This is an open access publication, available online and distributed under the terms of a Creative Commons Attribution-Non Commercial-No Derivatives 4.0 International licence (CC BY-NC-ND 4.0), a copy of which is available at https://creativecommons.org/licenses/by-nc-nd/4.0/. Subject to this license, all rights are reserved.
NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.
Noebels JL, Avoli M, Rogawski MA, et al., editors. Jasper's Basic Mechanisms of the Epilepsies. 5th edition. New York: Oxford University Press; 2024. doi: 10.1093/med/9780197549469.003.0056
Abstract
Progressive myoclonus epilepsies (PMEs) are genetic diseases that together account for about 1% of all epilepsies in childhood and adolescence. Even though the age of onset, symptom spectrum, and severity vary among different PMEs, all patients experience myoclonic seizures, generalized epilepsy, and progressive neurological decline, often intractable and fatal. Antisense oligonucleotides (ASOs) are synthetic single-stranded chemically modified nucleic acid polymers that target RNA to alter splicing, modulate translation, or promote/prevent mRNA degradation. Their gene-specific action and versatility has made ASO-based therapeutics one of the major pillars of drug development for genetic diseases with more than a dozen ASO drugs FDA-approved and many more in development and clinical trials. Advances in ASO technology already paved the way toward “N of 1” patient-customized therapies and hold great promise for personalized drug development that is much needed to treat rare genetic diseases such as PMEs. This chapter discusses the potential of ASO therapies for PMEs by giving an overview of the possible mechanisms of action employed to target disease genes and by reviewing ASO studies aimed to treat neuromuscular or neurodegenerative diseases, including PMEs.
Progressive Myoclonus Epilepsies—A Brief Overview
Progressive myoclonus epilepsies (PMEs) are a group of clinically diverse disorders caused by mutations in specific genes. All PMEs are extremely rare, but together they account for about 1% of all epilepsies in children and adolescents and certainly belong to the most serious epileptic syndromes known (Orsini et al., 2019). Commonalities among PMEs include the occurrence of myoclonus and generalized epilepsy as well as progressive neurological decline, including dementia and ataxia. As the disease progresses, PMEs in many cases become intractable and often fatal. With no cures available, treatments usually aim to reduce symptoms such as seizures combined with palliative, supportive, and rehabilitative measures (Kälviäinen, 2015; Minassian et al., 2016; Orsini et al., 2019).
The most common PMEs are Unverricht-Lundborg disease (ULD), Lafora disease (LD), the group of neuronal ceroid lipofuscinoses (NCLs/CLNs), and myoclonus epilepsy and ragged-red fibers (MERFF) (Kälviäinen, 2015). In most cases ULD is caused by mutations in the CSTB gene, but in some instances mutations in SCARB2 or PRICKLE1 were found instead (Table 56–1). Most PMEs are autosomal recessive disorders, but there are examples of autosomal dominant inheritance, including dentatorubral-pallidoluysian atrophy (DRPLA), CLN4B, progressive myoclonus epilepsy 11 (EPM11), myoclonus epilepsy and ataxia due to pathogenic variants in the potassium [K] channel (MEAK/EPM7), and familial encephalopathy with neuroserpin inclusion bodies (FENIB). MERRF is caused by mutations in mitochondrial genes and inheritance is therefore maternal. Some of the diseases affect the lysosome and because of that are also classified as lysosomal storage disorders, including NCLs, sialidosis, Gaucher disease (GD), and Niemann-Pick type C (NPC). Details on the symptom spectrum of the different PMEs and their basic disease mechanism (if known) are reviewed elsewhere and beyond the scope of this book chapter.

Table 56–1
Progressive Myoclonus Epilepsies with Preclinically or Clinically Tested ASO Strategies.
As mentioned above, there are no effective and curative therapies for most PME disorders. Exceptions are an FDA-approved enzyme replacement therapy for CLN2 (de Los Reyes et al., 2020) and gene therapy approaches that are in development or clinical trials for CLN1, CLN2, CLN3, CLN5, CLN6, and CLN7 (Brenner et al., 2020; Masten et al., 2020). In the following, different ways of utilizing antisense oligonucleotide technology to treat genetic diseases will be introduced and their potential as therapies for PMEs presented and discussed. References showing examples of ASO strategies for PMEs are also listed in Table 56–1.
Antisense Oligonucleotides and Their Different Modes of Action
Antisense oligonucleotides (ASOs) are synthetic single-stranded chemically modified nucleic acid (RNA or DNA) polymers. They target RNA via complementary Watson-Crick base pairing, which is the basis of their specificity and their capability to modulate RNA processing (Sharma and Watts, 2015). Their high specificity enables targeting of specific transcript isoforms (Han et al., 2020; Lim et al., 2020; Roberts et al., 2020) and disease alleles with only single base pair changes (Carroll et al., 2011; Southwell et al., 2018), without affecting the wild-type RNA, making even “undruggable” targets “druggable.” The therapeutic potential of ASOs is reflected by the recent approval of more than a dozen ASO drugs (Kuijper et al., 2021; Roberts et al., 2020; Xiong et al., 2021) and the high number of ASOs currently in clinical trials for genetic diseases (Bennett et al., 2017; Brenner et al., 2020; Roberts et al., 2020; Silva et al., 2020) and cancer (Xiong et al., 2021).
One major obstacle for ASO therapeutics to become even more widespread is the difficulty of efficient delivery, especially to extrahepatic tissues (Roberts et al., 2020). For central nervous system (CNS) diseases, direct injection of ASOs into the cerebrospinal fluid (CSF) leads to good distribution and therapeutic effects, as demonstrated with the ASO drug nusinersen for spinal muscular atrophy (Finkel et al., 2016). Active research is going into improving ASO drug delivery and stability by chemical modifications, tissue/cell-targeting or cell-penetrating covalent conjugates, and nanoparticle vehicles (Gagliardi and Ashizawa, 2021; Roberts et al., 2020).
ASO design is very flexible and allows for multiple mechanisms of action, depending on the specific ASO chemistry and on the target sequence (Chan et al., 2006; Scoles et al., 2019). The most widely used mechanisms either promote RNA degradation via RNase H or function without RNA degradation by steric hindrance, leading to translation arrest or splice modulation (Bennett et al., 2017). In the former case, protein knockdown is achieved by recruitment of endonuclease RNase H to the ASO-mRNA heteroduplex, which results in mRNA degradation, leaving the DNA-based ASO behind (Chan et al., 2006; Roberts et al., 2020). ASO-dependent protein knockdown is not only valuable for developing therapeutic approaches but has been utilized to generate PME disease models, for example, by targeting the ASAH1 ortholog in zebrafish for SMA-PME (Zhou et al., 2012) or murine Npc1 for Nieman-Pick type C (Rimkunas et al., 2008). Translation arrest and splice modulation are achieved by steric-block ASOs. In the case of splice modulation, the ASO targets the pre-mRNA to mask a specific sequence, for example, splice sites or exonic/intronic cis-regulatory elements. Altering splicing can either correct splicing defects due to mutations, or use exon skipping or inclusion to restore an open reading frame or to disrupt it, thereby causing mRNA degradation through nonsense-mediated mRNA decay (NMD). It can be used to remove exons with the disease-causing mutation or to induce isoform switching, for example, promoting the splicing of a beneficial alternatively spliced isoform (Kuijper et al., 2021; Roberts et al., 2020; Silva et al., 2020, 2014).
ASO Strategies to Treat PME Disorders
ASOs That Modulate Splicing
One mode of action of ASOs is the modulation of splicing. This can be advantageous for cases where (point) mutations cause splicing defects, or where deletions lead to frameshifts/premature termination codons (PTCs) and altering splicing restores the open reading frame, preventing NMD or the formation of strongly truncated proteins. Two well-known examples for ASO therapies targeting splicing are the FDA-approved drugs for the neuromuscular diseases spinal muscular atrophy (SMA, nusinersen; Finkel et al., 2017; Mercuri et al., 2018) and Duchenne muscular dystrophy (DMD, eteplirsen; Alfano et al., 2019; Mendell et al., 2016).
SMA is caused by mutations in SMN1 (Survival Motor Neuron 1). A paralogous gene, SMN2, exists and encodes a functional SMN protein. However, exon 7 is removed during splicing in about 90% of the SMN2 transcripts, resulting in truncated/nonfunctional proteins and leaving only about 10% of full-length mRNA and hence very low levels of functional SMN protein. Nusinersen binds to an intronic splicing silencer (ISS) element in intron 7 to prevent splicing, which leads to exon-7 inclusion and increased levels of functional SMN protein (Fig. 56–1A) (Chiriboga et al., 2016; Mercuri et al., 2020).

Figure 56–1.
ASO strategies for neuromuscular and neurodegenerative diseases. (A) Exon inclusion by nusinersen for SMA. (B) Exon skipping by eteplirsen for DMD. (C) ASO-dependent exon skipping for CLN3. (D) Exclusion of cryptic exon by milasen for CLN7. (more...)
DMD is caused by mutations in the DMD gene, encoding dystrophin. Most of the DMD mutations are deletions. The most severe DMD phenotype results from out-of-frame mutations with no or negligible amounts of dystrophin. In contrast, in-frame mutations cause the milder dystrophinopathy Becker MD (BMD). The ASO strategy for DMD is to delete an exon (e.g., exon-51 skipping by eteplirsen; Fig. 56–1B) adjacent to the deletion to generate an in-frame deletion that results in a truncated but still functional variant of dystrophin, shifting the disease phenotype from severe DMD to milder BMD (Duan et al., 2021; Verma, 2018).
