You are on page 1of 8

REVIEW

The progressive myoclonic epilepsies


Naveed Malek,1 William Stewart,2 John Greene1

1
Department of Neurology, ABSTRACT Sachs disease), then it is crucial to get the
Institute of Neurological Progressive myoclonic epilepsies are a group of diagnosis right.3–6
Sciences, Southern General
Hospital, Glasgow, UK disorders characterised by a relentlessly
2
Department of Neuropathology, progressive disease course until death; treatment- Unverricht–Lundborg disease
Institute of Neurological resistant epilepsy is just a part of the phenotype. Unverricht–Lundborg disease—also called
Sciences, Southern General
Hospital, Glasgow, UK
This umbrella term encompasses many diverse progressive myoclonic epilepsy type 1
conditions, ranging from Lafora body disease to (EPM1)—is autosomal-recessively inher-
Correspondence to Gaucher’s disease. These diseases as a group are ited and is characterised by stimulus-
Dr N Malek, Department of important because of a generally poor response sensitive myoclonus and tonic–clonic
Neurology, Southern General
Hospital, Glasgow G51 4TF, UK; to antiepileptic medication, an overall poor seizures. The age of onset is usually 6–16
nmalek@nhs.net prognosis and inheritance risks to siblings or years. Some years after the disease onset,
offspring (where there is a proven genetic cause). patients may develop cerebellar features,
Accepted 1 February 2015
A correct diagnosis also helps patients and their such as ataxia, incoordination, intention
families to accept and understand the nature of tremor and dysarthria. Individuals with
their disease, even if incurable. Here, we discuss Unverricht–Lundborg disease are men-
the phenotypes of these disorders and tally alert but show emotional lability,
summarise the relevant specific investigations to depression and mild decline in intellec-
identify the underlying cause. tual performance over time.7 EPM1
results from mutations in the CSTB gene
INTRODUCTION causing defective function of cystatin B, a
The progressive myoclonic epilepsies cysteine protease inhibitor.8
comprise a devastating group of rare dis-
orders that manifest with worsening Lafora body disease
action myoclonus; it is also present at rest Lafora body disease—also called progres-
but activates with stimuli such as noise, sive myoclonic epilepsy type 2 (EPM2)
light or touch.1 Other neurological fea- and named after the Spanish neurologist
tures that frequently but not reliably Gonzalo Lafora—is an autosomal-recessive
coexist include other seizure types ( par- form of progressive myoclonus epilepsy.
ticularly generalised tonic–clonic), pro- It is characterised by myoclonus, tonic–
gressive ataxia and dementia. Typically, clonic seizures, visual hallucinations,
the presentation is in late childhood or intellectual decline and progressive neuro-
adolescence; however, they may affect all logical deterioration.9 The age of onset is
ages. Ultimately, patients become wheel- usually 12–15 years, but an earlier onset
chair users and have a reduced life expect- variant begins at the age of 5 years.10 11
ancy. It may be challenging to distinguish Lafora body disease is caused by mutations
these conditions very early on from more either in the EPM2A gene (encoding for
common forms of genetic generalised epi- laforin, a dual-specificity protein phosphat-
lepsy, particularly juvenile myoclonic epi- ase) or in the EPM2B (NHLRC1) gene
lepsy. However, features suggesting (encoding malin, an E3- ubiquitin ligase).11
progressive myoclonic epilepsy are the These two proteins interact and, as a
presence or evolution of progressive complex, regulate glycogen synthesis.
neurological disability, failure to respond Lafora body disease is, therefore, a dis-
to antiepileptic drug therapy and back- order of carbohydrate metabolism resulting
ground slowing on EEGs.2 Many progres- in polyglucosan inclusion bodies in neural
sive myoclonic epilepsies have similar and other tissues.12
clinical presentations yet are genetically Tissue biopsy (axillary skin) reveals
To cite: Malek N, Stewart W, heterogeneous, making accurate diagnosis Lafora bodies, which are aggregates of poly-
Greene J. Pract Neurol difficult. Therein lies the importance of glucosans (poorly constructed glycogen
Published Online First: [ please
include Day Month Year] getting the diagnosis right: if we are to molecules). Lafora bodies are pathogno-
doi:10.1136/practneurol- investigate specific treatments for individ- monic and do not occur in any other
2014-000994 ual disorders (eg, gene therapy for Tay– condition.

