ Therapeutic approaches include symptomatic

serological remission in patients with otherwise therapy and immunomodulatory therapy. Some
refractory SPS.39 patients require (and can tolerate) very large
doses of diazepam; combination symptomatic
Other immunomodulatory agents treatments are often necessary. Intravenous
Corticosteroids have often been used in patients immunoglobulin is a mainstay treatment for
with SPS either as monotherapy or combined with those refractory to symptomatic treatment and
other therapeutic agents with improvement of is used increasingly early in the disease.
spasms and autoantibody titre. However, there has
never been a good quality clinical trial to determine
their overall role in SPS. Other immune system
modulating agents give variable benefit, including including DM and cancer. Further exploration
mycophenolate mofetil, azathioprine, cyclophos- of the link between SPS and DM could help to
phamide, cyclosporine, tacrolimus and sirolimus. predict and classify DM, to alter the implica-
tions of its therapy and to prevent its associated
Prognosis and future prospects morbidities.29
SPS follows a variable course over 6–28 years, For SPS, the aim should be earlier recognition
measured from symptom onset to either the and treatment under the care of a neurologist. SPS
last follow-up visit or death. Its rate of progres- is severely disabling, can substantially affect life
sion depends on several factors, including: (1) expectancy and impair physical and mental capa-
whether the initial presentation and symptoms bilities. Disability results in a reduced quality of
are of classical SPS, (2) belong to the ‘stiff man life and affects an individual’s potential for educa-
plus’ category or (3) whether it is associated with tion and earning. Therefore, a better understand-
disorders such as DM or malignancy.21 Patients ing of the natural history, mechanisms of disease
with classic SPS usually respond well to treatment and the impact of disease progression over time,
and their condition stabilises over time, although along with the long-term effects of treatment, are
paroxysmal autonomic dysfunction or sudden needed to make further progress.
death occurs in 10% of SPS patients.21 Autonomic
Acknowledgements The authors thank Dr Jeremy
dysfunction results from a succession of spasms or
Rees for permission to reproduce figure 3. This paper
sudden withdrawal of medication.40 was reviewed by Jeremy Brown, London, UK.
Although SPS is rare, it can cause significant Competing interests None.
morbidity and mortality; its pathogenesis relates Provenance and peer review Commissioned;
to diseases with major public health impact, externally peer reviewed.