Similar to the ASO-dependent exon-51 skipping in DMD, an exon-skipping approach has been studied for CLN3 Batten disease (Centa et al., 2020). In most cases this disease is caused by deletions of exon 7 and 8, which results in a PTC in exon 9 (Cotman et al., 2002; Munroe et al., 1997). ASO-mediated skipping of exon 5 through steric hindrance restored the open reading frame (Fig. 56–1C), leading to the inclusion of the lysosomal-targeting sequence (LTS) containing C terminus (Centa et al., 2020), the loss of which invariably leads to more severe forms of the disease (Centa et al., 2020; Kousi et al., 2012). In the disease mouse model, ASO treatment resulted in improved motor coordination, reduced histopathology, and increased survival (Centa et al., 2020).
Intriguingly, an ASO-based “N of 1” therapy was developed for a patient with CLN7. A unique mutation was found in the MFSD8 gene of this patient where an SVA retrotransposon insertion generated a cryptic splice site that resulted in a PTC-containing transcript. Milasen, a splicing-altering steric-block ASO, was developed to enable proper splicing of intron 6 and hence an increase in correctly spliced MFSD8 transcripts (Fig. 56–1D). Administration to the patient reduced frequency and duration of seizures, a few other symptoms stabilized, but neurological assessment scores continued to decline (Kim et al., 2019). Even though the ASO was not able to restore lost function, this case is an exciting example of precision medicine and patient-customized treatment that is possible today.
Disease-causing missense mutations are commonly assumed to disrupt protein function. However, 22%–25% of exonic disease mutations are estimated to be splicing sensitive (Lim et al., 2011; Sterne-Weiler et al., 2011), with up to 50% for genes that are particularly sensitive toward mutation-induced aberrant splicing (e.g., DMD; Sterne-Weiler and Sanford, 2014). Splicing defects are not only caused by missense mutations but also by deletions, silent and nonsense mutations, and intronic mutations, inactivating or weakening 3′ or 5′ splice sites or disrupting/creating splicing enhancer/silencer elements (Daguenet et al., 2015; Ward and Cooper, 2010). Overall, it is estimated that approximately one-third of all disease-causing mutations are splicing mutations (Lim et al., 2011). Splicing defects have been described for PMEs such as Gaucher type 3 (Tonin et al., 2019), sialidosis (Caciotti et al., 2009; Penzel et al., 2001), CLN3 (Mirza et al., 2019), CLN13 (Di Fabio et al., 2014), AMRF/EPM4 (Dibbens et al., 2009; Yari et al., 2021), ULD (Matos et al., 2018), and NPC (Encarnação et al., 2020; Paron et al., 2020; Zech et al., 2013). ASO approaches have been tested for the latter two diseases (Matos et al., 2018; Rodriguez-Pascau et al., 2009), both as proof-of-principle studies in patient fibroblasts. Splicing mutations are more frequent than commonly thought, and it will be interesting to revisit known PME-causing mutations for their possible effect on splicing to potentially find suitable ASO therapies targeting the root of the problem.
ASOs Targeting mRNA for Degradation via RNase H
Mutations in a disease gene often result in loss of function, in which case further downregulation of gene expression is not meaningful. There are, however, instances where downregulation of gene expression is beneficial. That is the case for autosomal dominant diseases with mutations causing dominant-negative effects. Another example could be a disease where a target can be identified, and the downregulation of which prevents or slows down disease progression.
The ASO strategy for Lafora disease, proven to be successful in a preclinical study, is a great example for the latter case (Ahonen et al., 2021). Lafora disease is caused by loss-of-function mutations in either EPM2A or NHLRC1/EPM2B, encoding the glycogen phosphatase laforin and E3 ubiquitin ligase malin, respectively (Chan et al., 2003; Minassian et al., 1998). Both proteins form a functional complex that is crucial for maintaining proper glycogen structure that ensures glycogen solubility. In Lafora disease, glycogen molecules are formed that have longer than normal glucan chains and fewer branch points, respectively, which can render the molecule insoluble. Over time, insoluble glycogen molecules accumulate and form pathognomonic, pathogenic polyglucosan bodies (better known as Lafora bodies [LBs]) in the brain and other organs (Nitschke et al., 2018; Sullivan et al., 2017). Inhibiting or slowing down glycogen synthesis has been proven to correct the neuropathological bases of Lafora disease in the disease’s mouse models (Duran et al., 2014; Gumusgoz et al., 2021; Israelian et al., 2020; Nitschke et al., 2021; Pederson et al., 2013; Turnbull et al., 2011, 2014; Varea et al., 2021). Glycogen synthase (GYS) is the rate-limiting enzyme during glycogen synthesis and therefore a valuable/promising therapeutic target in Lafora disease. ASO-mediated downregulation of Gys1 transcripts (Fig. 56–1E) efficiently prevents further disease progression in Lafora disease mice, including the halt of LB formation, glycogen accumulation, and neuroinflammation (Ahonen et al., 2021).
Although most PMEs are autosomal recessive disorders, there are a few that are autosomal dominant with dominant-negative effects caused by only one mutant allele such as CLN4B (Naseri et al., 2021), MEAK/EPM7 (Muona et al., 2015), EPM11 (Hamanaka et al., 2020), FENIB (Roussel et al., 2016), and DRPLA (Tsuji, 2012). Using ASOs that lead to RNase H-dependent degradation of the target mRNA can be beneficial for these diseases. DRPLA, for example, belongs to a group of nine neurodegenerative diseases that are caused by CAG repeat expansions, leading to expanded polyglutamine (polyQ) tracts in the resulting proteins, hence called polyQ disorders (Lieberman et al., 2019; Riley and Orr, 2006). Well-known examples of polyQ disorders are Huntington’s disease (HD) or spinocerebellar ataxias (SCA) for which several preclinical ASO trials showed promising results, with ASO drugs for HD even progressing to clinical trials (Silva et al., 2020), with some trials terminated but a new one launched that is still recruiting. Non-allele-specific ASO approaches targeting both wild-type and mutant huntingtin (HTT) mRNA (Kordasiewicz et al., 2012) as well as allele-specific strategies targeting only the mutant mRNA (Carroll et al., 2011; Datson et al., 2017; Southwell et al., 2018) (Fig. 56–1F) showed success in preclinical studies. It needs to be evaluated for each of the autosomal dominant disorders individually if a non-allele-specific ASO strategy is an option, depending on how essential the (wild-type) protein’s function is for cellular processes or even survival.
For DRPLA, a CAG repeat targeting ASO was tested in patient fibroblasts and led to a 98% downregulation of mutant atrophin-1 (ATN1), while the wild-type ATN1 mRNA was only reduced by 30% (Evers et al., 2011). Even though CAG repeat targeting ASO approaches can be allele-specific, it depends on the ASO and the disease allele how selectively the mutant mRNA is reduced because even the wild-type alleles contain CAG repeats only much less than the mutant alleles in polyQ disorders (for CAG repeat numbers, see Riley and Orr, 2006; and Lieberman et al., 2019). Another concern could be the nonspecific reduction of other genes containing CAG repeats. For DRPLA, ASO studies in mice have not been done, and it is unclear what consequences an additional reduction of the wild-type Atn1 mRNA would have. Interestingly, Atn1–/– null mice are viable and fertile (Shen et al., 2007) and mice expressing a truncated ATN1 protein lacking both the polyQ repeat and the following C-terminal peptides (Atn1t/t) display growth retardation and progressive male infertility, but no obvious signs of neurodegeneration (Yu et al., 2009). Other studies have used ASOs, targeting SNPs that are tightly linked to CAG expansion in the mutant HTT gene, which successfully and selectively silenced the latter and reduced cognitive and behavioral impairments (Carroll et al., 2011; Southwell et al., 2018).
The fact that ASO technology is so advanced that ASOs can be designed that selectively target a mutant allele with a single base pair difference from the wild-type allele holds great promise for the other autosomal dominant PMEs with dominant negative effects of the mutant allele. However, this approach may be impractical for diseases where ASOs need to be designed for multiple different mutations, and in these cases a combination of a non-allele-specific ASO drug with an enzyme or gene replacement therapy could be an alternative solution. Other RNase H-independent ASO strategies such as translation inhibition or exon skipping could be considered as well (Silva et al., 2020); specifically, proof-of-concept studies using ASOs that cause exon skipping have shown potential for HD (Evers et al., 2014) and SCA3 (Evers et al., 2013).
Upregulation of Gene Expression through ASOs
For some diseases it would be beneficial to upregulate gene expression or promote translation of a certain gene product. One example would be haploinsufficiencies, one type of autosomal dominant diseases where the remaining intact allele is not sufficiently expressed to compensate for loss of function of the other allele. Increasing the gene output could also help in cases where the disease-causing mutation did not result in a complete loss of function (hypomorphic alleles), or where only protein levels and not functionality of the protein are affected. A third example would be to promote expression of another target (not the disease gene) which, once upregulated, helps slowing or preventing disease progression. In sialidosis (caused by NEU1 mutations), such a target would be protective protein cathepsin A (PPCA, encoded by the CTSA gene), the natural chaperone of NEU1. Mutations in CTSA lead to galactosialidosis, involving secondary deficiency of NEU1. Proper folding, lysosomal localization, and catalytic activation of NEU1 depend on PPCA (d’Azzo et al., 2015; Mosca et al., 2020).