Malek N, et al. Pract Neurol 2015;0:1–8. doi:10.1136/practneurol-2014-000994 1


REVIEW
Action myoclonus renal failure syndrome Myoclonus does not occur in all mitochondrial dis-
Action myoclonus renal failure (AMRF) syndrome— eases (3.6% of 1086 patients in one study) but is prom-
also called progressive myoclonic epilepsy type 4 inent in MERRF.23 Their myoclonus is not inextricably
(EPM4)—is a distinctive form of progressive myoclonus linked to epilepsy, but more so to cerebellar ataxia.23
epilepsy associated with renal dysfunction.13 It is an MERRF is most commonly caused by a mutation in the
autosomal-recessive disease related to loss-of-function tRNALys gene in mitochondrial DNA at nucleotide pos-
mutations in SCARB2 gene.14 The onset is in the second ition 8344, leading to altered mitochondrial function,
and third decades, but there is also a late-onset form, but there are also other point mutations.24 25
starting in the fifth and sixth decades and without renal
failure.15 16 AMRF shows genotype–phenotype hetero-
Neuronal ceroid lipofuscinoses
geneity, such that affected people within the same
sibship, despite identical gene mutations, can present The neuronal ceroid lipofuscinoses (CLN) comprise a
differently: some with neurological features while others heterogeneous group of inherited, neurodegenerative,
with renal impairment.17 lysosomal storage disorders characterised by progres-
sive cognitive and motor deterioration, progressive
tonic–clonic as well as myoclonic seizures, and early
PRICKLE1-gene-related progressive myoclonic epilepsy death. Most forms have visual loss.26 The clinical phe-
with ataxia notypes traditionally divide into infantile, late infant-
PRICKLE1-gene-related progressive myoclonic epi- ile, juvenile and adult, based on age at onset.
lepsy—also called progressive myoclonic epilepsy type However, a new classification system takes into
5 (EPM5)—is characterised by myoclonic seizures, account both the gene involved (genes designated
generalised tonic–clonic seizures (often sleep-related) with CLN loci from 1 to 14) and the age at disease
and ataxia, but with normal cognition. The age of onset; for example, (CLN1 disease, infantile onset)
onset is 5–10 years. Action myoclonus may affect the and (CLN1 disease, juvenile onset) are both caused by
limbs or bulbar muscles, sometimes with spontaneous mutations in PPT1 but with differing age of onset,
myoclonus of facial muscles causing marked dysarth- and with considerable phenotype–genotype hetero-
ria.18 PRICKLE proteins, such as PRICKLE1, are core geneity (table 1).26 These are usually autosomal reces-
constituents of the planar cell polarity-signalling sive but there are also autosomal-dominant late-onset
pathway that establishes cell polarity during embry- forms.27 In childhood, the neuronal CLN are the
onic development.19 most common lysosomal diseases and the most
common neurodegenerative diseases, but, in adults,
‘North Sea’ progressive myoclonus epilepsy they represent a small fraction of the neurodegenera-
‘North Sea’ progressive myoclonus epilepsy—also tive diseases.28 The adult type (Kufs’ disease) is the
called progressive myoclonic epilepsy type 6 (EPM6) rarest of all the subtypes of neuronal CLN. The
—is characterised by ataxia starting around aged pathological findings on biopsy are abnormal auto-
2 years and followed by myoclonic seizures when fluorescent lipopigments from lysosomal inclusion
aged 6–7 years. Patients have multiple seizure types, bodies deposited in brain, myenteric plexus, muscle
including generalised tonic–clonic seizures, absence and skin. Electron microscopy has a special role in
seizures and drop attacks. Most patients develop scoli- their diagnosis. The characteristic ultrastructural
osis by adolescence: an important diagnostic clue. abnormalities in neuronal CLN, seen in conveniently
There may also be skeletal deformities, including pes available biopsy specimens (skin) or lymphocytes
cavus and syndactyly. The condition is caused by (buffy coat), include one (or a combination) of
mutations in the Golgi SNAP receptor complex 2
gene (GOSR2).20 Patients have elevated serum creatine
kinase concentrations (median ≈700 IU) in the
context of a normal muscle biopsy.21

Myoclonic epilepsy with ragged-red fibres


Myoclonic epilepsy with ragged-red fibres (MERRF)
is a multisystem mitochondrial disorder, named after
its characteristic muscle biopsy appearances (figure 1).
The onset is usually in childhood, after normal early
development. The first symptom is often myoclonus,
followed by generalised epilepsy, ataxia, weakness and
dementia. Common associated findings are hearing
loss, short stature, optic atrophy and cardiomyopathy
with Wolff–Parkinson–White syndrome. Some patients Figure 1 A ragged-red fibre in a muscle biopsy specimen (top
have pigmentary retinopathy and/or lipomatosis.22 left) on the Gomori trichome stain.

