You are on page 1of 11

Received: 24 April 2023

| Accepted: 19 August 2023

DOI: 10.1002/epd2.20152

REVIEW ARTICLE

ILAE Genetics Literacy series: Progressive myoclonus


epilepsies

Jillian M. Cameron1 | Colin A. Ellis2 | Samuel F. Berkovic1 |


for the ILAE Genetics Commission* | the ILAE Genetic Literacy Task Force*

1
Epilepsy Research Centre, Department
of Medicine, University of Melbourne, Abstract
Austin Health, Melbourne, Victoria, Progressive Myoclonus Epilepsy (PME) is a rare epilepsy syndrome characterized
Australia
2
by the development of progressively worsening myoclonus, ataxia, and seizures.
Department of Neurology, University
of Pennsylvania Perelman School of
A molecular diagnosis can now be established in approximately 80% of individu-
Medicine, Philadelphia, Pennsylvania, als with PME. Almost fifty genetic causes of PME have now been established,
USA although some remain extremely rare. Herein, we provide a review of clinical
Correspondence phenotypes and genotypes of the more commonly encountered PMEs. Using
Samuel F. Berkovic, Epilepsy Research an illustrative case example, we describe appropriate clinical investigation and
Centre, 245 Burgundy St., Heidelberg
therapeutic strategies to guide the management of this often relentlessly progres-
3084, VIC, Australia.
Email: s.berkovic@unimelb.edu.au sive and devastating epilepsy syndrome. This manuscript in the Genetic Literacy
series maps to Learning Objective 1.2 of the ILAE Curriculum for Epileptology
Funding information
(Epileptic Disord. 2019;21:129).
American Academy of Neurology
Susan S. Spencer Clinical Research
Training Scholarship; Mirowski Family KEYWORD
Foundation; National Health and progressive myoclonus epilepsies
Medical Research Council, Grant/
Award Number: APP1196637; National
Institute of Neurological Disorders
and Stroke, Grant/Award Number:
K23NS121520

1 | P RO G RE SSIVE M YOCLON US is often in later childhood or adolescence and patients


EPILEPSY typically become extremely functionally impaired by
their symptoms. As the name suggests, myoclonus is core
Progressive Myoclonus Epilepsy (PME) is a rare general- to the PME phenotype. Stimulus-­sensitive myoclonus
ized epilepsy syndrome, clinically characterized by the de- (also referred to as reflex myoclonus, that is myoclonus
velopment of progressively worsening myoclonus, ataxia, induced or exacerbated by a variety of stimuli including
and tonic–­clonic seizures,1 with variable associated cog- light, sound, touch, and action) is a key manifestation.
nitive decline and neuropsychiatric disturbance.2,3 Onset Action-­induced myoclonus is a characteristic feature and

*The ILAE Genetics Commission and The ILAE Genetic Literacy Task Force members are listed in Appendix 1.
This report was written by experts selected by the International League Against Epilepsy (ILAE) and was approved for publication by the ILAE.
Opinions expressed by the authors, however, do not necessarily represent the policy or position of the ILAE.

This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium, provided
the original work is properly cited.
© 2023 The Authors. Epileptic Disorders published by Wiley Periodicals LLC on behalf of International League Against Epilepsy.

670 | wileyonlinelibrary.com/journal/epd2
 Epileptic Disorders. 2023;25:670–680.
|

19506945, 2023, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/epd2.20152, Wiley Online Library on [02/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
CAMERON et al.    671

is typically fragmentary and multifocal. Myoclonus tends


to be refractory to treatment and cause the most function- Key points
ally significant impairment.4
There are many forms of PME, with now almost fifty • In the evaluation of PME, certain clinical and
established genetic causes, although some are extremely EEG findings can help narrow the list of differ-
rare (see Table S15). The most common cause of PME ential diagnoses to specific diseases.
worldwide is Unverricht-­Lundborg Disease (ULD),6 with • Important PMEs include Unverricht-­Lundborg
highest reported prevalence in Finland (1.9/100 000).7 Disease, Lafora Disease, Neuronal Ceroid
Other important causes of PME include Lafora Disease Lipofuscinoses, and Myoclonic Epilepsy with
(LD, prevalence 1–­9/106,8), the Neuronal Ceroid Lipofus- Ragged Red Fibers.
cinoses (NCL), and Myoclonic epilepsy with ragged red fi- • A molecular diagnosis can now be established
bers (MERRF). Geographical distribution varies for some in approximately 80% of patients with PME.
forms of PME, particularly those with recessive inheri- While PME gene panels are currently useful,
tance, dependent on the frequency of consanguinity and exome or genome sequencing will likely sup-
local founder effects. plant these in the future.
• Treatment mainly symptomatic.

2 | ET I O LOGY AN D G E N ET ICS OF
PME
myoclonus, generalized tonic–­clonic seizures (GTCS)
A molecular diagnosis can now be established in approxi- are the most common seizure type, with absence and
mately 80% of patients with PME.9,10 Clinical phenotypes focal seizures with impaired awareness reported rarely.4
and genotypes associated with some of the more common GTCS typically respond to antiseizure medication, but
forms of PME are outlined below. also independently decrease in frequency as the disease
progresses.17 The most commonly associated neurological
manifestation is ataxia. Cognition is relatively preserved,
2.1 | Unverricht-­Lundborg disease although subtle impaired processing and executive func-
tion have been reported.18,19 Psychiatric co-­morbidities are
ULD is an autosomal recessive condition due to ho- common, with high rates of depression and suicidal be-
mozygous or compound heterozygous pathogenic haviour.18 Reduced life expectancy is seen, with a median
variants in CSTB.6 The most common type of genetic age at death of 53.9 years.7
variant, seen in 90% of disease alleles worldwide, is an
unstable expansion of a 12-­nucleotide dodecamer repeat
(5′-­CCCCGCCCCGCG-­3′).11 ULD-­associated alleles 2.2 | Lafora disease
typically contain at least 30 repeat copies.12 Although
initial small studies did not show a correlation between Lafora Disease is an autosomal recessive condition due
the expansion size and clinical features, a larger study to loss-­of-­function variants in EPM2A (65%–­70% of cases)
suggested that the size of the CSTB expansion is likely or NHLRC1/EPM2B.20 No definitive genotype–­phenotype
to have a modulating effect on age of disease onset, se- correlations have been established, though a longer life
verity of myoclonus, and neurophysiological markers.13 expectancy and milder disease course have been described
Anticipation (that is increase in expansion from parent with some NHLRC1 variants, particularly the recurrent
to child) has not been identified. Missense pathogenic missense variant p.Asp146Asn.21,22
variants in CSTB account for a minority of causative EPM2A encodes laforin23 and NHLRC1 encodes
variants.14 Individuals who are compound heterozy- malin.24 Whilst their roles are incompletely understood,
gotes, with one missense and one repeat expansion vari- both proteins are involved in glycogen metabolism. Evi-
ant, have been reported to have a more severe disease dence suggests they act as a complex to prevent the accu-
phenotype.15,16 mulation of insoluble glycogen.25–­27 LD is associated with
Typical onset of ULD is in late childhood to adolescence the pathognomonic finding of Lafora bodies (diastase-­
(peak 12–­13 years of age). Myoclonus is the predominant resistant periodic acid–­Schiff (PAS-­D)-­positive aggregates
feature at onset, progressing over months to years before of abnormally branched, insoluble polyglucosans), which
plateauing in middle life (17). Medication refractory reflex are found in the brain, liver, skeletal and cardiac myocytes
myoclonus, particularly action myoclonus, is common and sweat glands. It is thought a deficiency of functional
and the main cause of functional disability. Other than laforin or malin results in normally soluble glycogen
|