Even though ASOs are more commonly known for modulating splicing, promoting RNA degradation, or causing translation arrest, there are ways to increase protein production utilizing ASO technology. Antagonizing microRNAs which otherwise suppress protein production has been shown as one possible mode of action (Esau et al., 2006; Lima et al., 2018; Rotllan et al., 2013). Another possibility to increase levels of therapeutic proteins would be for the ASO to bind to upstream ORFs (uORFs) or other translation inhibitory elements in the 5′ UTR, thereby blocking inhibition of translation initiation and hence promoting translation (Liang et al., 2016, 2017). A third way to upregulate protein levels is to prevent nonproductive alternative splicing (AS) events that induce NMD due to the presence of PTCs in the resulting transcript isoforms (Lim et al., 2020). This approach is called TANGO (Targeted Augmentation of Nuclear Gene Output) and basically utilizes the capability of ASOs to alter splicing in a gene-specific and AS type-dependent fashion.
TANGO has been further tested, and an ASO (STK-001) has been selected and is currently undergoing a phase 1/2 clinical trial for Dravet syndrome (DS) (Han et al., 2020; Hill and Meisler, 2021). Dravet syndrome (OMIM # 607208) is classified as developmental and epileptic encephalopathy (DEE) and involves, very similar to PMEs, severe epilepsy (including myoclonic seizures) resistant to antiepileptic drugs and cognitive decline. In most cases, the disease is caused by mutations in one of the two SCN1A alleles, encoding the type I voltage-gated Na2+ channel Nav1.1, leading to haploinsufficiency (Depienne et al., 2009; Dravet and Oguni, 2013; Holmes, 2020). The steric-block ASO prevents the naturally occurring alternative splicing event, characterized by inclusion of an alternative (poison) exon (part of intron 20; Fig. 56–1G), thereby increasing the levels of productive full-length Scn1a transcripts and Nav1.1 protein in a DS mouse model. In this haploinsufficient DS mouse model, seizures were reduced and sudden unexpected death in epilepsy (SUDEP) prevented for up to 90 days upon ASO treatment (Han et al., 2020).
Interestingly, around 90% of the human genes undergo alternative splicing (McGlincy and Smith, 2008; Wang et al., 2008). This is not just a noise in the splicing process, but it generates distinct protein isoforms that either have different functions or regulate each other post-translationally. Approximately a third of all AS events in humans lead to nonproductive mRNAs, which are part of an important post-transcriptional mechanism (called AS-NMD), that is tightly regulated, to control gene expression (Hillman et al., 2004; McGlincy and Smith, 2008). Lim et al. analyzed 83 publicly available RNA-sequencing datasets from different human tissues and discovered 7757 genes undergoing at least one nonproductive AS event, 1246 of which are potentially disease-associated (Lim et al., 2020). According to these results, a significant number of PME-causing disease genes such as SCARB2 (ULD, AMRF/EPM4), EPM2A (LD), PPT1 (CLN1), TPP1 (CLN2), CLN3 (CLN3), DNAJC5 (CLN4B), CLN5 (CLN5), CLN6 (CLN6), MFSD8 (CLN7), CLN8 (CLN8), CTSD (CLN10), and KCNC1 (MEAK/EPM7), as well as CTSA, which could be beneficial to upregulate in sialidosis as described above, were found to be subjected to at least 1, and up to 13 different, nonproductive AS events. Together, this means there is an opportunity to possibly develop TANGO approaches for specific PMEs.
Combination of ASOs with Readthrough Drugs to Target Nonsense Mutations
Approximately 10%–12% of monogenic diseases are caused by nonsense mutations (Bidou et al., 2012; Mort et al., 2008), and there are several PMEs for which nonsense mutations have been described, including LD (Singh and Ganesh, 2009), sialidosis (Pattison et al., 2004), GD (Hruska et al., 2008), AMRF/EPM4 (Balreira et al., 2008), and NCL (Kousi et al., 2012). Interestingly, around 50% of infantile NCL-causing PPT1 mutations are nonsense mutations (Miller et al., 2015), and hence compounds called nonsense suppression or readthrough drugs that override PTCs to enable translation of full-length proteins (Bidou et al., 2012; Dabrowski et al., 2018) could be beneficial for CLN1 patients with these mutations. Readthrough drugs such as ataluren (PTC-124) or gentamicin have been tested in CLN1 patient cell lines (Miller et al., 2013; Sarkar et al., 2011; Sleat et al., 2001) and a CLN1 mouse model that carries the most common nonsense mutation in CLN1 patients (p.R151X) (Miller et al., 2015; Thada et al., 2016). Even though the results showed significant increase in full-length PPT1 protein and PPT1 activity, the overall efficacy of readthrough drugs is limited by NMD (Dabrowski et al., 2018).
NMD not only regulates gene expression through AS-NMD (as explained above) but also serves as a mRNA surveillance mechanism that degrades mRNAs with PTCs, resulting from nonsense mutations, frameshift mutations, deletions, splicing-altering mutations, or nonproductive alternative splicing, to prevent production of aberrant/truncated proteins (Brogna and Wen, 2009). Therefore, it reduces the amount of mRNA that can be targeted by readthrough drugs, and a combination of readthrough drugs with NMD-inhibiting drugs could be promising (Geraets et al., 2016; Keeling and Bedwell, 2011; Nickless et al., 2017). This strategy has been successfully applied in a mouse model of Hurler’s syndrome (Keeling et al., 2013) and in cancer cells (Martin et al., 2014) using NMD-attenuating small molecules. Intriguingly, ASOs can also act as NMD suppressors and improve efficacy of readthrough drugs (Keenan et al., 2019; Nomakuchi et al., 2016). ASOs that target translation termination even had synergistic effects in combination with the readthrough drug geneticin in a mouse model of hemophilia (Huang et al., 2019). A similar approach could be envisioned for PTC-containing pathogenic variants that cause PMEs such as in CLN1 patients (Fig. 56–1H).
Conclusions and Future Perspectives
In 2015, ASO-based therapeutics were predicted to become the third major pillar of drug development after small molecules and protein therapeutics (Sharma and Watts, 2015). Today, several ASO drugs are approved (Brenner et al., 2020; Kuijper et al., 2021; Xiong et al., 2021) and many are in development and clinical trials for diseases such as Huntington’s disease, Alzheimer’s disease, and amyotrophic lateral sclerosis (Bennett et al., 2017; Brenner et al., 2020; Roberts et al., 2020; Silva et al., 2020). Using ASOs has become a prominent therapeutic strategy for the treatment of genetic diseases, including CNS disorders, and cancer (Brenner et al., 2020; Hill and Meisler, 2021; Schoch and Miller, 2017; Xiong et al., 2021).
One problem that medicine faces when treating CNS disorders is the delivery of the drug to the brain. ASOs, like many drugs, do not cross the blood–brain barrier (BBB) and therefore intrathecal injections into the CSF are necessary to target the brain sufficiently (Brenner et al., 2020; Roberts et al., 2020). Once in the CSF, ASOs distribute broadly and are quite stable (up to months) in the brain (Mazur et al., 2019; Rigo et al., 2014). CNS-specific application is an advantage because adverse effects in other organs due to systemic administration can be circumvented and lower doses are sufficient, which is more cost-effective. On the other hand, untreated tissues may become clinically relevant later in the disease course. Also, repeated lumbar punctures are not only inconvenient but are invasive, and ways to overcome the BBB are being actively investigated and will allow for easier and less invasive dosing in the future.
Another advantage of ASOs is that they are relatively short-lived and elicit limited or no immunogenicity or toxicity, while virus-mediated gene therapy is strongly immunogenic and long-lasting. In case of adverse effects of the ASO drug, the treatment can be stopped, while gene therapy is more permanent (Brenner et al., 2020). On the other hand, longer stability of ASOs could allow for less frequent dosing. Different ASO chemistries and modifications, conjugations, or packaging of ASOs into vehicles such as nanoparticles will help improving ASO stability and bioavailability, and potentially allow for delivery into selective tissues, including BBB crossing (Gagliardi and Ashizawa, 2021; Roberts et al., 2020).
ASO therapy is generally very safe and efficacious and most importantly immensely versatile because of the many different possible modes of action (Fig. 56–1). This allows for truly individualized drug development as seen with milasen for a CLN7 patient (Kim et al., 2019). In this case the precedent of nusinersen’s success made the rapid development of milasen possible. This is certainly a very exciting and encouraging example of today’s precision medicine. As discussed above, there are multiple ways to utilize ASO technology to tackle the genetic defects that cause PMEs. The small number of patients carrying a specific mutation, however, is undoubtedly an obstacle in terms of safety testing and cost. The hope, though, is that ASO strategies will be developed further and that with more experience and additional successes ASO platforms can be established which enable the rapid development of customized therapies, even for extremely rare diseases such as PMEs.
Acknowledgments
This work was funded by the National Institute of Neurological Disorders and Stroke under award number P01NS097197. Dr. Minassian holds the UT Southwestern Jimmy Elizabeth Westcott Chair in Pediatric Neurology and is Chief Medical Advisor to Taysha Gene Therapies.