2 Malek N, et al. Pract Neurol 2015;0:1–8. doi:10.1136/practneurol-2014-000994


REVIEW
Table 1 Neuronal ceroid lipofuscinosis (CLN) loci and genotype–phenotype correlations with characteristic electron microscopy
findings in biopsy samples
Locus Age of onset (phenotype) Gene Electron microscopy
CLN1 Infantile, late infantile, juvenile, adult (Kufs’ disease) PPT1 Granular osmophilic deposits
CLN2 Late infantile, juvenile TPP1 Curvilinear profiles
CLN3 Adult (Kufs’ disease) CLN3 Fingerprint profiles
CLN4 Adult (Parry’s disease) DNAJC5 Granular osmophilic deposits, mixed
CLN5 Late infantile CLN5 Fingerprint profiles
CLN6 Late infantile, adult (Kufs’ disease) CLN6 Curvilinear profiles, fingerprint profiles, Rectilinear complex
CLN7 Late infantile MFSD8 Curvilinear profiles, fingerprint profiles, rectilinear complex
CLN8 Late infantile (Northern epilepsy) CLN8 Curvilinear profiles or granular osmophilic deposits
CLN9 Juvenile Unknown Granular osmophilic deposits, curvilinear profiles
CLN10 Congenital CTSD Granular osmophilic deposits
CLN11 Adult (Kufs’ disease) GRN Fingerprint profiles
CLN12 Juvenile ATP13A2 Granular osmophilic deposits, mixed
CLN13 Adult (Kufs’ disease) CTSF Fingerprint profiles or none
CLN14 Infantile KCTD7 Granular osmophilic deposits, fingerprint profiles
Adapted from ref. 30.
Mixed indicates curvilinear profiles, fingerprint profiles, rectilinear complex and granular osmophilic deposits.
PPT-1, palmitoyl-protein thioesterase 1; TPP-1, tripeptidyl peptidase 1.

fingerprint, curvilinear and membranous profile inclu- progressive myoclonic epilepsy phenotype have larger
sions in the lysosomes (table 1).29 expansions (62–79 repeats) and earlier age of onset
(before aged 20 years) while those with a non-
progressive myoclonic epilepsy phenotype have a later
Dentatorubral-pallidoluysian atrophy
age of onset (after aged 20 years) and smaller expan-
Dentatorubral-pallidoluysian atrophy (DRPLA), unlike
sions (54–67 repeats).32
other progressive myoclonus epilepsies, is an
autosomal-dominant disorder characterised by epi-
Sialidosis type 1 (cherry-red spot myoclonus syndrome)
lepsy, cerebellar ataxia, choreoathetosis, myoclonus,
dementia and psychiatric symptoms in varying combi- Sialidosis, also called mucolipidosis type I, is an
nations. DRPLA is caused by an unstable expansion of autosomal-recessive lysosomal storage disease caused
CAG repeats in exon 5 of the DRPLA gene on by a deficiency of the enzyme α-N-acetyl
chromosome 12, coding for polyglutamine tracts. neuraminidase-1 (coded by the NEU1 gene on
Being a trinucleotide repeat disorder, it also shows the chromosome 6p21). It is classified into two main clin-
phenomenon of anticipation, with paternal transmis- ical variants: type 1, the milder variant, and type 2,
sion resulting in more prominent anticipation than usually more severe and with an earlier onset.33 The
maternal transmission. DRPLA protein (also called disease is characterised by myoclonic epilepsy, visual
atrophin-1) is located in the nucleus and functions as problems, hyperreflexia and ataxia that develop in the
a transcription coregulator.31 Patients with a second or the third decade of life.34 Although patients
with sialidosis always have macular cherry-red spots
(figure 2), this is not a pathognomonic finding since
they also occur in central retinal artery occlusion and
metabolic storage diseases such as Tay–Sachs disease,
Sandhoff ’s disease, Niemann–Pick disease, Fabry’s
disease and Gaucher’s disease, some of which can also
have a progressive myoclonic epilepsy phenotype.35
The characteristic pathology of sialidosis reflects tissue
accumulation and urinary excretion of sialylated
oligosaccharides.36

GM2 gangliosidosis (Tay–Sachs disease and variants)


The GM2 gangliosidoses comprise a group of
autosomal-recessive disorders characterised by accu-
mulation of GM2 ganglioside, one type of glycolipid,
Figure 2 A cherry-red spot in the macula on a fundus in neuronal cells. The genes responsible for these dis-
photograph from the left eye. orders are HEXA (Tay–Sachs disease and variants),