19506945, 2023, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/epd2.20152, Wiley Online Library on [02/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
672    CAMERON et al.

containing abnormally long chains and precipitating and 2.4 | Myoclonic epilepsy and ataxia due
aggregating as Lafora bodies.28 to KCNC1 mutation (MEAK)
Typical onset of LD is in late childhood or early ad-
olescence (8–­19 years, peak at 14–­16 years). Focal oc- MEAK is a form of PME caused by a recurrent het-
cipital seizures are a characteristic feature particularly erozygous missense variant, p.Arg320His in KCNC1.
early in disease.29 Myoclonus manifests early and es- Most cases are sporadic due to de novo variants but a
calates rapidly over months to years to intractable ac- few families with autosomal dominant inheritance
tion-­ and stimulus-­sensitive myoclonus. Various other are reported.35 KCNC1 encodes Kv3.1, a subunit of the
seizure types (GTCS, absence, atonic) rapidly develop. Kv3 subfamily of voltage-­gated tetrameric potassium
LD is associated with a rapidly progressive dementia channels.35 The p.Arg320His variant has a dominant-­
with significant apraxia, visual loss, and neuropsychi- negative loss-­of-­function effect in vitro.35 Kv3.1 expres-
atric disturbance. The prognosis is poor. Early studies sion is largely restricted to the central nervous system,
suggested death occurred within 10 years of disease with predominant expression in GABAergic interneu-
onset,28,30 but more recent data suggests the prognosis rons.36 Thus, disinhibition due to impaired activity of
may be less grim, with a median survival of 11 years.31 these inhibitory interneurons is a plausible pathogenic
Late onset (>18 years) appears to be related to longer mechanism.35
disease duration and slower progression.31 The clinical phenotype is characterized by the onset of
myoclonus between 6 and 14 years of age.8 Myoclonus is
the most prominent clinical feature, typically becoming
2.3 | Neuronal ceroid lipofuscinoses very severe during adolescence and impacting mobility.
(NCL) Infrequent GTCS are seen, and cognition is largely pre-
served. Early death has not been observed.
The NCLs, also known as Batten Disease, are a group
of monogenic neurodegenerative conditions grouped
together due to a shared histopathological signature, 2.5 | Myoclonic epilepsy associated with
namely abnormal intracellular lysosomal lipopigment ragged red fibers (MERRF)
storage material with various characteristic forms on
ultrastructural examination of brain and peripheral tis- MERRF is one of many mitochondrial disorders due to
sues such as skin or lymphocytes. To date, 13 different pathogenic variants in mitochondrial DNA. Mitochon-
NCL-­related genes have been identified. Although some drial DNA encodes polypeptides involved in the mito-
clinical heterogeneity is seen, broadly the clinical phe- chondrial respiratory chain. Almost all reported MERRF
notype includes a combination of seizures, visual failure, pathogenic variants affect the gene for tRNA lysine MT-­
dementia, and decline in motor function. Most forms of TK, with the m.8344A>G variant identified in 80%–­90%
NCL have onset in infancy or early childhood. Important of patients.37
childhood forms of NCL that can present with a PME MERRF is characterized by myoclonus, generalized
phenotype include CLN2 disease, CLN5 disease, and the seizures, myopathy, and progressive ataxia. Onset is
late-­infantile form of CLN6 disease. CLN2 disease, due typically in childhood, although adult onset is seen. As
to pathogenic variants in TPP1, is associated with visual is typical of mitochondrial disorders, the phenotype is
loss and significant language delay. 32 It is the only form extremely variable, and multisystem involvement (par-
of PME for which there is disease-­modifying treatment, ticularly affecting tissues with high metabolic demand)
hence prompt recognition and diagnosis is of critical im- is seen.38 Common associated features include hearing
portance (see treatment section below). KCTD7 patho- impairment, sensorimotor peripheral neuropathy, cog-
genic variants have been described in individuals with a nitive impairment, short stature, and optic atrophy.
PME phenotype both with and without histopathologi- Less common features include cardiomyopathy, diabe-
cal evidence of abnormal lysosomal storage material on tes mellitus, pyramidal signs, retinitis pigmentosa, and
biopsy.33,34 ophthalmoplegia. Multiple lipomas, ranging from small
A PME phenotype can also be associated with the re- subcutaneous nodules to large masses, can be seen, typi-
cessive adult-­onset NCLs (also referred to as Kufs disease), cally on the neck or trunk.38,39 There is some phenotypic
particularly CLN6 disease. Vision is typically preserved in overlap with other mitochondrial disorders, particularly
the adult-­onset NCLs. CLN4 disease, due to pathogenic Mitochondrial Encephalopathy, Lactic Acidosis, and
variants in DNAJC5, is the only autosomal dominant form Stroke-­like episodes (MELAS). Although acute episodes
of NCL described, and also presents with an adult-­onset of neurological impairment are the most common neu-
PME phenotype. rological presentation associated with MELAS, a broad
|