References
- Ahonen S, Nitschke S, Grossman TR, Kordasiewicz H, Wang P, Zhao X, Guisso DR, Kasiri S, Nitschke F, Minassian BA. Gys1 antisense therapy rescues neuropathological bases of murine Lafora disease. Brain. 2021; 144(10):2985-2993. doi: 10.1093/brain/awab194. PMID: 33993268. [PMC free article: PMC8634080] [PubMed: 33993268]
- Alfano LN, Charleston JS, Connolly AM, Cripe L, Donoghue C, Dracker R, Dworzak J, Eliopoulos H, Frank DE, Lewis S, Lucas K, Lynch J, Milici AJ, Flynt A, Naughton E, Rodino-Klapac LR, Sahenk Z, Schnell FJ, Young GD, Mendell JR, Lowes LP. Long-term treatment with eteplirsen in nonambulatory patients with Duchenne muscular dystrophy. Medicine (Baltimore). 2019;98(26):e15858. doi: 10.1097/MD.0000000000015858. PMID: 31261494. [PMC free article: PMC6617421] [PubMed: 31261494]
- Balreira A, Gaspar P, Caiola D, Chaves J, Beirao I, Lima JL, Azevedo JE, Miranda MC. A nonsense mutation in the LIMP-2 gene associated with progressive myoclonic epilepsy and nephrotic syndrome. Hum Mol Genet. 2008;17(14):2238–43. doi: 10.1093/hmg/ddn124. PMID: 18424452. [PubMed: 18424452]
- Bennett CF, Baker BF, Pham N, Swayze E, Geary RS. Pharmacology of Antisense Drugs. Annu Rev Pharmacol Toxicol. 2017;57:81–105. doi: 10.1146/annurev-pharmtox-010716-104846. PMID: 27732800. [PubMed: 27732800]
- Bidou L, Allamand V, Rousset JP, Namy O. Sense from nonsense: therapies for premature stop codon diseases. Trends Mol Med. 2012;18(11):679–88. doi: 10.1016/j.molmed.2012.09.008. PMID: 23083810. [PubMed: 23083810]
- Brenner D, Ludolph AC, Weishaupt JH. Gene specific therapies—the next therapeutic milestone in neurology. Neurol Res Pract. 2020;2:25. doi: 10.1186/s42466-020-00075-z. PMID: 33324928. [PMC free article: PMC7650126] [PubMed: 33324928]
- Brogna S, Wen J. Nonsense-mediated mRNA decay (NMD) mechanisms. Nat Struct Mol Biol. 2009;16(2):107–13. doi: 10.1038/nsmb.1550. PMID: 19190664. [PubMed: 19190664]
- Caciotti A, Di Rocco M, Filocamo M, Grossi S, Traverso F, d’Azzo A, Cavicchi C, Messeri A, Guerrini R, Zammarchi E, Donati MA, Morrone A. Type II sialidosis: review of the clinical spectrum and identification of a new splicing defect with chitotriosidase assessment in two patients. J Neurol. 2009;256(11):1911–5. doi: 10.1007/s00415-009-5213-4. PMID: 19568825. [PubMed: 19568825]
- Carroll JB, Warby SC, Southwell AL, Doty CN, Greenlee S, Skotte N, Hung G, Bennett CF, Freier SM, Hayden MR. Potent and selective antisense oligonucleotides targeting single-nucleotide polymorphisms in the Huntington disease gene/allele-specific silencing of mutant huntingtin. Mol Ther. 2011;19(12):2178–85. doi: 10.1038/mt.2011.201. PMID: 21971427. [PMC free article: PMC3242664] [PubMed: 21971427]
- Centa JL, Jodelka FM, Hinrich AJ, Johnson TB, Ochaba J, Jackson M, Duelli DM, Weimer JM, Rigo F, Hastings ML. Therapeutic efficacy of antisense oligonucleotides in mouse models of CLN3 Batten disease. Nat Med. 2020;26(9):1444–51. doi: 10.1038/s41591-020-0986-1. PMID: 32719489. [PMC free article: PMC8008709] [PubMed: 32719489]
- Chan EM, Bulman DE, Paterson AD, Turnbull J, Andermann E, Andermann F, Rouleau GA, Delgado-Escueta AV, Scherer SW, Minassian BA. Genetic mapping of a new Lafora progressive myoclonus epilepsy locus (EPM2B) on 6p22. J Med Genet. 2003;40(9):671–675. PMID: 12960212. [PMC free article: PMC1735578] [PubMed: 12960212]
- Chan JH, Lim S, Wong WS. Antisense oligonucleotides: from design to therapeutic application. Clin Exp Pharmacol Physiol. 2006;33(5-6):533–40. doi: 10.1111/j.1440-1681.2006.04403.x. PMID: 16700890. [PubMed: 16700890]
- Chiriboga CA, Swoboda KJ, Darras BT, Iannaccone ST, Montes J, De Vivo DC, Norris DA, Bennett CF, Bishop KM. Results from a phase 1 study of nusinersen (ISIS-SMN(Rx)) in children with spinal muscular atrophy. Neurology. 2016;86(10):890–897. doi: 10.1212/WNL.0000000000002445. PMID: 26865511. [PMC free article: PMC4782111] [PubMed: 26865511]
- Cotman SL, Vrbanac V, Lebel LA, Lee RL, Johnson KA, Donahue LR, Teed AM, Antonellis K, Bronson RT, Lerner TJ, MacDonald ME. Cln3(Deltaex7/8) knock-in mice with the common JNCL mutation exhibit progressive neurologic disease that begins before birth. Hum Mol Genet. 2002;11(22):2709–21. doi: 10.1093/hmg/11.22.2709. PMID: 12374761. [PubMed: 12374761]
- d’Azzo A, Machado E, Annunziata I. Pathogenesis, Emerging therapeutic targets and Treatment in Sialidosis. Expert Opin Orphan Drugs. 2015;3(5):491–504. doi: 10.1517/21678707.2015.1025746. PMID: 26949572. [PMC free article: PMC4777310] [PubMed: 26949572]
- Dabrowski M, Bukowy-Bieryllo Z, Zietkiewicz E. Advances in therapeutic use of a drug-stimulated translational readthrough of premature termination codons. Mol Med. 2018;24(1):25. doi: 10.1186/s10020-018-0024-7. PMID: 30134808. [PMC free article: PMC6016875] [PubMed: 30134808]
- Daguenet E, Dujardin G, Valcarcel J. The pathogenicity of splicing defects: mechanistic insights into pre-mRNA processing inform novel therapeutic approaches. EMBO Rep. 2015;16(12):1640–55. doi: 10.15252/embr.201541116. PMID: 26566663. [PMC free article: PMC4693517] [PubMed: 26566663]
- Datson NA, Gonzalez-Barriga A, Kourkouta E, Weij R, van de Giessen J, Mulders S, Kontkanen O, Heikkinen T, Lehtimaki K, van Deutekom JC. The expanded CAG repeat in the huntingtin gene as target for therapeutic RNA modulation throughout the HD mouse brain. PLoS One. 2017;12(2):e0171127. doi: 10.1371/journal.pone.0171127. PMID: 28182673. [PMC free article: PMC5300196] [PubMed: 28182673]
- de Los Reyes E, Lehwald L, Augustine EF, Berry-Kravis E, Butler K, Cormier N, Demarest S, Lu S, Madden J, Olaya J, See S, Vierhile A, Wheless JW, Yang A, Cohen-Pfeffer J, Chu D, Leal-Pardinas F, Wang RY. Intracerebroventricular Cerliponase Alfa for Neuronal Ceroid Lipofuscinosis Type 2 Disease: Clinical Practice Considerations From US Clinics. Pediatr Neurol. 2020;110:64–70. doi: 10.1016/j.pediatrneurol.2020.04.018. PMID: 32684372. [PubMed: 32684372]
- Depienne C, Trouillard O, Saint-Martin C, Gourfinkel-An I, Bouteiller D, Carpentier W, Keren B, Abert B, Gautier A, Baulac S, Arzimanoglou A, Cazeneuve C, Nabbout R, LeGuern E. Spectrum of SCN1A gene mutations associated with Dravet syndrome: analysis of 333 patients. J Med Genet. 2009;46(3):183–91. doi: 10.1136/jmg.2008.062323. PMID: 18930999. [PubMed: 18930999]
- Di Fabio R, Moro F, Pestillo L, Meschini MC, Pezzini F, Doccini S, Casali C, Pierelli F, Simonati A, Santorelli FM. Pseudo-dominant inheritance of a novel CTSF mutation associated with type B Kufs disease. Neurology. 2014;83(19):1769–70. doi: 10.1212/WNL.0000000000000953. PMID: 25274848. [PubMed: 25274848]
- Dibbens LM, Michelucci R, Gambardella A, Andermann F, Rubboli G, Bayly MA, Joensuu T, Vears DF, Franceschetti S, Canafoglia L, Wallace R, Bassuk AG, Power DA, Tassinari CA, Andermann E, Lehesjoki AE, Berkovic SF. SCARB2 mutations in progressive myoclonus epilepsy (PME) without renal failure. Ann Neurol. 2009;66(4):532–6. doi: 10.1002/ana.21765. PMID: 19847901. [PubMed: 19847901]
- Dravet C, Oguni H. Dravet syndrome (severe myoclonic epilepsy in infancy). Handb Clin Neurol. 2013;111:627–633. doi: 10.1016/B978-0-444-52891-9.00065-8. PMID: 23622210. [PubMed: 23622210]
- Duan D, Goemans N, Takeda S, Mercuri E, Aartsma-Rus A. Duchenne muscular dystrophy. Nat Rev Dis Primers. 2021;7(1):1-19. doi: 10.1038/s41572-021-00248-3. PMID: 33602943. [PMC free article: PMC10557455] [PubMed: 33602943]