Malek N, et al. Pract Neurol 2015;0:1–8. doi:10.1136/practneurol-2014-000994 3


REVIEW

HEXB (Sandhoff ’s disease and variants) and GM2A NEUROPHYSIOLOGY


(AB variant of GM2 gangliosidosis).37 The proteins Not all progressive myoclonic epilepsies have well-
encoded by these three genes are the α-subunits characterised neurophysiological findings, but we shall
(HEXA gene) and β-subunits (HEXB gene) of describe those of the most common disorders. Lafora
β-hexosaminidase A enzyme. The third is a small gly- body patients’ EEGs show varying degrees of slowing
colipid transport protein, the GM2 activator protein of background activity (in the vast majority), general-
(GM2A), which acts as a substrate specific cofactor ised epileptiform discharges (in 85%), focal discharges
for the enzyme. A deficiency of any one of these pro- (in about a third) and photosensitivity to a
teins leads to storage of the ganglioside, primarily in fast-frequency stimulus (in a quarter). About two-thirds
the lysosomes of neuronal cells, causing cell death.38 of patients show giant somatosensory-evoked poten-
The three subtypes are clinically indistinguishable tials (14–175 mV) (figure 3).45
apart from subtle visceral and skeletal manifestations In Unverricht–Lundborg disease, the scalp EEG
in some people with Sandhoff ’s disease.39 The most shows generalised epileptiform discharges in the
severe early onset form of Tay–Sachs disease is charac- majority of patients; about half of the patients show
terised by a progressive and relentless neuronal dys- giant somatosensory-evoked potentials.45 In MERRF,
function, manifesting as hypotonia, blindness, EEG in about two-thirds of patients shows slowing
dementia, seizures and subsequently death, usually by of background activity with/without generalised
aged 3–5 years.40 Late-onset GM2 gangliosidosis epileptiform discharges; a minority show giant
shows a wide range of symptoms, including cerebellar somatosensory-evoked potentials. There may be coex-
ataxia, dystonia, motor neurone disease, psychiatric istent neuropathy or myopathy.45
symptoms, dementia and rarely polyneuropathy.41 In neuronal CLN, EEG shows varying degrees of
diffuse slowing of background activity (in 95%) and
generalised epileptiform discharges (in 80%); about a
Gaucher’s disease quarter of patients show giant somatosensory-evoked
Gaucher’s disease is the most prevalent lysosomal potentials. Nerve conduction studies show evidence of
storage disease and is caused by >200 mutations that axonal neuropathy in about 30% of patients.45
produce abnormal glucocerebrosidase.42 Gaucher’s
disease can have several phenotypes, ranging from a Diagnostic testing
perinatal lethal form to an asymptomatic type.43 Investigations should be directed towards the pheno-
There are three major clinical phenotypes (1, 2 and type that best fits the patient’s condition; however,
3). Only types 2 and 3 involve the central nervous the clinical phenotypes of the progressive myoclonic
system and are distinguished by age at onset and the epilepsies overlap sufficiently such that it is not pos-
rate of disease progression, but these distinctions are sible to make a definitive clinical diagnosis of individ-
not absolute. Type 3 cases can cause several neuro- ual disorders, and hence the reliance on laboratory,
logical impairments, including a horizontal gaze genetic and ancillary investigations. Some of the pro-
abnormality, progressive dementia, generalised epi- gressive myoclonic epilepsies are metabolic disorders
lepsy, ataxia and spasticity but also a progressive myo- such as Gaucher’s disease and Tay–Sachs disease, and
clonic epilepsy phenotype.44 these can be diagnosed with white cell enzyme ana-
lysis; other conditions such as Lafora body disease
and the neuronal CLN have characteristic histopatho-
logical findings on skin biopsy. Others such as AMRF
syndrome and DRPLA can be diagnosed only with
genetic tests (table 2).

TREATMENT
There are specific treatments for some forms of pro-
gressive myoclonus epilepsy. For example, metabolic
disorders such as Gaucher’s disease may respond to
substrate reduction therapy or enzyme replacement
therapy. Enzyme replacement therapy is now consid-
ered the gold standard for the management of
Gaucher’s disease type 1 and for the non-neurological
manifestations of Gaucher’s disease type 3.46 For con-
Figure 3 Giant somatosensory-evoked N20/P22 potentials at
the central scalp (Cc) electrode on the left side, with normal
ditions where there is no specific therapy, such as
somatosensory-evoked potentials from another person on the Gaucher’s disease type 2, clinicians can offer only
right side. Fc, Fz, Fi, Cc and Cz refer to scalp electrode symptom treatment with antiepileptic drugs to control
positions. seizures.

4 Malek N, et al. Pract Neurol 2015;0:1–8. doi:10.1136/practneurol-2014-000994


REVIEW

Table 2 The investigative workup for progressive myoclonic epilepsies


Disorder Investigations
1. Unverricht–Lundborg disease (EPM1) Gene test: EPM1 (CSTB) mutation analysis
2. Lafora body disease (EPM2) 1. Skin biopsy: Lafora bodies
2. Gene tests: EPM2A or EPM2B (NHLRC1) mutation analysis
3. Action myoclonus renal failure syndrome (EPM4) Gene test: SCARB2/LIMP2 mutation analysis
4. PRICKLE1-related progressive myoclonus epilepsy with Gene test: PRICKLE1 mutation analysis
ataxia (EPM5)
5. ‘North Sea’ progressive myoclonus epilepsy (EPM6) Gene test: GOSR2 mutation analysis
6. MERRF 1. Plasma lactate, pyruvate (as screen)
2. Muscle biopsy: for ragged-red fibres
3. Gene test: MT-TK mutation analysis
7. Neuronal ceroid lipofuscinosis (CLN) 1. Skin biopsy (electron microscopy): curvilinear profiles, fingerprint profiles, granular
osmophilic deposits
2. Leucocyte enzyme analysis (PPT1,TPP1, CTSD)
3. Gene tests: PPT1, TPP1, CLN3, CLN5, CLN6, MFSD8, CLN8, CTSD, DNAJC5, CTSF,
ATP13A2, GRN, KCTD7 mutation analysis
8. DRPLA Gene test: DRPLA mutation analysis
(CAG repeats)
9. Sialidosis type I 1. Sialo-oligosaccharides in urine
(cherry-red spot myoclonus syndrome) 2. Leucocyte enzyme analysis (neuraminidase)
3. Gene test: NEU1 mutation analysis
10. Tay–Sachs disease (GM2 gangliosidosis) 1. Leucocyte enzyme analysis (hexosaminidase A, B)
2. Gene test: HEXA mutation analysis