Practical Neurology 2011;11:272–282. doi:10.1136/practneurol-2011-000071 281


REVIEW

References 21. Gershanik OS. Stiff-person syndrome. Parkinsonism Relat


1. Dalakas MC. Stiff person syndrome: advances in pathogenesis Disord 2009;15(Suppl 3):S130–4.
and therapeutic interventions. Curr Treat Options Neurol 22. Meinck HM. Stiff man syndrome. CNS Drugs 2001;
2009;11:102–10. 15:515–26.
2. Moersch FP, Woltman HW. Progressive fluctuating muscular 23. Ameli R, Snow J, Rakocevic G, et al. A neuropsychological
rigidity and spasm (“stiff-man” syndrome); report of a case and assessment of phobias in patients with stiff person syndrome.
some observations in 13 other cases. Proc Staff Meet Mayo Clin Neurology 2005;64:1961–3.
1956;31:421–7. 24. Gerschlager W, Schrag A, Brown P. Quality of life in stiff-
3. Gordon EE, Januszko DM, Kaufman L. A critical survey of person syndrome. Mov Disord 2002;17:1064–7.
stiff-man syndrome. Am J Med 1967;42:582–99. 25. Ruivard M, Berger C, Achaibi A, et al. Physician compliance
4. Lorish TR, Thorsteinsson G, Howard FM Jr. Stiff-man with outpatient oral anticoagulant guidelines in Auvergne,
syndrome updated. Mayo Clin Proc 1989;64:629–36. France. J Gen Intern Med 2003;18:903–7.
5. Solimena M, Folli F, Denis-Donini S, et al. Autoantibodies 26. Economides JR, Horton JC. Eye movement abnormalities in
to glutamic acid decarboxylase in a patient with stiff-man stiff person syndrome. Neurology 2005;65:1462–4.
syndrome, epilepsy, and type I diabetes mellitus. N Engl J Med 27. Barker RA, Revesz T, Thom M, et al. Review of 23 patients
1988;318:1012–20. affected by the stiff man syndrome: clinical subdivision
6. Howard FM Jr. A new and effective drug in the treatment of into stiff trunk (man) syndrome, stiff limb syndrome, and
the stiff-man syndrome: preliminary report. Proc Staff Meet progressive encephalomyelitis with rigidity. J Neurol Neurosurg
Mayo Clin 1963;38:203–12. Psychiatr 1998;65:633–40.
7. Piccolo G, Cosi V, Zandrini C, et al. Steroid-responsive 28. Leslie RD. Predicting adult-onset autoimmune diabetes: clarity
and dependent stiff-man syndrome: a clinical and from complexity. Diabetes 2010;59:330–1.
electrophysiological study of two cases. Ital J Neurol Sci 29. Brown P, Marsden CD. The stiff man and stiff man plus
1988;9:559–66. syndromes. J Neurol 1999;246:648–52.
8. Vicari AM, Folli F, Pozza G, et al. Plasmapheresis in 30. Gouider-Khouja N, Mekaouar A, Larnaout A, et al. Progressive
the treatment of stiff-man syndrome. N Engl J Med encephalomyelitis with rigidity presenting as a stiff-person
1989;320:1499. syndrome. Parkinsonism Relat Disord 2002;8:285–8.
9. Amato AA, Cornman EW, Kissel JT. Treatment of stiff-man 31. McCombe PA, Chalk JB, Searle JW, et al. Progressive
syndrome with intravenous immunoglobulin. Neurology encephalomyelitis with rigidity: a case report with magnetic
1994;44:1652–4. resonance imaging findings. J Neurol Neurosurg Psychiatr
10. Raju R, Rakocevic G, Chen Z, et al. Autoimmunity to GABAA- 1989;52:1429–31.
receptor-associated protein in stiff-person syndrome. Brain 32. Stayer C, Tronnier V, Dressnandt J, et al. Intrathecal
2006;129:3270–6. baclofen therapy for stiff-man syndrome and progressive
11. Folli F, Solimena M, Cofiell R, et al. Autoantibodies to a encephalomyelopathy with rigidity and myoclonus. Neurology
128-kd synaptic protein in three women with the stiff-man 1997;49:1591–7.
syndrome and breast cancer. N Engl J Med 1993;328:546–51. 33. Silbert PL, Matsumoto JY, McManis PG, et al. Intrathecal
12. Butler MH, Hayashi A, Ohkoshi N, et al. Autoimmunity to baclofen therapy in stiff-man syndrome: a double-blind,
gephyrin in Stiff-Man syndrome. Neuron 2000;26:307–12. placebo-controlled trial. Neurology 1995;45:1893–7.
13. Levy LM, Dalakas MC, Floeter MK. The stiff-person syndrome: 34. Meinck HM, Tronnier V, Rieke K, et al. Intrathecal baclofen
an autoimmune disorder affecting neurotransmission of gamma- treatment for stiff-man syndrome: pump failure may be fatal.
aminobutyric acid. Ann Intern Med 1999;131:522–30. Neurology 1994;44:2209–10.
14. Thompson PD. The stiff-man syndrome and related disorders. 35. Bardutzky J, Tronnier V, Schwab S, et al. Intrathecal baclofen
Parkinsonism Relat Disord 2001;8:147–53. for stiff-person syndrome: life-threatening intermittent catheter
15. Nollet H, Vanderstraeten G, Sustronck B, et al. Suspected case leakage. Neurology 2003;60:1976–8.
of stiff-horse syndrome. Vet Rec 2000;146:282–4. 36. Sechi G, Barrocu M, Piluzza MG, et al. Levetiracetam in stiff-
16. Solimena M, Folli F, Aparisi R, et al. Autoantibodies to GABA- person syndrome. J Neurol 2008;255:1721–5.
ergic neurons and pancreatic beta cells in stiff-man syndrome. 37. Dalakas MC, Fujii M, Li M, et al. High-dose intravenous
N Engl J Med 1990;322:1555–60. immune globulin for stiff-person syndrome. N Engl J Med
17. Lohmann T, Hawa M, Leslie RD, et al. Immune reactivity to 2001;345:1870–6.
glutamic acid decarboxylase 65 in stiffman syndrome and type 38. Elovaara I, Apostolski S, van Doorn P, et al. EFNS guidelines
1 diabetes mellitus. Lancet 2000;356:31–5. for the use of intravenous immunoglobulin in treatment
18. Ellis TM, Atkinson MA. The clinical significance of an of neurological diseases: EFNS task force on the use of
autoimmune response against glutamic acid decarboxylase. intravenous immunoglobulin in treatment of neurological
Nat Med 1996;2:148–53. diseases. Eur J Neurol 2008;15:893–908.
19. Alberts B, Johnson A, Lewis J, et al. Molecular Biology of the 39. Baker MR, Das M, Isaacs J, et al. Treatment of stiff person
Cell. 4th edition. New York, NY: Garland Science Publishing syndrome with rituximab. J Neurol Neurosurg Psychiatr
Company 2001:711–26. 2005;76:999–1001.
20. Dalakas MC, Fujii M, Li M, et al. The clinical spectrum of anti- 40. Mitsumoto H, Schwartzman MJ, Estes ML, et al. Sudden death
GAD antibody-positive patients with stiff-person syndrome. and paroxysmal autonomic dysfunction in stiff-man syndrome.
Neurology 2000;55:1531–5. J Neurol 1991;238:91–6.

282 Practical Neurology 2011;11:272–282. doi:10.1136/practneurol-2011-000071

You might also like