- Duran J, Gruart A, Garcia-Rocha M, Delgado-Garcia JM, Guinovart JJ. Glycogen accumulation underlies neurodegeneration and autophagy impairment in Lafora disease. Hum Mol Genet. 2014;23(12):3147–56. doi: ddu024 [pii]10.1093/hmg/ddu024. PMID: 24452334. [PubMed: 24452334]
- Encarnação M, Coutinho MF, Cho SM, Cardoso MT, Ribeiro I, Chaves P, Santos JI, Quelhas D, Lacerda L, Leao Teles E, Futerman AH, Vilarinho L, Alves S. NPC1 silent variant induces skipping of exon 11 (p.V562V) and unfolded protein response was found in a specific Niemann-Pick type C patient. Mol Genet Genomic Med. 2020;8(11):e1451. doi: 10.1002/mgg3.1451. PMID: 32931663. [PMC free article: PMC7667330] [PubMed: 32931663]
- Esau C, Davis S, Murray SF, Yu XX, Pandey SK, Pear M, Watts L, Booten SL, Graham M, McKay R, Subramaniam A, Propp S, Lollo BA, Freier S, Bennett CF, Bhanot S, Monia BP. miR-122 regulation of lipid metabolism revealed by in vivo antisense targeting. Cell Metab. 2006;3(2):87–98. doi: 10.1016/j.cmet.2006.01.005. PMID: 16459310. [PubMed: 16459310]
- Evers MM, Pepers BA, van Deutekom JC, Mulders SA, den Dunnen JT, Aartsma-Rus A, van Ommen GJ, van Roon-Mom WM. Targeting several CAG expansion diseases by a single antisense oligonucleotide. PLoS One. 2011;6(9):e24308. doi: 10.1371/journal.pone.0024308. PMID: 21909428. [PMC free article: PMC3164722] [PubMed: 21909428]
- Evers MM, Tran HD, Zalachoras I, Meijer OC, den Dunnen JT, van Ommen GJ, Aartsma-Rus A, van Roon-Mom WM. Preventing formation of toxic N-terminal huntingtin fragments through antisense oligonucleotide-mediated protein modification. Nucleic Acid Ther. 2014;24(1):4–12. doi: 10.1089/nat.2013.0452. PMID: 24380395. [PubMed: 24380395]
- Evers MM, Tran HD, Zalachoras I, Pepers BA, Meijer OC, den Dunnen JT, van Ommen GJ, Aartsma-Rus A, van Roon-Mom WM. Ataxin-3 protein modification as a treatment strategy for spinocerebellar ataxia type 3: removal of the CAG containing exon. Neurobiol Dis. 2013;58:49–56. doi: 10.1016/j.nbd.2013.04.019. PMID: 23659897. [PubMed: 23659897]
- Finkel RS, Chiriboga CA, Vajsar J, Day JW, Montes J, De Vivo DC, Yamashita M, Rigo F, Hung G, Schneider E, Norris DA, Xia S, Bennett CF, Bishop KM. Treatment of infantile-onset spinal muscular atrophy with nusinersen: a phase 2, open-label, dose-escalation study. Lancet. 2016;388(10063):3017–26. doi: 10.1016/S0140-6736(16)31408-8. PMID: 27939059. [PubMed: 27939059]
- Finkel RS, Mercuri E, Darras BT, Connolly AM, Kuntz NL, Kirschner J, Chiriboga CA, Saito K, Servais L, Tizzano E, Topaloglu H, Tulinius M, Montes J, Glanzman AM, Bishop K, Zhong ZJ, Gheuens S, Bennett CF, Schneider E, Farwell W, De Vivo DC, Group ES. Nusinersen versus Sham Control in Infantile-Onset Spinal Muscular Atrophy. N Engl J Med. 2017;377(18):1723–32. doi: 10.1056/NEJMoa1702752. PMID: 29091570. [PubMed: 29091570]
- Gagliardi M, Ashizawa AT. The Challenges and Strategies of Antisense Oligonucleotide Drug Delivery. Biomedicines. 2021;9(4). doi: 10.3390/biomedicines9040433. PMID: 33923688. [PMC free article: PMC8072990] [PubMed: 33923688]
- Geraets RD, Koh S, Hastings ML, Kielian T, Pearce DA, Weimer JM. Moving towards effective therapeutic strategies for Neuronal Ceroid Lipofuscinosis. Orphanet J Rare Dis. 2016;11:40. doi: 10.1186/s13023-016-0414-2. PMID: 27083890. [PMC free article: PMC4833901] [PubMed: 27083890]
- Gumusgoz E, Guisso DR, Kasiri S, Wu J, Dear M, Verhalen B, Nitschke S, Mitra S, Nitschke F, Minassian BA. Targeting Gys1 with AAV-SaCas9 Decreases Pathogenic Polyglucosan Bodies and Neuroinflammation in Adult Polyglucosan Body and Lafora Disease Mouse Models. Neurotherapeutics. 2021;18(2):1414–25. doi: 10.1007/s13311-021-01040-7. PMID: 33830476. [PMC free article: PMC8423949] [PubMed: 33830476]
- Hamanaka K, Imagawa E, Koshimizu E, Miyatake S, Tohyama J, Yamagata T, Miyauchi A, Ekhilevitch N, Nakamura F, Kawashima T, Goshima Y, Mohamed AR, Ch’ng GS, Fujita A, Azuma Y, Yasuda K, Imamura S, Nakashima M, Saitsu H, Mitsuhashi S, Mizuguchi T, Takata A, Miyake N, Matsumoto N. De Novo Truncating Variants in the Last Exon of SEMA6B Cause Progressive Myoclonic Epilepsy. Am J Hum Genet. 2020;106(4):549–58. doi: 10.1016/j.ajhg.2020.02.011. PMID: 32169168. [PMC free article: PMC7118575] [PubMed: 32169168]
- Han Z, Chen C, Christiansen A, Ji S, Lin Q, Anumonwo C, Liu C, Leiser SC, Meena, Aznarez I, Liau G, Isom LL. Antisense oligonucleotides increase Scn1a expression and reduce seizures and SUDEP incidence in a mouse model of Dravet syndrome. Sci Transl Med. 2020;12(558). doi: 10.1126/scitranslmed.aaz6100. PMID: 32848094. [PubMed: 32848094]
- Hill SF, Meisler MH. Antisense Oligonucleotide Therapy for Neurodevelopmental Disorders. Dev Neurosci. 2021;43(3-4):247–52. doi: 10.1159/000517686. PMID: 34412058. [PMC free article: PMC8440367] [PubMed: 34412058]
- Hillman RT, Green RE, Brenner SE. An unappreciated role for RNA surveillance. Genome Biol. 2004;5(2):R8. doi: 10.1186/gb-2004-5-2-r8. PMID: 14759258. [PMC free article: PMC395752] [PubMed: 14759258]
- Holmes GL. Drug Treatment of Progressive Myoclonic Epilepsy. Paediatr Drugs. 2020;22(2):149–64. doi: 10.1007/s40272-019-00378-y. PMID: 31939107. [PMC free article: PMC7901803] [PubMed: 31939107]
- Hruska KS, LaMarca ME, Scott CR, Sidransky E. Gaucher disease: mutation and polymorphism spectrum in the glucocerebrosidase gene (GBA). Hum Mutat. 2008;29(5):567–83. doi: 10.1002/humu.20676. PMID: 18338393. [PubMed: 18338393]
- Huang L, Aghajan M, Quesenberry T, Low A, Murray SF, Monia BP, Guo S. Targeting Translation Termination Machinery with Antisense Oligonucleotides for Diseases Caused by Nonsense Mutations. Nucleic Acid Ther. 2019;29(4):175–86. doi: 10.1089/nat.2019.0779. PMID: 31070517. [PMC free article: PMC6686700] [PubMed: 31070517]
- Israelian L, Nitschke S, Wang P, Zhao X, Perri AM, Lee JPY, Verhalen B, Nitschke F, Minassian BA. Ppp1r3d deficiency preferentially inhibits neuronal and cardiac Lafora body formation in a mouse model of the fatal epilepsy Lafora disease. J Neurochem. 2020. doi: 10.1111/jnc.15176. PMID: 32892347. [PMC free article: PMC7936008] [PubMed: 32892347]
- Kälviäinen R. Progressive Myoclonus Epilepsies. Semin Neurol. 2015;35(3):293–9. doi: 10.1055/s-0035-1552620. PMID: 26060909. [PubMed: 26060909]
- Keeling KM, Bedwell DM. Suppression of nonsense mutations as a therapeutic approach to treat genetic diseases. Wiley Interdiscip Rev RNA. 2011;2(6):837–52. doi: 10.1002/wrna.95. PMID: 21976286. [PMC free article: PMC3188951] [PubMed: 21976286]
- Keeling KM, Wang D, Dai Y, Murugesan S, Chenna B, Clark J, Belakhov V, Kandasamy J, Velu SE, Baasov T, Bedwell DM. Attenuation of nonsense-mediated mRNA decay enhances in vivo nonsense suppression. PLoS One. 2013;8(4):e60478. doi: 10.1371/journal.pone.0060478. PMID: 23593225. [PMC free article: PMC3622682] [PubMed: 23593225]
- Keenan MM, Huang L, Jordan NJ, Wong E, Cheng Y, Valley HC, Mahiou J, Liang F, Bihler H, Mense M, Guo S, Monia BP. Nonsense-mediated RNA Decay Pathway Inhibition Restores Expression and Function of W1282X CFTR. Am J Respir Cell Mol Biol. 2019;61(3):290–300. doi: 10.1165/rcmb.2018-0316OC. PMID: 30836009. [PubMed: 30836009]