11. Gaucher’s disease 1. Leucocyte enzyme analysis (β-glucocerebrosidase)


2. Gene test: GBA mutation analysis
DRPLA, dentatorubral-pallidoluysian atrophy; EPM, progressive myoclonus epilepsy.

Valproate is often the first choice to treat myoclonic myoclonic epilepsies, the combination of valproate,
seizures because of its broad spectrum of antiepileptic clonazepam and phenobarbital was superior to con-
action and its good myoclonus suppression potential. ventional antiepileptic drugs such as phenytoin,
It may be less preferable in women owing to its terato- carbamazepine, primidone and diazepam.49 In fact,
genicity but should not be denied to women who clonazepam is the only drug approved by the US
have no plans to conceive, in the face of an incurable Food and Drug Administration as monotherapy for
disease. The rationale for using valproate to treat the treatment of myoclonic seizures,50 but clobazam
myoclonus in progressive myoclonic epilepsies is can be used for brief periods to control seizure clus-
based on trials of its efficacy in juvenile myoclonic ters. Levetiracetam and topiramate are also highly
epilepsy, a myoclonic epilepsy with a comparatively effective for myoclonic seizures and are often used in
benign prognosis.47 Sometimes valproate alone cannot combination or as second-line treatment. On the
achieve seizure control. Also, valproate should be other hand, some antiepileptic drugs may exacerbate
avoided in MERRF due to concerns about liver failure or even induce myoclonus51 and clearly should be
when used in patients with other mitochondrial disor- avoided, particularly sodium channel blockers (carba-
ders.48 In a small trial of 26 adults with progressive mazepine, oxcarbazepine) and GABAergic drugs (tia-
gabine, vigabatrin), as well as gabapentin and
pregabalin.7
Lamotrigine does not appear to be useful in pro-
Table 3 Antiepileptic drugs that may be useful in progressive
gressive myoclonic epilepsies due to poor efficacy,
myoclonic epilepsies (on left) and drugs that may worsen
myoclonus (on the right) dose-related exacerbation of myoclonus and putative
late-onset worsening.52 Lamotrigine also may exacer-
Drugs useful in progressive Drugs that exacerbate bate myoclonus in non-progressive myoclonic epilepsy
myoclonic epilepsies myoclonus
seizure states.53 54 The evidence for levetiracetam,
Sodium valproate* Lamotrigine topiramate and zonisamide in progressive myoclonic
Levetiracetam Phenytoin epilepsies comes from case series; these drugs may
Topiramate Carbamazepine reduce myoclonus besides their efficacy in treating
Clonazepam Oxcarbazepine generalised tonic–clonic seizures.55–57
Zonisamide Tiagabine Finally, high-dose piracetam has been found to be
Phenobarbital Vigabatrin useful in the treatment of progressive myoclonic epi-
*Avoid valproate in myoclonic epilepsy with ragged-red fibres. lepsies58 (table 3).