19506945, 2023, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/epd2.20152, Wiley Online Library on [02/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
CAMERON et al.    673

range of neurological manifestations including PME are pathways involved are diverse and constantly expanding,
well-­recognized. as highlighted by the recent novel association of dolichol-­
dependent glycosylation with the PME phenotype due
to pathogenic variants impacting NUS1, DHDDS, and
2.6 | Sialidosis ALG10.9 There are increasing descriptions of cases of PME
due to pathogenic variants in genes associated with other
Sialidosis is an autosomal recessive lysosomal storage neurodevelopmental conditions, broadening the spectrum
disorder due to pathogenic variants in NEU1, resulting of clinical phenotypes associated with these genes. This
in sialidase deficiency and abnormal accumulation of includes but is not limited to the developmental and epi-
sialic acid-­rich substrates in the CNS and other organ leptic encephalopathies (for example, TBC1D24, DHDDS,
systems.40 There are two forms of sialidosis, type I (nor- CHD2, and CACNA2D2).
momorphic) and II (dysmorphic). Patients with type I
sialidosis have some residual sialidase activity, and ex-
hibit a PME phenotype characterized by progressively 5 | THE UNSOLVED RESIDUUM
worsening multifocal myoclonus with seizures and
ataxia. Typical age of onset is between 10 and 20 years. Despite the rapidly evolving understanding of the genetic
A characteristic macular “cherry-­red spot” can be de- architecture of PME, 20% of individuals remain without a
tected, due to storage material in perifoveal ganglionic molecular diagnosis. This unsolved residuum will likely
cells, and can result in visual impairment.41 Early in the be a collection of ultra-­rare causes in novel genes, or due
disease, the cherry-­red spot may be clinically undetect- to pathogenic variants, which are more challenging to
able, and it may also disappear in the later stages of the identify and interpret with current available technologies,
disease.42 such as repeat expansion mutations, or variants in non-
coding regions affecting mechanisms such as transcrip-
tional regulation or epigenetic modification. Nongenetic
3 | F URT HE R FOR M S OF PM E etiologies, including autoimmune, may warrant consider-
ation, as highlighted by the association of coeliac disease
Other well-­described forms of PME include Action with various neurological manifestations including myo-
Myoclonus Renal Failure Syndrome (AMRF, SCARB2), clonus and ataxia.44,45
GOSR2 “North Sea” PME, and Spinomuscular Atrophy-­ Of note, the Progressive Myoclonic Ataxias (PMAs)
PME (SMA-­PME, ASAH1) amongst others. The most are defined as conditions generally presenting first with
distinguishing (but not universal) feature of AMRF is prominent ataxia, before subsequently developing my-
renal involvement, initially presenting as proteinuria oclonus and in some cases epilepsy.46 There is clearly a
and then progressing to nephrotic syndrome and end-­ significant clinical phenotypic overlap between PME and
stage renal failure. “North-­Sea” PME is so-­called as all PMA, and several conditions are described under both cat-
described cases have birthplaces clustered around the egories in the literature. An awareness of this when con-
North Sea, a result of a founder effect. A persistently sidering differential diagnoses and potential underlying
raised CK in the context of a normal muscle biopsy is etiologies in those who remain without a genetic diagno-
typical, and associated clinical features include scoliosis sis is important.
and peripheral neuropathy. SMA-­PME is a rare condi-
tion characterized by the presence of both lower motor
neuron degeneration and classical PME features; either 6 | INV ESTIGATION STRATEGIE S
can occur first.43 Furthermore, a PME phenotype can
also be seen as part of a wider neurological spectrum 6.1 | History and examination
of disease in various conditions including Dentatorubral
pallidoluysian atrophy (DRPLA), Gaucher's Disease, PME should be considered in patients with myoclonus
and Juvenile Huntington's Disease. (especially if refractory to medication), progressive motor
impairment, cognitive deterioration, sensory, and cerebel-
lar signs. Because most genetic causes are recessive, fam-
4 | R A RE A N D E ME RG IN G FORMS ily history may be unrevealing, or may include affected
siblings only. Inquiring about consanguinity is essential.
With now almost fifty genes associated with PME, Some features of history and examination may suggest
many forms remain extremely rare. The biochemical specific PME syndromes (Table 1).
|

19506945, 2023, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/epd2.20152, Wiley Online Library on [02/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
674    CAMERON et al.

T A B L E 1 History and examination features suggestive of


specific PME syndromes.

Finding Associated syndrome


Visual seizures (history) Lafora disease
Occipital seizures (EEG)
Vision loss, retinopathy NCLs (but rarely in adult-­onset
cases)
Sialidosis
Cherry-­red spot in macula Sialidosis (also Niemann–­Pick
disease, Gaucher disease)
Myopathy MERRF
Hearing loss
Cutaneous lipomas
Stroke-­like attacks MELAS
Hepatosplenomegaly Gaucher disease
Peripheral neuropathy Multiple PME syndromes,
Ataxia nonspecific
Optic atrophy
FIGURE 1 Diagnostic approach to suspected PME.
Photosensitivity (at low
flash rate)
Psychiatric symptoms
targeted single-­gene testing based on phenotype. Gene
panels for PME or comprehensive epilepsy panels are
6.2 | EEG and imaging available from most commercial laboratories. Exome or
genome sequencing have additional advantages and are
EEG should be performed in all patients. It is important replacing gene panels as first-­line genetic tests because
to recognize that there can be considerable variabil- they can detect findings not detected by a gene panel and
ity in EEG characteristics between the various PMEs. enable future reanalysis for undiagnosed cases. Many di-
In most, but not all, forms of PME, the background is agnostic laboratories now offer a gene panel off an exome
typically normal early in the disease course, progressing or genome backbone, which allows for reanalysis with an
to irregular diffuse slowing as the disease progresses. updated gene list after 18 months–­2 years.
Epileptiform discharges are typically generalized spike-­ Clinicians should be aware of several pitfalls in genetic
and-­wave or polyspike-­and-­wave, but focal or multifocal testing for PMEs. Repeat expansions (as seen in ULD
epileptiform activity has also been reported in several and DRPLA) are typically not captured well by current
different PME subtypes.47,48 Occipital discharges and/ sequencing-­based tests (including gene panels and exome
or seizures are often seen in PME, particularly in La- sequencing) and may require specialized PCR testing or
fora disease.28 Photosensitivity is common in many of Southern blot, or specific bioinformatics added to genome
the PMEs.49 In particular, photosensitivity at low flash sequencing to detect expansion variants. Thus the ability
frequencies (<6 Hz) should raise suspicion for a PME, of a test to cover these expansion variants should be dis-
although it is not entirely specific for PME nor for an cussed with the diagnostic laboratory. An additional pit-
individual PME subtype.50,51 Neuroimaging in patients fall of gene panel or exome sequencing is that sequencing
with PME is typically normal or shows nonspecific of mitochondrial DNA is not standard. It is important that
atrophy.52 clinicians are aware of this, though typically mitochon-
drial DNA testing is only appropriate to include as a first-­
line genetic test if there are specific clinical features, which
6.3 | Genetic testing suggest an underlying mitochondrial pathology. Whole
genome sequencing has the advantage of being able to
Genetic testing should now be the first-­line diagnostic test cover both expansion variants and mitochondrial variants,
whenever PME is suspected (Figure 1), and should be per- as well as complex structural and noncoding variants, and
formed early given the profound prognostic implications so is likely to become the test of choice for PME as costs of
of the diagnosis and emerging targeted therapies. Genetic WGS come down and availability increases.
testing for PME is now widely available, noninvasive, and Many PMEs are autosomal recessive. This is import-
high-­yield. The long and growing list of causative genes ant when interpreting the results of genetic testing. For
(Table S1) warrants a broad testing strategy, rather than recessive disorders, a single heterozygous pathogenic
|