- Kim J, Hu C, Moufawad El Achkar C, Black LE, Douville J, Larson A, Pendergast MK, Goldkind SF, Lee EA, Kuniholm A, Soucy A, Vaze J, Belur NR, Fredriksen K, Stojkovska I, Tsytsykova A, Armant M, DiDonato RL, Choi J, Cornelissen L, Pereira LM, Augustine EF, Genetti CA, Dies K, Barton B, Williams L, Goodlett BD, Riley BL, Pasternak A, Berry ER, Pflock KA, Chu S, Reed C, Tyndall K, Agrawal PB, Beggs AH, Grant PE, Urion DK, Snyder RO, Waisbren SE, Poduri A, Park PJ, Patterson A, Biffi A, Mazzulli JR, Bodamer O, Berde CB, Yu TW. Patient-Customized Oligonucleotide Therapy for a Rare Genetic Disease. N Engl J Med. 2019;381(17):1644–52. doi: 10.1056/NEJMoa1813279. PMID: 31597037. [PMC free article: PMC6961983] [PubMed: 31597037]
- Kordasiewicz HB, Stanek LM, Wancewicz EV, Mazur C, McAlonis MM, Pytel KA, Artates JW, Weiss A, Cheng SH, Shihabuddin LS, Hung G, Bennett CF, Cleveland DW. Sustained therapeutic reversal of Huntington’s disease by transient repression of huntingtin synthesis. Neuron. 2012;74(6):1031–1044. doi: 10.1016/j.neuron.2012.05.009. PMID: 22726834. [PMC free article: PMC3383626] [PubMed: 22726834]
- Kousi M, Lehesjoki AE, Mole SE. Update of the mutation spectrum and clinical correlations of over 360 mutations in eight genes that underlie the neuronal ceroid lipofuscinoses. Hum Mutat. 2012;33(1):42–63. doi: 10.1002/humu.21624. PMID: 21990111. [PubMed: 21990111]
- Kuijper EC, Bergsma AJ, Pijnappel W, Aartsma-Rus A. Opportunities and challenges for antisense oligonucleotide therapies. J Inherit Metab Dis. 2021;44(1):72–87. doi: 10.1002/jimd.12251. PMID: 32391605. [PMC free article: PMC7891411] [PubMed: 32391605]
- Liang XH, Shen W, Sun H, Migawa MT, Vickers TA, Crooke ST. Translation efficiency of mRNAs is increased by antisense oligonucleotides targeting upstream open reading frames. Nat Biotechnol. 2016;34(8):875–80. doi: 10.1038/nbt.3589. PMID: 27398791. [PubMed: 27398791]
- Liang XH, Sun H, Shen W, Wang S, Yao J, Migawa MT, Bui HH, Damle SS, Riney S, Graham MJ, Crooke RM, Crooke ST. Antisense oligonucleotides targeting translation inhibitory elements in 5’ UTRs can selectively increase protein levels. Nucleic Acids Res. 2017;45(16):9528–46. doi: 10.1093/nar/gkx632. PMID: 28934489. [PMC free article: PMC5766168] [PubMed: 28934489]
- Lieberman AP, Shakkottai VG, Albin RL. Polyglutamine Repeats in Neurodegenerative Diseases. Annu Rev Pathol. 2019;14:1–27. doi: 10.1146/annurev-pathmechdis-012418-012857. PMID: 30089230. [PMC free article: PMC6387631] [PubMed: 30089230]
- Lim KH, Ferraris L, Filloux ME, Raphael BJ, Fairbrother WG. Using positional distribution to identify splicing elements and predict pre-mRNA processing defects in human genes. Proc Natl Acad Sci U S A. 2011;108(27):11093–11098. doi: 10.1073/pnas.1101135108. PMID: 21685335. [PMC free article: PMC3131313] [PubMed: 21685335]
- Lim KH, Han Z, Jeon HY, Kach J, Jing E, Weyn-Vanhentenryck S, Downs M, Corrionero A, Oh R, Scharner J, Venkatesh A, Ji S, Liau G, Ticho B, Nash H, Aznarez I. Antisense oligonucleotide modulation of non-productive alternative splicing upregulates gene expression. Nat Commun. 2020;11(1):3501. doi: 10.1038/s41467-020-17093-9. PMID: 32647108. [PMC free article: PMC7347940] [PubMed: 32647108]
- Lima JF, Cerqueira L, Figueiredo C, Oliveira C, Azevedo NF. Anti-miRNA oligonucleotides: A comprehensive guide for design. RNA Biol. 2018;15(3):338–52. doi: 10.1080/15476286.2018.1445959. PMID: 29570036. [PMC free article: PMC5927725] [PubMed: 29570036]
- Martin L, Grigoryan A, Wang D, Wang J, Breda L, Rivella S, Cardozo T, Gardner LB. Identification and characterization of small molecules that inhibit nonsense-mediated RNA decay and suppress nonsense p53 mutations. Cancer Res. 2014;74(11):3104–13. doi: 10.1158/0008-5472.CAN-13-2235. PMID: 24662918. [PMC free article: PMC4040335] [PubMed: 24662918]
- Masten MC, Mink JW, Augustine EF. Batten disease: an expert update on agents in preclinical and clinical trials. Expert Opin Investig Drugs. 2020;29(12):1317–22. doi: 10.1080/13543784.2020.1837110. PMID: 33135495. [PMC free article: PMC7736171] [PubMed: 33135495]
- Matos L, Duarte AJ, Ribeiro D, Chaves J, Amaral O, Alves S. Correction of a Splicing Mutation Affecting an Unverricht-Lundborg Disease Patient by Antisense Therapy. Genes (Basel). 2018;9(9):455. doi: 10.3390/genes9090455. PMID: 30208654. [PMC free article: PMC6162617] [PubMed: 30208654]
- Mazur C, Powers B, Zasadny K, Sullivan JM, Dimant H, Kamme F, Hesterman J, Matson J, Oestergaard M, Seaman M, Holt RW, Qutaish M, Polyak I, Coelho R, Gottumukkala V, Gaut CM, Berridge M, Albargothy NJ, Kelly L, Carare RO, Hoppin J, Kordasiewicz H, Swayze EE, Verma A. Brain pharmacology of intrathecal antisense oligonucleotides revealed through multimodal imaging. JCI Insight. 2019;4(20):e129240. doi: 10.1172/jci.insight.129240. PMID: 31619586. [PMC free article: PMC6824309] [PubMed: 31619586]
- McGlincy NJ, Smith CW. Alternative splicing resulting in nonsense-mediated mRNA decay: what is the meaning of nonsense? Trends Biochem Sci. 2008;33(8):385–393. doi: 10.1016/j.tibs.2008.06.001. PMID: 18621535. [PubMed: 18621535]
- Mendell JR, Goemans N, Lowes LP, Alfano LN, Berry K, Shao J, Kaye EM, Mercuri E, Eteplirsen Study G, Telethon Foundation DMDIN. Longitudinal effect of eteplirsen versus historical control on ambulation in Duchenne muscular dystrophy. Ann Neurol. 2016;79(2):257–71. doi: 10.1002/ana.24555. PMID: 26573217. [PMC free article: PMC5064753] [PubMed: 26573217]
- Mercuri E, Darras BT, Chiriboga CA, Day JW, Campbell C, Connolly AM, Iannaccone ST, Kirschner J, Kuntz NL, Saito K, Shieh PB, Tulinius M, Mazzone ES, Montes J, Bishop KM, Yang Q, Foster R, Gheuens S, Bennett CF, Farwell W, Schneider E, De Vivo DC, Finkel RS, Group CS. Nusinersen versus Sham Control in Later-Onset Spinal Muscular Atrophy. N Engl J Med. 2018;378(7):625–35. doi: 10.1056/NEJMoa1710504. PMID: 29443664. [PubMed: 29443664]
- Mercuri E, Pera MC, Scoto M, Finkel R, Muntoni F. Spinal muscular atrophy—insights and challenges in the treatment era. Nat Rev Neurol. 2020;16(12):706–15. doi: 10.1038/s41582-020-00413-4. PMID: 33057172. [PubMed: 33057172]
- Miller JN, Chan CH, Pearce DA. The role of nonsense-mediated decay in neuronal ceroid lipofuscinosis. Hum Mol Genet. 2013;22(13):2723–34. doi: 10.1093/hmg/ddt120. PMID: 23539563. [PMC free article: PMC4439521] [PubMed: 23539563]
- Miller JN, Kovacs AD, Pearce DA. The novel Cln1(R151X) mouse model of infantile neuronal ceroid lipofuscinosis (INCL) for testing nonsense suppression therapy. Hum Mol Genet. 2015;24(1):185–96. doi: 10.1093/hmg/ddu428. PMID: 25205113. [PMC free article: PMC4326326] [PubMed: 25205113]
- Minassian BA, Lee JR, Herbrick JA, Huizenga J, Soder S, Mungall AJ, Dunham I, Gardner R, Fong CY, Carpenter S, Jardim L, Satishchandra P, Andermann E, Snead OC, 3rd, Lopes-Cendes I, Tsui LC, Delgado-Escueta AV, Rouleau GA, Scherer SW. Mutations in a gene encoding a novel protein tyrosine phosphatase cause progressive myoclonus epilepsy. Nat Genet. 1998;20(2):171–4. doi: 10.1038/2470. PMID: 9771710. [PubMed: 9771710]
- Minassian BA, Striano P, Avanzini G, editors. Progressive Myoclonus Epilepsies: State-of-the-Art. Epileptic Disorders. 2016;18(2):1–164. [PMC free article: PMC5777179] [PubMed: 27621064]
- Mirza M, Vainshtein A, DiRonza A, Chandrachud U, Haslett LJ, Palmieri M, Storch S, Groh J, Dobzinski N, Napolitano G, Schmidtke C, Kerkovich DM. The CLN3 gene and protein: What we know. Mol Genet Genomic Med. 2019;7(12):e859. doi: 10.1002/mgg3.859. PMID: 31568712. [PMC free article: PMC6900386] [PubMed: 31568712]