Malek N, et al. Pract Neurol 2015;0:1–8. doi:10.1136/practneurol-2014-000994 5


REVIEW

Table 4 Some take home points from this review Provenance and peer review Not commissioned; externally
peer reviewed. This paper was reviewed by Reetta Kälviäinen,
Hint Consider Kuopio, Finland.
If there is deafness, cardiac problems, optic atrophy, MERRF
short stature or lipomas
If the disease onset is between 5 and 15 years after Lafora body REFERENCES
normal early development, with visual hallucinations disease
1 Lozsadi D. Myoclonus: a pragmatic approach. Pract Neurol
(occipital seizures) and normal fundi
2012;12:215–24.
If there is a macular cherry-red spot in addition to the Sialidosis
2 Knupp K, Wirrell E. Progressive myoclonic epilepsies: It takes a
myoclonus and sialo-oligosaccharides in urine
village to make a diagnosis. Neurology 2014;82:378–9.
If there is associated renal failure and/or family AMRF
3 Cachon-Gonzalez MB, Wang SZ, Ziegler R, et al. Reversibility
members have renal failure
of neuropathology in Tay-Sachs-related diseases. Hum Mol
If phenotype and family history suggest an DRPLA
Genet 2014;23:730–48.
autosomal-dominant inheritance and anticipation
4 Chang JC, Liu KH, Chuang CS, et al. Treatment of human
AMRF, action myoclonus renal failure syndrome, DRPLA,
cells derived from MERRF syndrome by peptide-mediated
dentatorubral-pallidoluysian atrophy; MERRF, myoclonic epilepsy with
ragged-red fibres. mitochondrial delivery. Cytotherapy 2013;15:1580–96.
5 Meng Y, Sohar I, Sleat DE, et al. Effective intravenous therapy
Despite all of the available antiepileptic drugs, clini- for neurodegenerative disease with a therapeutic enzyme and a
cians should recognise a risk of overmedication in peptide that mediates delivery to the brain. Mol Ther
treating drug-resistant myoclonus. Generalised convul- 2014;22:547–53.
sive seizures are often part of the phenotype of the 6 Bennett LL, Mohan D. Gaucher disease and its treatment
options. Ann Pharmacother 2013;47:1182–93.
progressive myoclonic epilepsies and these are well
7 Kalviainen R, Khyuppenen J, Koskenkorva P, et al. Clinical
controlled with classic antiepileptic drugs, whereas
picture of EPM1-Unverricht-Lundborg disease. Epilepsia
myoclonus—which can be the most disabling 2008;49:549–56.
symptom resulting in the patient becoming wheelchair 8 Lehesjoki AE, Kalviainen R. Unverricht-Lundborg disease.
user—is usually refractory to standard antiepileptic In: Pagon RA, Adam MP, Bird TD, Dolan CR, Fong CT,
drugs.59 It may be that the drug-resistant myoclonus is Stephens K, eds. GeneReviews. Seattle, WA: University of
subcortical in origin.60 Washington, 1993.
Progressive myoclonic epilepsies as a group are not 9 Couarch P, Vernia S, Gourfinkel-An I, et al. Lafora progressive
amenable to surgical resection. Vagus nerve stimulation myoclonus epilepsy: NHLRC1 mutations affect glycogen
can help control epileptic seizures and status epilepticus, metabolism. J Mol Med 2011;89:915–25.
but will not help the myoclonus or cerebellar symptoms, 10 Singh S, Ganesh S. Lafora progressive myoclonus epilepsy:
a meta-analysis of reported mutations in the first decade
and has no effect on cognitive impairment.61
following the discovery of the EPM2A and NHLRC1 genes.
Hum Mutat 2009;30:715–23.
PROGNOSIS 11 Turnbull J, Girard JM, Lohi H, et al. Early-onset Lafora body
The prognosis of this group of epilepsies is poor in terms disease. Brain 2012;135:2684–98.
of seizure control with antiepileptic drugs; however, the 12 Spuch C, Ortolano S, Navarro C. Lafora progressive myoclonus
natural histories of the individual disorders vary. In the epilepsy: recent insights into cell degeneration. Recent Pat Endocr
most devastating disorders, such as Lafora body disease, Metab Immune Drug Discov 2012;6:99–107.
there is a progressive neurological deterioration begin- 13 Badhwar A, Berkovic SF, Dowling JP, et al. Action
ning in adolescence, leading to a vegetative state in status myoclonus-renal failure syndrome: characterization of a unique
myoclonicus and death within 10 years9 (table 4). cerebro-renal disorder. Brain 2004;127:2173–82.
14 Hopfner F, Schormair B, Knauf F, et al. Novel SCARB2
mutation in action myoclonus-renal failure syndrome and
FUTURE DIRECTIONS
evaluation of SCARB2 mutations in isolated AMRF features.
There are some forms of progressive myoclonic epi- BMC Neurol 2011;11:134.
lepsies that have yet to be clinically and genetically 15 Higashiyama Y, Doi H, Wakabayashi M, et al. A novel SCARB2
characterised. Although the progressive myoclonic mutation causing late-onset progressive myoclonus epilepsy.
epilepsies as a group may perhaps be the rarest of the Mov Disord 2013;28:552–3.
inherited epilepsies, recent molecular genetic advances 16 Dibbens LM, Michelucci R, Gambardella A, et al. SCARB2
are unravelling the causes of previously nebulous mutations in progressive myoclonus epilepsy (PME) without
entities such as AMRF syndrome. In time, the pro- renal failure. Ann Neurol 2009;66:532–6.
gressive myoclonic epilepsies may well become the 17 Perandones C, Pellene LA, Micheli F. A new SCARB2 mutation
best understood of the epilepsies at the cellular level, in a patient with progressive myoclonus ataxia without renal
failure. Mov Disord 2014;29:158–9.
with the promise of specific treatments.
18 Fox MH, Bassuk AG. PRICKLE1-related progressive
Acknowledgements We thank Dr Graeme Williams, Consultant myoclonus epilepsy with ataxia. In: Pagon RA, Adam MP, Bird
Neuro-ophthalmologist, Institute of Neurological Sciences, TD, Dolan CR, Fong CT, Smith RJH, et al, eds. GeneReviews.
Glasgow, for the image of cherry-red spot in the retina. Seattle, WA: University of Washington, 1993.
Contributors All authors contributed equally to the paper. 19 Liu C, Lin C, Whitaker DT, et al. Prickle1 is expressed in
Competing interests None. distinct cell populations of the central nervous system and