19506945, 2023, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/epd2.20152, Wiley Online Library on [02/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
CAMERON et al.    675

variant is not sufficient to cause disease; such a patient assays are ordered and performed individually, and sys-
is an unaffected carrier. On the other hand, when a par- tematic approaches to the selection of tests are limited.58
ticular syndrome is strongly suspected but only a single
heterozygous pathogenic variant is detected, an occult
second pathogenic variant may exist, but not yet have 6.4.2 | Neurophysiology
been detected by the testing which has been ordered to
date.53 This means that broadening the testing, for ex- Giant evoked potentials in response to both visual and
ample, to whole genome sequencing, which can detect a somatosensory stimulation have been reported in several
wider variety of genomic mechanisms, may be appropri- PMEs.48,59,60 Although the diagnostic utility of this finding
ate, and discussion with a clinical genetics team can be has not been studied systematically, this is an accessible
helpful to discuss the next best steps. When two different and noninvasive test, which can be used to demonstrate
pathogenic variants are identified in the same gene (com- cortical hyperexcitability in individuals with PME. Nerve
pound heterozygous), it is necessary to confirm that the conduction and EMG studies may be indicated to evalu-
two variants are on different alleles (“in trans”), that is ate specific comorbid features such as neuropathy or
one inherited from each parent, rather than on the same myopathy.
allele (“in cis”). This can be checked by segregating the
variants in both parents, with the anticipation that each
will carry one of the two variants found in the patient, 6.4.3 | Tissue biopsies
although very occasionally one variant may be de novo
and not inherited. Finally, variants of uncertain signif- Before the era of readily available molecular testing, tissue-­
icance (VUS) must be interpreted with caution, and in based diagnosis was a standard part of the diagnostic evalu-
most cases should not be considered diagnostic nor used ation of PME.57,61 Today, biopsies are not necessary in cases
to guide management. Discussion with a genetics team with a clear genetic diagnosis and compatible phenotype.
can be helpful to consider if further options, such as func- Biopsies may be useful in selected patients with nondiag-
tional studies, may be possible to help further investigate nostic genetic testing. Tissue diagnosis pertains mostly to
VUS. Reanalysis after time with updated knowledge and LD and the NCLs. The pathognomonic Lafora bodies seen
gene lists is also helpful for families without a confirmed in LD can be identified on axillary skin biopsy, though the
molecular diagnosis. sensitivity and specificity are not known, and are likely to
vary across institutions based on local experience.
Similarly, the sensitivity and specificity of biopsy for
6.4 | Ancillary tests NCL are unknown. In adult-­onset NCL, intracellular stor-
age material may be absent in peripheral tissues (false
In the past, the evaluation of suspected PME often in- negatives), while normal accumulation of age-­related
cluded an assortment of specialized tests, such as enzyme lipofuscin may be misinterpreted as pathological (false
assays and tissue biopsies. In the modern genetic era, these positives).62 When a mitochondrial disorder is suspected,
should be considered ancillary, and may not be necessary muscle biopsy may show ragged red fibers. Again, the
If genetic testing is diagnostic. Ancillary testing still plays sensitivity and specificity of this test are unknown, and
a role when an orthogonal test may be helpful to support a muscle biopsy is now primarily reserved for patients with
genetic diagnosis, for example, to help determine the sig- nondiagnostic genetic testing.
nificance of a VUS.

7 | TREATMENT STRATEGIES IN
6.4.1 | Laboratory tests PME

Some common laboratory tests may suggest particular Management of patients with PME presents several chal-
PME syndromes. Renal impairment is often a feature of lenges. For most types of PME, there are no available
AMRF.54 Elevated CK has been associated with North-­Sea disease-­modifying therapies and treatment strategies are
PME, despite normal muscle biopsies.55 Elevated lactate symptomatic, aimed at seizure control and suppression of
may suggest a mitochondrial disorder, but is neither sensi- myoclonus. The rarity of the condition means evidence-­
tive nor specific. based efficacy data is minimal, and is largely in small pop-
Enzyme assays have been developed for several of the ulations of ULD or LD if at all.
NCLs caused by lysosomal enzyme deficiencies.56–­58 Urine Antiseizure medications (ASMs) are the current main-
sialo-­oligosaccharides are elevated in sialidosis. These stay of treatment. Valproate was the initial ASM shown to
|

19506945, 2023, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/epd2.20152, Wiley Online Library on [02/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
676    CAMERON et al.