- Mort M, Ivanov D, Cooper DN, Chuzhanova NA. A meta-analysis of nonsense mutations causing human genetic disease. Hum Mutat. 2008;29(8):1037–47. doi: 10.1002/humu.20763. PMID: 18454449. [PubMed: 18454449]
- Mosca R, van de Vlekkert D, Campos Y, Fremuth LE, Cadaoas J, Koppaka V, Kakkis E, Tifft C, Toro C, Allievi S, Gellera C, Canafoglia L, Visser G, Annunziata I, d’Azzo A. Conventional and Unconventional Therapeutic Strategies for Sialidosis Type I. J Clin Med. 2020;9(3). doi: 10.3390/jcm9030695. PMID: 32143456. [PMC free article: PMC7141319] [PubMed: 32143456]
- Munroe PB, Mitchison HM, O’Rawe AM, Anderson JW, Boustany RM, Lerner TJ, Taschner PE, de Vos N, Breuning MH, Gardiner RM, Mole SE. Spectrum of mutations in the Batten disease gene, CLN3. Am J Hum Genet. 1997;61(2):310–6. doi: 10.1086/514846. PMID: 9311735. [PMC free article: PMC1715900] [PubMed: 9311735]
- Muona M, Berkovic SF, Dibbens LM, Oliver KL, Maljevic S, Bayly MA, Joensuu T, Canafoglia L, Franceschetti S, Michelucci R, Markkinen S, Heron SE, Hildebrand MS, Andermann E, Andermann F, Gambardella A, Tinuper P, Licchetta L, Scheffer IE, Criscuolo C, Filla A, Ferlazzo E, Ahmad J, Ahmad A, Baykan B, Said E, Topcu M, Riguzzi P, King MD, Ozkara C, Andrade DM, Engelsen BA, Crespel A, Lindenau M, Lohmann E, Saletti V, Massano J, Privitera M, Espay AJ, Kauffmann B, Duchowny M, Moller RS, Straussberg R, Afawi Z, Ben-Zeev B, Samocha KE, Daly MJ, Petrou S, Lerche H, Palotie A, Lehesjoki AE. A recurrent de novo mutation in KCNC1 causes progressive myoclonus epilepsy. Nat Genet. 2015;47(1):39–46. doi: 10.1038/ng.3144. PMID: 25401298. [PMC free article: PMC4281260] [PubMed: 25401298]
- Naseri N, Sharma M, Velinov M. Autosomal dominant neuronal ceroid lipofuscinosis: Clinical features and molecular basis. Clin Genet. 2021;99(1):111–8. doi: 10.1111/cge.13829. PMID: 32783189. [PMC free article: PMC7899141] [PubMed: 32783189]
- Nickless A, Bailis JM, You Z. Control of gene expression through the nonsense-mediated RNA decay pathway. Cell Biosci. 2017;7:26. doi: 10.1186/s13578-017-0153-7. PMID: 28533900. [PMC free article: PMC5437625] [PubMed: 28533900]
- Nitschke F, Ahonen SJ, Nitschke S, Mitra S, Minassian BA. Lafora disease—from pathogenesis to treatment strategies. Nat Rev Neurol. 2018;14(10):606–17. doi: 10.1038/s41582-018-0057-0. PMID: 30143794. [PMC free article: PMC6317072] [PubMed: 30143794]
- Nitschke S, Chown EE, Zhao X, Gabrielian S, Petkovic S, Guisso DR, Perri AM, Wang P, Ahonen S, Nitschke F, Minassian BA. An inducible glycogen synthase-1 knockout halts but does not reverse Lafora disease progression in mice. J Biol Chem. 2021;296:100150. doi: 10.1074/jbc.RA120.015773. PMID: 33277363. [PMC free article: PMC7857511] [PubMed: 33277363]
- Nomakuchi TT, Rigo F, Aznarez I, Krainer AR. Antisense oligonucleotide-directed inhibition of nonsense-mediated mRNA decay. Nat Biotechnol. 2016;34(2):164–6. doi: 10.1038/nbt.3427. PMID: 26655495. [PMC free article: PMC4744113] [PubMed: 26655495]
- Orsini A, Valetto A, Bertini V, Esposito M, Carli N, Minassian BA, Bonuccelli A, Peroni D, Michelucci R, Striano P. The best evidence for progressive myoclonic epilepsy: A pathway to precision therapy. Seizure. 2019;71:247–57. doi: 10.1016/j.seizure.2019.08.012. PMID: 31476531. [PMC free article: PMC7288863] [PubMed: 31476531]
- Paron F, Dardis A, Buratti E. Pre-mRNA splicing defects and RNA binding protein involvement in Niemann Pick type C disease. J Biotechnol. 2020;318:20–30. doi: 10.1016/j.jbiotec.2020.03.012. PMID: 32387451. [PubMed: 32387451]
- Pattison S, Pankarican M, Rupar CA, Graham FL, Igdoura SA. Five novel mutations in the lysosomal sialidase gene (NEU1) in type II sialidosis patients and assessment of their impact on enzyme activity and intracellular targeting using adenovirus-mediated expression. Hum Mutat. 2004;23(1):32–9. doi: 10.1002/humu.10278. PMID: 14695530. [PubMed: 14695530]
- Pederson BA, Turnbull J, Epp JR, Weaver SA, Zhao X, Pencea N, Roach PJ, Frankland PW, Ackerley CA, Minassian BA. Inhibiting glycogen synthesis prevents lafora disease in a mouse model. Annals of Neurology. 2013;74(2):297–300. doi: 10.1002/ana.23899. [PMC free article: PMC3823666] [PubMed: 23913475]
- Penzel R, Uhl J, Kopitz J, Beck M, Otto HF, Cantz M. Splice donor site mutation in the lysosomal neuraminidase gene causing exon skipping and complete loss of enzyme activity in a sialidosis patient. FEBS Lett. 2001;501(2–3):135–8. doi: 10.1016/s0014-5793(01)02645-x. PMID: 11470272. [PubMed: 11470272]
- Rigo F, Chun SJ, Norris DA, Hung G, Lee S, Matson J, Fey RA, Gaus H, Hua Y, Grundy JS, Krainer AR, Henry SP, Bennett CF. Pharmacology of a central nervous system delivered 2’-O-methoxyethyl-modified survival of motor neuron splicing oligonucleotide in mice and nonhuman primates. J Pharmacol Exp Ther. 2014;350(1):46–55. doi: 10.1124/jpet.113.212407. PMID: 24784568. [PMC free article: PMC4056267] [PubMed: 24784568]
- Riley BE, Orr HT. Polyglutamine neurodegenerative diseases and regulation of transcription: assembling the puzzle. Genes Dev. 2006;20(16):2183–92. doi: 10.1101/gad.1436506. PMID: 16912271. [PubMed: 16912271]
- Rimkunas VM, Graham MJ, Crooke RM, Liscum L. In vivo antisense oligonucleotide reduction of NPC1 expression as a novel mouse model for Niemann Pick type C- associated liver disease. Hepatology. 2008;47(5):1504–12. doi: 10.1002/hep.22327. PMID: 18438776. [PMC free article: PMC2440313] [PubMed: 18438776]
- Roberts TC, Langer R, Wood MJA. Advances in oligonucleotide drug delivery. Nat Rev Drug Discov. 2020;19(10):673–94. doi: 10.1038/s41573-020-0075-7. PMID: 32782413. [PMC free article: PMC7419031] [PubMed: 32782413]
- Rodriguez-Pascau L, Coll MJ, Vilageliu L, Grinberg D. Antisense oligonucleotide treatment for a pseudoexon-generating mutation in the NPC1 gene causing Niemann-Pick type C disease. Hum Mutat. 2009;30(11):E993–E1001. doi: 10.1002/humu.21119. PMID: 19718781. [PubMed: 19718781]
- Rotllan N, Ramirez CM, Aryal B, Esau CC, Fernandez-Hernando C. Therapeutic silencing of microRNA-33 inhibits the progression of atherosclerosis in Ldlr-/- mice--brief report. Arterioscler Thromb Vasc Biol. 2013;33(8):1973–7. doi: 10.1161/ATVBAHA.113.301732. PMID: 23702658. [PMC free article: PMC4157595] [PubMed: 23702658]
- Roussel BD, Lomas DA, Crowther DC. Progressive myoclonus epilepsy associated with neuroserpin inclusion bodies (neuroserpinosis). Epileptic Disord. 2016;18(S2):103–10. doi: 10.1684/epd.2016.0847. PMID: 27618835. [PubMed: 27618835]
- Sarkar C, Zhang Z, Mukherjee AB. Stop codon read-through with PTC124 induces palmitoyl-protein thioesterase-1 activity, reduces thioester load and suppresses apoptosis in cultured cells from INCL patients. Mol Genet Metab. 2011;104(3):338–45. doi: 10.1016/j.ymgme.2011.05.021. PMID: 21704547. [PMC free article: PMC3220191] [PubMed: 21704547]
- Schoch KM, Miller TM. Antisense Oligonucleotides: Translation from Mouse Models to Human Neurodegenerative Diseases. Neuron. 2017;94(6):1056–70. doi: 10.1016/j.neuron.2017.04.010. PMID: 28641106. [PMC free article: PMC5821515] [PubMed: 28641106]
- Scoles DR, Minikel EV, Pulst SM. Antisense oligonucleotides: A primer. Neurol Genet. 2019;5(2):e323. doi: 10.1212/NXG.0000000000000323. PMID: 31119194. [PMC free article: PMC6501637] [PubMed: 31119194]