6 Malek N, et al. Pract Neurol 2015;0:1–8. doi:10.1136/practneurol-2014-000994


REVIEW
contributes to neuronal morphogenesis. Hum Mol Gen 39 Smith NJ, Winstone AM, Stellitano L, et al. GM2
2013;22:2234–46. gangliosidosis in a UK study of children with progressive
20 Corbett MA, Schwake M, Bahlo M, et al. A mutation in the neurodegeneration: 73 cases reviewed. Dev Med Child Neurol
Golgi Qb-SNARE gene GOSR2 causes progressive myoclonus 2012;54:176–82.
epilepsy with early ataxia. AmJ Hum Gen 2011;88: 40 Kaback M, Lim-Steele J, Dabholkar D, et al. Tay-Sachs
657–63. disease--carrier screening, prenatal diagnosis, and the molecular
21 Boisse Lomax L, Bayly MA, Hjalgrim H, et al. ‘North Sea’ era. An international perspective, 1970 to 1993. The
progressive myoclonus epilepsy: phenotype of subjects with International TSD Data Collection Network. JAMA
GOSR2 mutation. Brain 2013;136:1146–54. 1993;270:2307–15.
22 DiMauro S, Hirano M. MERRF. In: Pagon RA, Adam MP, Bird 41 Hund E, Grau A, Fogel W, et al. Progressive cerebellar ataxia,
TD, Dolan CR, Fong CT, Stephens K, eds. GeneReviews. proximal neurogenic weakness and ocular motor disturbances:
Seattle, WA: University of Washington, 1993. hexosaminidase A deficiency with late clinical onset in four
23 Mancuso M, Orsucci D, Angelini C, et al. Myoclonus in siblings. J Neurol Sci 1997;145:25–31.
mitochondrial disorders. Mov Disord 2014;29:722. 42 Tajima A, Ohashi T, Hamano S, et al. Gaucher disease patient
24 Noer AS, Sudoyo H, Lertrit P, et al. A tRNA(Lys) mutation in with myoclonus epilepsy and a novel mutation. Pediatr Neurol
the mtDNA is the causal genetic lesion underlying myoclonic 2010;42:65–8.
epilepsy and ragged-red fiber (MERRF) syndrome. Am J 43 Sidransky E. Gaucher disease: insights from a rare Mendelian
Human Genet 1991;49:715–22. disorder. Discov Med 2012;14:273–81.
25 Hahn A, Schanzer A, Neubauer BA, et al. MERRF-like 44 Park JK, Orvisky E, Tayebi N, et al. Myoclonic epilepsy in
phenotype associated with a rare mitochondrial trnaile Gaucher disease: genotype-phenotype insights from a rare
mutation (m.4284 G>A). Neuropediatrics 2011;42:148–51. patient subgroup. Pediatr Res 2003;53:387–95.
26 Mole SE, Williams RE. Neuronal Ceroid-Lipofuscinoses. In: 45 Sinha S, Satishchandra P, Gayathri N, et al. Progressive
Pagon RA, Adam MP, Bird TD, Dolan CR, Fong CT, Stephens myoclonic epilepsy: a clinical, electrophysiological and
K, eds. GeneReviews. Seattle, WA: University of Washington, pathological study from South India. J Neurol Sci
1993. 2007;252:16–23.
27 Burneo JG, Arnold T, Palmer CA, et al. Adult-onset neuronal 46 Anderson LJ, Henley W, Wyatt KM, et al. Long-term
ceroid lipofuscinosis (Kufs disease) with autosomal dominant effectiveness of enzyme replacement therapy in adults with
inheritance in Alabama. Epilepsia 2003;44:841–6. Gaucher disease: results from the NCS-LSD cohort study.
28 Anderson GW, Goebel HH, Simonati A. Human pathology in J Inherit Metab Dis 2014;37:953–60.
NCL. Biochim Biophys Acta 2013;1832:1807–26. 47 Sundqvist A, Tomson T, Lundkvist B. Valproate as
29 Carpenter S, Karpati G, Andermann F, et al. The monotherapy for juvenile myoclonic epilepsy: dose-effect study.
ultrastructural characteristics of the abnormal cytosomes in Ther Drug Monit 1998;20:149–57.
Batten-Kufs’ disease. Brain 1977;100(Pt 1):137–56. 48 Pronicka E, Weglewska-Jurkiewicz A, Pronicki M, et al.
30 Mole SEWR. In: Pagon RA, Adam MP, Bird TD, et al., editors. Drug-resistant epilepsia and fulminant valproate liver toxicity.
GeneReviews™ [Internet]. Seattle, WA: University of Alpers-Huttenlocher syndrome in two children confirmed post
Washington, 1993–2014. http://www.ncbi.nlm.nih.gov/books/ mortem by identification of p.W748S mutation in POLG gene.