be effective in convulsive seizure and myoclonus control 7.3 | Gene therapies


in ULD.63 Benzodiazepines, including clonazepam and
clobazam, have well-­established efficacy for the manage- Gene and nucleotide-­based therapies are not yet clinically
ment of myoclonus, and are typically used as an add-­on available for any of the PMEs. In mouse models of CLN6
therapy. More recently several ASMs including levetirace- disease, bilateral intracerebroventricular injections of
tam, topiramate, zonisamide, and brivaracetam have been adeno-­associated viral 6 carrying functional copies of CLN6
reported to be effective in PME. Piracetam is a useful an- have been shown to increase lifespan and reduce neuro-
timyoclonic agent,64 and perampanel has been associated pathological features.72 Antisense oligonucleotide (ASO)
with a beneficial effect on action myoclonus, disability, therapy has been shown both in vitro and in vivo to have
and seizures in PME.40 beneficial impacts. Patient-­derived fibroblasts from an in-
dividual with ULD homozygous for the c.66G>A CSTB
pathogenic variant expressed normal CSTB protein levels
7.1 | Adjunctive therapies when treated with a specific locked nucleic acid ASO target-
ing the cryptic donor splice site in intron 1 of CSTB.73 Such
Even when used in combination, ASMs typically fail to precision medicine therapeutic strategies are clearly still in
adequately control the complex and progressive symp- their initial stages but hold promise that future improved
tomatology experienced by patients with PME, and targeted therapies will be feasible for patients with PME.
consideration needs to be given to adjunctive thera-
pies. There is limited data exploring dietary therapeutic ACKNOWLEDGMENTS
strategies in PME, with only small case series of the ke- CAE is supported by the National Institute of Neurologi-
togenic diet and modified Atkins diet in LD and North cal Disorders and Stroke award number K23NS121520;
Sea PME, respectively, showing some improvement in the American Academy of Neurology Susan S. Spencer
myoclonus.65,66 Similarly, limited case reports and case Clinical Research Training Scholarship and the Mirowski
series of neuromodulatory therapies including VNS, Family Foundation. SFB is supported by an NHMRC In-
DBS and rTMS have variable results, and all warrant vestigator Grant (APP1196637). Open access publishing
further exploration. With the increasing appreciation facilitated by The University of Melbourne, as part of the
of the role of neuroinflammation in neurodegenerative Wiley -­ The University of Melbourne agreement via the
disorders, immunomodulatory medications may also Council of Australian University Librarians.
have a therapeutic role, though this is yet to be estab-
lished.67,68 Animal data suggests that metformin may ORCID
have a disease-­modifying role in Lafora disease,69,70 Colin A. Ellis https://orcid.org/0000-0003-2152-8106
though the very limited clinical data that is currently Samuel F. Berkovic https://orcid.
available suggests that its use is not associated with clin- org/0000-0003-4580-841X
ically meaningful benefit.71 Further data is required to
clarify this. REFERENCES
1. Blumcke I, Arzimanoglou A, Beniczky S, Wiebe S. Roadmap for
a competency-­based educational curriculum in epileptology: re-
7.2 | Enzyme replacement therapies port of the Epilepsy Education Task Force of the International
League Against Epilepsy. Epileptic Disord. 2019;21(2):129–­40.
2. Marseille Consensus Group. Classification of progressive
Enzyme replacement is currently available for treatment
myoclonus epilepsies and related disorders. Ann Neurol.
of CLN2 disease, caused by pathogenic variants in the 1990;28(1):113–­6.
gene TPP1, leading to a deficiency in the lysosomal en- 3. Berg AT, Berkovic SF, Brodie MJ, Buchhalter J, Cross JH,
zyme tripeptidyl peptidase 1. Enzyme replacement ther- van Emde BW, et al. Revised terminology and concepts for
apy via intraventricular infusion of recombinant human organization of seizures and epilepsies: report of the ILAE
tripeptidyl peptidase 1 (cerliponase alfa) results in less Commission on Classification and Terminology, 2005-­2009.
decline in motor and language function that in historical Epilepsia. 2010;51(4):676–­85.
4. Kalviainen R, Khyuppenen J, Koskenkorva P, Eriksson K,
controls.57 This is the first disease-­modifying treatment
Vanninen R, Mervaala E. Clinical picture of EPM1-­Unverricht-­
for a type of PME and holds the promise that similar ap-
Lundborg disease. Epilepsia. 2008;49(4):549–­56.
proaches may be successful in other forms of PME. Sev- 5. Oliver KL, Scheffer IE, Bennett MF, Grinton BE, Bahlo M,
eral PME-­causing pathogenic variants result in enzyme Berkovic SF. Genes4Epilepsy: an epilepsy gene resource.
deficiencies (including Sialidosis, SMA-­PME, and Gau- Epilepsia. 2023;64:1368–­75.
cher's disease), and may be good targets for enzyme re- 6. Pennacchio LA, Lehesjoki AE, Stone NE, Willour VL,
placement therapies. Virtaneva K, Miao J, et al. Mutations in the gene encoding
|

19506945, 2023, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/epd2.20152, Wiley Online Library on [02/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
CAMERON et al.    677

cystatin B in progressive myoclonus epilepsy (EPM1). Science. 22. Gomez-­Abad C, Gomez-­Garre P, Gutierrez-­Delicado E, Saygi
1996;271(5256):1731–­4. S, Michelucci R, Tassinari CA, et al. Lafora disease due to
7. Sipila JOT, Hypponen J, Kyto V, Kalviainen R. Unverricht-­ EPM2B mutations: a clinical and genetic study. Neurology.
Lundborg disease (EPM1) in Finland: a nationwide population-­ 2005;64(6):982–­6.
based study. Neurology. 2020;95(23):e3117–­23. 23. Minassian BA, Lee JR, Herbrick JA, Huizenga J, Soder S,
8. Oliver KL, Franceschetti S, Milligan CJ, Muona M, Mandelstam Mungall AJ, et al. Mutations in a gene encoding a novel protein
SA, Canafoglia L, et al. Myoclonus epilepsy and ataxia due to tyrosine phosphatase cause progressive myoclonus epilepsy.
KCNC1 mutation: analysis of 20 cases and K(+) channel prop- Nat Genet. 1998;20(2):171–­4.
erties. Ann Neurol. 2017;81(5):677–­89. 24. Chan EM, Omer S, Ahmed M, Bridges LR, Bennett C, Scherer SW,
9. Courage C, Oliver KL, Park EJ, Cameron JM, Grabinska KA, et al. Progressive myoclonus epilepsy with polyglucosans (Lafora
Muona M, et al. Progressive myoclonus epilepsies-­residual disease): evidence for a third locus. Neurology. 2004;63(3):565–­7.
unsolved cases have marked genetic heterogeneity including 25. Worby CA, Gentry MS, Dixon JE. Malin decreases glycogen ac-
dolichol-­dependent protein glycosylation pathway genes. Am J cumulation by promoting the degradation of protein targeting
Hum Genet. 2021;108(4):722–­38. to glycogen (PTG). J Biol Chem. 2008;283(7):4069–­76.
10. Canafoglia L, Franceschetti S, Gambardella A, Striano P, 26. Vilchez D, Ros S, Cifuentes D, Pujadas L, Valles J, Garcia-­Fojeda
Giallonardo AT, Tinuper P, et al. Progressive myoclonus epilep- B, et al. Mechanism suppressing glycogen synthesis in neurons
sies: diagnostic yield with next-­generation sequencing in previ- and its demise in progressive myoclonus epilepsy. Nat Neurosci.
ously unsolved cases. Neurol Genet. 2021;7(6):e641. 2007;10(11):1407–­13.
11. Lalioti MD, Scott HS, Buresi C, Rossier C, Bottani A, Morris 27. Sanchez-­Martin P, Roma-­Mateo C, Viana R, Sanz P. Ubiquitin
MA, et al. Dodecamer repeat expansion in cystatin B gene conjugating enzyme E2-­N and sequestosome-­1 (p62) are com-
in progressive myoclonus epilepsy. Nature. 1997;386(6627):​ ponents of the ubiquitination process mediated by the Malin-­
847–­51. laforin E3-­ubiquitin ligase complex. Int J Biochem Cell Biol.
12. Lafreniere RG, Rochefort DL, Chretien N, Rommens JM, 2015;69:204–­14.
Cochius JI, Kalviainen R, et al. Unstable insertion in the 5′ 28. Nitschke F, Ahonen SJ, Nitschke S, Mitra S, Minassian BA.
flanking region of the cystatin B gene is the most common Lafora disease –­ from pathogenesis to treatment strategies. Nat
mutation in progressive myoclonus epilepsy type 1, EPM1. Nat Rev Neurol. 2018;14(10):606–­17.
Genet. 1997;15(3):298–­302. 29. Roger J, Pellissier JF, Bureau M, Dravet C, Revol M, Tinuper
13. Hypponen J, Aikia M, Joensuu T, Julkunen P, Danner N, P. Early diagnosis of Lafora disease. Significance of paroxys-
Koskenkorva P, et al. Refining the phenotype of Unverricht-­ mal visual manifestations and contribution of skin biopsy. Rev
Lundborg disease (EPM1): a population-­wide Finnish study. Neurol (Paris). 1983;139(2):115–­24.
Neurology. 2015;84(15):1529–­36. 30. Turnbull J, Tiberia E, Striano P, Genton P, Carpenter S, Ackerley
14. Joensuu T, Lehesjoki AE, Kopra O. Molecular back- CA, et al. Lafora disease. Epileptic Disord. 2016;18(S2):38–­62.
ground of EPM1-­Unverricht-­Lundborg disease. Epilepsia. 31. Pondrelli F, Muccioli L, Licchetta L, Mostacci B, Zenesini C,
2008;49(4):557–­63. Tinuper P, et al. Natural history of Lafora disease: a prognos-
15. Koskenkorva P, Hypponen J, Aikia M, Mervaala E, Kiviranta T, tic systematic review and individual participant data meta-­
Eriksson K, et al. Severer phenotype in Unverricht-­Lundborg analysis. Orphanet J Rare Dis. 2021;16(1):362.
disease (EPM1) patients compound heterozygous for the do- 32. Fietz M, AlSayed M, Burke D, Cohen-­Pfeffer J, Cooper JD,
decamer repeat expansion and the c.202C>T mutation in the Dvorakova L, et al. Diagnosis of neuronal ceroid lipofuscinosis
CSTB gene. Neurodegener Dis. 2011;8(6):515–­22. type 2 (CLN2 disease): expert recommendations for early detection
16. Canafoglia L, Gennaro E, Capovilla G, Gobbi G, Boni A, and laboratory diagnosis. Mol Genet Metab. 2016;119(1–­2):160–­7.
Beccaria F, et al. Electroclinical presentation and genotype-­ 33. Staropoli JF, Karaa A, Lim ET, Kirby A, Elbalalesy N, Romansky
phenotype relationships in patients with Unverricht-­Lundborg SG, et al. A homozygous mutation in KCTD7 links neuronal
disease carrying compound heterozygous CSTB point and indel ceroid lipofuscinosis to the ubiquitin-­proteasome system. Am J
mutations. Epilepsia. 2012;53(12):2120–­7. Hum Genet. 2012;91(1):202–­8.
17. Magaudda A, Ferlazzo E, Nguyen VH, Genton P. Unverricht-­ 34. Kousi M, Anttila V, Schulz A, Calafato S, Jakkula E, Riesch E,
Lundborg disease, a condition with self-­limited progression: et al. Novel mutations consolidate KCTD7 as a progressive my-
long-­term follow-­up of 20 patients. Epilepsia. 2006;47(5):860–­6. oclonus epilepsy gene. J Med Genet. 2012;49(6):391–­9.
18. Ferlazzo E, Gagliano A, Calarese T, Magaudda A, Striano P, 35. Muona M, Berkovic SF, Dibbens LM, Oliver KL, Maljevic S,
Cortese L, et al. Neuropsychological findings in patients with Bayly MA, et al. A recurrent de novo mutation in KCNC1 causes
Unverricht-­Lundborg disease. Epilepsy Behav. 2009;14(3):545–­9. progressive myoclonus epilepsy. Nat Genet. 2015;47(1):39–­46.
19. Aikia M, Hypponen J, Mervaala E, Kalviainen R. Cognitive func- 36. Gan L, Kaczmarek LK. When, where, and how much?
tioning in progressive myoclonus epilepsy type 1 (Unverricht-­ Expression of the Kv3.1 potassium channel in high-­frequency
Lundborg disease, EPM1). Epilepsy Behav. 2021;122:108157. firing neurons. J Neurobiol. 1998;37(1):69–­79.
20. Singh S, Ganesh S. Lafora progressive myoclonus epilepsy: 37. Enriquez JA, Chomyn A, Attardi G. MtDNA mutation in
a meta-­analysis of reported mutations in the first decade fol- MERRF syndrome causes defective aminoacylation of tR-
lowing the discovery of the EPM2A and NHLRC1 genes. Hum NA(Lys) and premature translation termination. Nat Genet.
Mutat. 2009;30(5):715–­23. 1995;10(1):47–­55.
21. Baykan B, Striano P, Gianotti S, Bebek N, Gennaro E, Gurses 38. Mancuso M, Orsucci D, Angelini C, Bertini E, Carelli V, Comi
C, et al. Late-­onset and slow-­progressing Lafora disease in four GP, et al. Phenotypic heterogeneity of the 8344A>G mtDNA
siblings with EPM2B mutation. Epilepsia. 2005;46(10):1695–­7. "MERRF" mutation. Neurology. 2013;80(22):2049–­54.
|

19506945, 2023, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/epd2.20152, Wiley Online Library on [02/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
678    CAMERON et al.