NBK1428/. 2001 Oct 10 [Updated 2013 Aug 1]. Med Sci Monit 2011;17:CR203–9.
31 Tsuji S. Dentatorubral-pallidoluysian atrophy. Handbook Clin 49 Iivanainen M, Himberg JJ. Valproate and clonazepam in the
Neurol 2012;103:587–94. treatment of severe progressive myoclonus epilepsy. Arch
32 Ikeuchi T, Onodera O, Oyake M, et al. Dentatorubral- Neurol 1982;39:236–8.
pallidoluysian atrophy (DRPLA): close correlation of CAG 50 Shahwan A, Farrell M, Delanty N. Progressive myoclonic
repeat expansions with the wide spectrum of clinical epilepsies: a review of genetic and therapeutic aspects. Lancet
presentations and prominent anticipation. Semin Cell Biol Neurol 2005;4:239–48.
1995;6:37–44. 51 Striano P, Belcastro V. Treatment of myoclonic seizures. Expert
33 Caciotti A, Di Rocco M, Filocamo M, et al. Type II sialidosis: Rev Neurother 2012;12:1411–17; quiz 8.
review of the clinical spectrum and identification of a new 52 Genton P, Gelisse P, Crespel A. Lack of efficacy and potential
splicing defect with chitotriosidase assessment in two patients. aggravation of myoclonus with lamotrigine in
J Neurol 2009;256:1911–15. Unverricht-Lundborg disease. Epilepsia 2006;47:2083–5.
34 Federico A, Cecio A, Battini GA, et al. Macular cherry-red spot 53 Crespel A, Genton P, Berramdane M, et al. Lamotrigine
and myoclonus syndrome. Juvenile form of sialidosis. J Neurol associated with exacerbation or de novo myoclonus in
Sci 1980;48:157–69. idiopathic generalized epilepsies. Neurology 2005;65:762–4.
35 Heroman JW, Rychwalski P, Barr CC. Cherry red spot in 54 Biraben A, Allain H, Scarabin JM, et al. Exacerbation of
sialidosis (mucolipidosis type I). Arch Ophthalmol juvenile myoclonic epilepsy with lamotrigine. Neurology
2008;126:270–1. 2000;55:1758.
36 Seyrantepe V, Poupetova H, Froissart R, et al. Molecular 55 Magaudda A, Gelisse P, Genton P. Antimyoclonic effect of
pathology of NEU1 gene in sialidosis. Hum Mutat levetiracetam in 13 patients with Unverricht-Lundborg disease:
2003;22:343–52. clinical observations. Epilepsia 2004;45:678–81.
37 Cordeiro P, Hechtman P, Kaplan F. The GM2 gangliosidoses 56 Demir CF, Ozdemir HH, Mungen B. Efficacy of topiramate as
databases: allelic variation at the HEXA, HEXB, and GM2A add-on therapy in two different types of progressive myoclonic
gene loci. Genet Med 2000;2:319–27. epilepsy. Acta Medica (Hradec Kralove) 2013;56:36–8.
38 Mahuran DJ. Biochemical consequences of mutations causing 57 Yoshimura I, Kaneko S, Yoshimura N, et al. Long-term
the GM2 gangliosidoses. Biochim Biophys Acta observations of two siblings with Lafora disease treated with
1999;1455:105–38. zonisamide. Epilepsy Res 2001;46:283–7.

Malek N, et al. Pract Neurol 2015;0:1–8. doi:10.1136/practneurol-2014-000994 7


REVIEW
58 Koskiniemi M, Van Vleymen B, Hakamies L, et al. Piracetam 60 Manninen O, Koskenkorva P, Lehtimaki KK, et al. White
relieves symptoms in progressive myoclonus epilepsy: matter degeneration with Unverricht-Lundborg progressive
a multicentre, randomised, double blind, crossover study myoclonus epilepsy: a translational diffusion-tensor imaging
comparing the efficacy and safety of three dosages of oral study in patients and cystatin B-deficient mice. Radiology
piracetam with placebo. J Neurol Neurosurg Psychiatry 2013;269:232–9.
1998;64:344–8. 61 Fujimoto A, Yamazoe T, Yokota T, et al. Clinical utility of vagus
59 Uthman BM, Reichl A. Progressive Myoclonic Epilepsies. Curr nerve stimulation for progressive myoclonic epilepsy. Seizure
Treat Options Neurol 2002;4:3–17. 2012;21:810–12.

8 Malek N, et al. Pract Neurol 2015;0:1–8. doi:10.1136/practneurol-2014-000994

You might also like