39. Altmann J, Buchner B, Nadaj-­Pakleza A, Schafer J, Jackson 55. Boisse Lomax L, Bayly MA, Hjalgrim H, Moller RS, Vlaar
S, Lehmann D, et al. Expanded phenotypic spectrum of the AM, Aaberg KM, et al. 'North Sea' progressive myoclonus ep-
m.8344A>G "MERRF" mutation: data from the German mi- ilepsy: phenotype of subjects with GOSR2 mutation. Brain.
toNET registry. J Neurol. 2016;263(5):961–­72. 2013;136(Pt 4):1146–­54.
40. Assenza G, Nocerino C, Tombini M, Di Gennaro G, D'Aniello 56. Mole SE, Anderson G, Band HA, Berkovic SF, Cooper JD,
A, Verrotti A, et al. Perampanel improves cortical myoclonus Kleine Holthaus SM, et al. Clinical challenges and future ther-
and disability in progressive myoclonic epilepsies: a case se- apeutic approaches for neuronal ceroid lipofuscinosis. Lancet
ries and a systematic review of the literature. Front Neurol. Neurol. 2019;18(1):107–­16.
2021;12:630366. 57. Schulz A, Ajayi T, Specchio N, de Los RE, Gissen P, Ballon D,
41. Sobral I, Cachulo Mda L, Figueira J, Silva R. Sialidosis type I: et al. Study of intraventricular cerliponase Alfa for CLN2 dis-
ophthalmological findings. BMJ Case Rep. 2014;2014:bcr2014​ ease. N Engl J Med. 2018;378(20):1898–­907.
205871. 58. Nita DA, Mole SE, Minassian BA. Neuronal ceroid lipofuscino-
42. Kivlin JD, Sanborn GE, Myers GG. The cherry-­red spot ses. Epileptic Disord. 2016;18(S2):73–­88.
in Tay-­Sachs and other storage diseases. Ann Neurol. 59. Sinha S, Satishchandra P, Gayathri N, Yasha TC, Shankar SK.
1985;17(4):356–­60. Progressive myoclonic epilepsy: a clinical, electrophysiolog-
43. Rubboli G, Veggiotti P, Pini A, Berardinelli A, Cantalupo G, ical and pathological study from South India. J Neurol Sci.
Bertini E, et al. Spinal muscular atrophy associated with pro- 2007;252(1):16–­23.
gressive myoclonic epilepsy: a rare condition caused by muta- 60. Malek N, Stewart W, Greene J. The progressive myoclonic epi-
tions in ASAH1. Epilepsia. 2015;56(5):692–­8. lepsies. Pract Neurol. 2015;15(3):164–­71.
44. Hadjivassiliou M, Sanders DS, Grunewald RA, Woodroofe 61. Berkovic SF, Carpenter S, Andermann F, Andermann E, Wolfe
N, Boscolo S, Aeschlimann D. Gluten sensitivity: from gut to LS. Kufs' disease: a critical reappraisal. Brain. 1988;111(Pt
brain. Lancet Neurol. 2010;9(3):318–­30. 1):27–­62.
45. Sarrigiannis PG, Hoggard N, Aeschlimann D, Sanders DS, 62. Berkovic SF, Staropoli JF, Carpenter S, Oliver KL, Kmoch
Grunewald RA, Unwin ZC, et al. Myoclonus ataxia and refrac- S, Anderson GW, et al. Diagnosis and misdiagnosis of adult
tory coeliac disease. Cerebellum Ataxias. 2014;1:11. neuronal ceroid lipofuscinosis (Kufs disease). Neurology.
46. van der Veen S, Zutt R, Elting JWJ, Becker CE, de Koning TJ, 2016;87(6):579–­84.
Tijssen MAJ. Progressive myoclonus ataxia: time for a new defi- 63. Iivanainen M, Himberg JJ. Valproate and clonazepam in the
nition? Mov Disord. 2018;33(8):1281–­6. treatment of severe progressive myoclonus epilepsy. Arch
47. Crespel A, Ferlazzo E, Franceschetti S, Genton P, Gouider R, Neurol. 1982;39(4):236–­8.
Kalviainen R, et al. Unverricht-­Lundborg disease. Epileptic 64. Orsini A, Valetto A, Bertini V, Esposito M, Carli N, Minassian
Disord. 2016;18(S2):28–­37. BA, et al. The best evidence for progressive myoclonic epilepsy:
48. So N, Berkovic S, Andermann F, Kuzniecky R, Gendron a pathway to precision therapy. Seizure. 2019;71:247–­57.
D, Quesney LF. Myoclonus epilepsy and ragged-­red fibres 65. Cardinali S, Canafoglia L, Bertoli S, Franceschetti S, Lanzi G,
(MERRF). 2. Electrophysiological studies and comparison with Tagliabue A, et al. A pilot study of a ketogenic diet in patients
other progressive myoclonus epilepsies. Brain. 1989;112(Pt with Lafora body disease. Epilepsy Res. 2006;69(2):129–­34.
5):1261–­76. 66. van Egmond ME, Weijenberg A, van Rijn ME, Elting JW,
49. Avanzini G, Shibasaki H, Rubboli G, Canafoglia L, Panzica F, Gelauff JM, Zutt R, et al. The efficacy of the modified Atkins
Franceschetti S, et al. Neurophysiology of myoclonus and pro- diet in North Sea progressive myoclonus epilepsy: an obser-
gressive myoclonus epilepsies. Epileptic Disord. 2016;18(S2):1 vational prospective open-­label study. Orphanet J Rare Dis.
1–­27. 2017;12(1):45.
50. Rubboli G, Meletti S, Gardella E, Zaniboni A, d'Orsi G, Dravet 67. Groh J, Berve K, Martini R. Fingolimod and teriflunomide
C, et al. Photic reflex myoclonus: a neurophysiological study attenuate neurodegeneration in mouse models of neuronal
in progressive myoclonus epilepsies. Epilepsia. 1999;40(Suppl ceroid Lipofuscinosis. Mol Ther. 2017;25(8):1889–­99.
4):50–­8. 68. Macauley SL, Wong AM, Shyng C, Augner DP, Dearborn JT,
51. Guellerin J, Hamelin S, Sabourdy C, Vercueil L. Low-­frequency Pearse Y, et al. An anti-­neuroinflammatory that targets dysreg-
photoparoxysmal response in adults: an early clue to diagnosis. ulated glia enhances the efficacy of CNS-­directed gene therapy
J Clin Neurophysiol. 2012;29(2):160–­4. in murine infantile neuronal ceroid lipofuscinosis. J Neurosci.
52. Berkovic SF, Oliver KL, Canafoglia L, Krieger P, Damiano JA, 2014;34(39):13077–­82.
Hildebrand MS, et al. Kufs disease due to mutation of CLN6: 69. Sanchez-­Elexpuru G, Serratosa JM, Sanz P, Sanchez MP.
clinical, pathological and molecular genetic features. Brain. 4-­Phenylbutyric acid and metformin decrease sensitivity to
2019;142(1):59–­69. pentylenetetrazol-­induced seizures in a malin knockout model
53. Kim J, Hu C, Moufawad El Achkar C, Black LE, Douville J, of Lafora disease. Neuroreport. 2017;28(5):268–­71.
Larson A, et al. Patient-­customized oligonucleotide therapy for 70. Berthier A, Paya M, Garcia-­Cabrero AM, Ballester MI, Heredia
a rare genetic disease. N Engl J Med. 2019;381(17):1644–­52. M, Serratosa JM, et al. Pharmacological interventions to ame-
54. Rubboli G, Franceschetti S, Berkovic SF, Canafoglia L, liorate neuropathological symptoms in a mouse model of
Gambardella A, Dibbens LM, et al. Clinical and neurophys- Lafora disease. Mol Neurobiol. 2016;53(2):1296–­309.
iologic features of progressive myoclonus epilepsy with- 71. Bisulli F, Muccioli L, d'Orsi G, Canafoglia L, Freri E, Licchetta
out renal failure caused by SCARB2 mutations. Epilepsia. L, et al. Treatment with metformin in twelve patients with
2011;52(12):2356–­63. Lafora disease. Orphanet J Rare Dis. 2019;14(1):149.
|

19506945, 2023, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/epd2.20152, Wiley Online Library on [02/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
CAMERON et al.    679

72. Kleine Holthaus SM, Herranz-­Martin S, Massaro G, Aristorena


M, Hoke J, Hughes MP, et al. Neonatal brain-­directed gene ther-
Memphis, USA; Andreas Brunklaus, Institute of Health
apy rescues a mouse model of neurodegenerative CLN6 batten and Wellbeing, University of Glasgow, UK; Gaetan
disease. Hum Mol Genet. 2019;28(23):3867–­79. Lesca, Department of Genetics, Hospices Civils de
73. Matos L, Duarte AJ, Ribeiro D, Chaves J, Amaral O, Alves S. Lyon, Bron, France.
Correction of a splicing mutation affecting an Unverricht-­
Lundborg disease patient by antisense therapy. Genes (Basel). Full list of ILAE Genetic Literacy Task
2018;9(9):455. Force members
Elizabeth Emma Palmer, Centre of Clinical Genetics.
SUPPORTING INFORMATION Sydney Children's Hospitals Network, and University
Additional supporting information can be found online of New South Wales, Randwick, NSW, Australia;
in the Supporting Information section at the end of this Amy McTague, Developmental Neurosciences, UCL
article. Great Ormond Street Institute of Child Health, and
Department of Neurology, Great Ormond Street Hospital
for Children, London, UK; Faiza Fakhfakh, Laboratory
of molecular and functional genetics, Faculty of sci-
How to cite this article: Cameron JM, Ellis CA,
ence, Sfax University of Sfax, Tunisia; Norman Delanty,
Berkovic SF. ILAE Genetics Literacy series:
Department of Neurology, Beaumont Hospital, Dublin,
Progressive myoclonus epilepsies. Epileptic Disord.
and FutureNeuro Research Centre, RCSI, Dublin; Daniel
2023;25:670–680. https://doi.org/10.1002/epd2.20152
H. Lowenstein, Department of Neurology, University
of California, San Francisco, USA; Nigel C. K. Tan,
Department of Neurology, National Neuroscience
APPENDIX 1 Institute, Singapore, Singapore; Ingo Helbig, Division
of Neurology, The Children's Hospital of Philadelphia,
Full list of ILAE Genetics Commission Philadelphia, USA; Alina I. Esterhuizen, UCT/MRC
members Genomic and Precision Medicine Research Unit, Division
Piero Perucca, Bladin-­Berkovic Comprehensive of Human Genetics, Department of Pathology, Institute
Epilepsy Program, Austin Health, Australia, and of Infectious Disease and Molecular Medicine, Faculty of
Department of Medicine (Austin Health), Melbourne Health Sciences, University of Cape Town, Cape Town,
Medical School, The University of Melbourne, Australia; South Africa.
J. Helen Cross, UCL-­Institute of Child Health, Great
Ormond Street Hospital for Children, London & Young APPENDIX 2
Epilepsy, Lingfield, UK; Holger Lerche, Department of
Neurology and Epileptology, Hertie Institute for Clinical Case study
Brain Research, University of Tuebingen, Tuebingen, A 12-­year-­old female was referred to neurology out-
Germany; Alina I. Esterhuizen, UCT/MRC Genomic patients with a six-­month history of increasing upper
and Precision Medicine Research Unit, Division of and lower limb jerks, which were starting to impact
Human Genetics, Department of Pathology, Institute her ability to complete schoolwork. She had a history
of Infectious Disease and Molecular Medicine, Faculty of three generalized tonic–­clonic seizures over a two-­
of Health Sciences, University of Cape Town, Cape year period, on the background of unremarkable birth
Town, South Africa; Iscia Lopes-­Cendes, Department and developmental milestones. There was no signifi-
of Translational Medicine, University of Campinas, cant family history. Examination revealed some subtle
Campinas, Brazil; Meng-­Han Tsai, Department of difficulty with tandem gait, with no other focal neuro-
Neurology, Kaohsiung Chang Gung Memorial Hospital logical deficit. The remainder of her general physical
and Chang Gung University College of Medicine, examination was unremarkable. EEG revealed normal
Kaohsiung, Taiwan; Daniel H. Lowenstein, Department background rhythm, with rare generalized spike–­wave
of Neurology, University of California, San Francisco, discharges and a grade IV photoparoxysmal response.
USA; Nigel C. K. Tan, Department of Neurology, A diagnosis of Juvenile Myoclonic Epilepsy was made,
National Neuroscience Institute, Singapore, Singapore; and the patient was commenced on 500 mg bd lev-
Ingo Helbig, Division of Neurology, The Children's etiracetam. Over the subsequent 12 months, she had
Hospital of Philadelphia, Philadelphia, USA; Heather no further convulsive seizures, though she had in-
C. Mefford, Center for Pediatric Neurological Disease creasingly severe and frequent upper and lower limb
Research, St. Jude Children's Research Hospital, jerks, was quite clumsy, and had several falls despite
|

19506945, 2023, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/epd2.20152, Wiley Online Library on [02/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
680    CAMERON et al.

adjustments to her antiseizure medication including Case resolution


uptitration of levetiracetam. Examination revealed ac- The targeted PME gene panel did not reveal any patho-
tion-­ and stimulus-­sensitive myoclonus and appendicu- genic variants for our patient. CSTB repeat expansion test-
lar and truncal ataxia. Further investigations including ing was subsequently requested, revealing a homozygous
MRI Brain and basic biochemical testing provided no 12-­nucleotide dodecamer repeat expansion, thus provid-
further diagnostic clues. Cognitive function remained ing molecular confirmation of a diagnosis of Unverricht-­
intact. Given the progressive nature of her condition, Lundborg Disease. Myoclonus continued to progress over
the diagnosis of Progressive Myoclonic Epilepsy (PME) the next 3 years, despite the addition of sodium valproate,
was considered. After appropriate patient and fam- piracetam, and clonazepam. A wheelchair was required
ily genetic counseling, a targeted PME gene panel was for mobilization at the age of 30 years due to the severity
requested. of myoclonus.

Test yourself
1. In a 15-­year-­old male with a history of myoclonus and epilepsy, which of the following features might sug-
gest he has a PME?
A. Macular cherry-­red spot
B. Occipital seizures in his EEG
C. Hepatosplenomegaly
D. All of the above
2. The Progressive Myoclonic Epilepsies
A. Are very rarely associated with ataxia
B. Typically present in the 4th and 5th decades of life
C. Present with myoclonus that is fragmentary and multifocal
D. Can be diagnosed using genetic testing in about 25% of cases
Answers may be found in the supporting information.

You might also like