- Sharma VK, Watts JK. Oligonucleotide therapeutics: chemistry, delivery and clinical progress. Future Med Chem. 2015;7(16):2221–42. doi: 10.4155/fmc.15.144. PMID: 26510815. [PubMed: 26510815]
- Shen Y, Lee G, Choe Y, Zoltewicz JS, Peterson AS. Functional architecture of atrophins. J Biol Chem. 2007;282(7):5037–44. doi: 10.1074/jbc.M610274200. PMID: 17150957. [PubMed: 17150957]
- Silva AC, Lobo DD, Martins IM, Lopes SM, Henriques C, Duarte SP, Dodart JC, Nobre RJ, Pereira de Almeida L. Antisense oligonucleotide therapeutics in neurodegenerative diseases: the case of polyglutamine disorders. Brain. 2020;143(2):407–29. doi: 10.1093/brain/awz328. PMID: 31738395. [PubMed: 31738395]
- Singh S, Ganesh S. Lafora progressive myoclonus epilepsy: a meta-analysis of reported mutations in the first decade following the discovery of the EPM2A and NHLRC1 genes. Hum Mutat. 2009;30(5):715–23. doi: 10.1002/humu.20954. PMID: 19267391. [PubMed: 19267391]
- Siva K, Covello G, Denti MA. Exon-skipping antisense oligonucleotides to correct missplicing in neurogenetic diseases. Nucleic Acid Ther. 2014;24(1):69–86. doi: 10.1089/nat.2013.0461. PMID: 24506781. [PMC free article: PMC3922311] [PubMed: 24506781]
- Sleat DE, Sohar I, Gin RM, Lobel P. Aminoglycoside-mediated suppression of nonsense mutations in late infantile neuronal ceroid lipofuscinosis. Eur J Paediatr Neurol. 2001;5 Suppl A:57–62. doi: 10.1053/ejpn.2000.0436. PMID: 11589009. [PubMed: 11589009]
- Southwell AL, Kordasiewicz HB, Langbehn D, Skotte NH, Parsons MP, Villanueva EB, Caron NS, Ostergaard ME, Anderson LM, Xie Y, Cengio LD, Findlay-Black H, Doty CN, Fitsimmons B, Swayze EE, Seth PP, Raymond LA, Frank Bennett C, Hayden MR. Huntingtin suppression restores cognitive function in a mouse model of Huntington’s disease. Sci Transl Med. 2018;10(461):eaar3959. doi: 10.1126/scitranslmed.aar3959. PMID: 30282695. [PubMed: 30282695]
- Sterne-Weiler T, Howard J, Mort M, Cooper DN, Sanford JR. Loss of exon identity is a common mechanism of human inherited disease. Genome Res. 2011;21(10):1563–71. doi: 10.1101/gr.118638.110. PMID: 21750108. [PMC free article: PMC3202274] [PubMed: 21750108]
- Sterne-Weiler T, Sanford JR. Exon identity crisis: disease-causing mutations that disrupt the splicing code. Genome Biol. 2014;15(1):201. doi: 10.1186/gb4150. PMID: 24456648. [PMC free article: PMC4053859] [PubMed: 24456648]
- Sullivan M, Nitschke S, Steup M, Minassian B, Nitschke F. Pathogenesis of Lafora Disease: Transition of Soluble Glycogen to Insoluble Polyglucosan. International Journal of Molecular Sciences. 2017;18(8):1743. doi: 10.3390/ijms18081743. PMID: 28800070. [PMC free article: PMC5578133] [PubMed: 28800070]
- Thada V, Miller JN, Kovacs AD, Pearce DA. Tissue-specific variation in nonsense mutant transcript level and drug-induced read-through efficiency in the Cln1(R151X) mouse model of INCL. J Cell Mol Med. 2016;20(2):381–385. doi: 10.1111/jcmm.12744. PMID: 26648046. [PMC free article: PMC4727554] [PubMed: 26648046]
- Tonin R, Catarzi S, Caciotti A, Procopio E, Marini C, Guerrini R, Morrone A. Progressive myoclonus epilepsy in Gaucher Disease due to a new Gly-Gly mutation causing loss of an Exonic Splicing Enhancer. J Neurol. 2019;266(1):92–101. doi: 10.1007/s00415-018-9084-4. PMID: 30382391. [PMC free article: PMC6342868] [PubMed: 30382391]
- Tsuji S. Dentatorubral-pallidoluysian atrophy. Handb Clin Neurol. 2012;103:587–94. doi: 10.1016/B978-0-444-51892-7.00041-3. PMID: 21827919. [PubMed: 21827919]
- Turnbull J, DePaoli-Roach AA, Zhao X, Cortez MA, Pencea N, Tiberia E, Piliguian M, Roach PJ, Wang P, Ackerley CA, Minassian BA. PTG depletion removes Lafora bodies and rescues the fatal epilepsy of Lafora disease. PLoS Genet. 2011;7(4):e1002037. doi: 10.1371/journal.pgen.1002037. PMID: 21552327. [PMC free article: PMC3084203] [PubMed: 21552327]
- Turnbull J, Epp JR, Goldsmith D, Zhao X, Pencea N, Wang P, Frankland PW, Ackerley CA, Minassian BA. PTG protein depletion rescues malin-deficient Lafora disease in mouse. Annals of Neurology. 2014;75(3):442–6. doi: 10.1002/ana.24104. PMID: 24419970. [PubMed: 24419970]
- Varea O, Duran J, Aguilera M, Prats N, Guinovart JJ. Suppression of glycogen synthesis as a treatment for Lafora disease: Establishing the window of opportunity. Neurobiol Dis. 2021;147:105173. doi: 10.1016/j.nbd.2020.105173. PMID: 33171226. [PMC free article: PMC7736642] [PubMed: 33171226]
- Verma A. Recent Advances in Antisense Oligonucleotide Therapy in Genetic Neuromuscular Diseases. Ann Indian Acad Neurol. 2018;21(1):3–8. doi: 10.4103/aian.AIAN_298_17. PMID: 29720791. [PMC free article: PMC5909143] [PubMed: 29720791]
- Wang ET, Sandberg R, Luo S, Khrebtukova I, Zhang L, Mayr C, Kingsmore SF, Schroth GP, Burge CB. Alternative isoform regulation in human tissue transcriptomes. Nature. 2008;456(7221):470–6. doi: 10.1038/nature07509. PMID: 18978772. [PMC free article: PMC2593745] [PubMed: 18978772]
- Ward AJ, Cooper TA. The pathobiology of splicing. J Pathol. 2010;220(2):152–63. doi: 10.1002/path.2649. PMID: 19918805. [PMC free article: PMC2855871] [PubMed: 19918805]
- Xiong H, Veedu RN, Diermeier SD. Recent Advances in Oligonucleotide Therapeutics in Oncology. Int J Mol Sci. 2021;22(7). doi: 10.3390/ijms22073295. PMID: 33804856. [PMC free article: PMC8036554] [PubMed: 33804856]
- Yari A, Ali-Nejad RM, Saleh-Gohari N. A novel homozygous splice-site mutation in SCARB2 is associated with progressive myoclonic epilepsy with renal failure. Neurol Sci. 2021. doi: 10.1007/s10072-021-05196-0. PMID: 33772352. [PubMed: 33772352]
- Yu J, Ying M, Zhuang Y, Xu T, Han M, Wu X, Xu R. C-terminal deletion of the atrophin-1 protein results in growth retardation but not neurodegeneration in mice. Dev Dyn. 2009;238(10):2471–8. doi: 10.1002/dvdy.22063. PMID: 19681162. [PubMed: 19681162]
- Zech M, Nubling G, Castrop F, Jochim A, Schulte EC, Mollenhauer B, Lichtner P, Peters A, Gieger C, Marquardt T, Vanier MT, Latour P, Klunemann H, Trenkwalder C, Diehl-Schmid J, Perneczky R, Meitinger T, Oexle K, Haslinger B, Lorenzl S, Winkelmann J. Niemann-Pick C disease gene mutations and age-related neurodegenerative disorders. PLoS One. 2013;8(12):e82879. doi: 10.1371/journal.pone.0082879. PMID: 24386122. [PMC free article: PMC3875432] [PubMed: 24386122]
- Zhou J, Tawk M, Tiziano FD, Veillet J, Bayes M, Nolent F, Garcia V, Servidei S, Bertini E, Castro-Giner F, Renda Y, Carpentier S, Andrieu-Abadie N, Gut I, Levade T, Topaloglu H, Melki J. Spinal muscular atrophy associated with progressive myoclonic epilepsy is caused by mutations in ASAH1. Am J Hum Genet. 2012; 91(1):5–14. doi: 10.1016/j.ajhg.2012.05.001. PMID: 22703880. [PMC free article: PMC3397266] [PubMed: 22703880]
- Review Epilepsy Genomics: Disease-Causing Sequence Variants.[Jasper's Basic Mechanisms of t...]Review Epilepsy Genomics: Disease-Causing Sequence Variants.Delgado-Escueta AV. Jasper's Basic Mechanisms of the Epilepsies. 2024
- Review The molecular genetic bases of the progressive myoclonus epilepsies.[Adv Neurol. 1999]Review The molecular genetic bases of the progressive myoclonus epilepsies.Serratosa JM, Gardiner RM, Lehesjoki AE, Pennacchio LA, Myers RM. Adv Neurol. 1999; 79:383-98.
- Review Strategies on Gene Therapy in Progressive Myoclonus Epilepsies.[Jasper's Basic Mechanisms of t...]Review Strategies on Gene Therapy in Progressive Myoclonus Epilepsies.Kayani S, Gumusgoz E, Minassian BA, Dolce A. Jasper's Basic Mechanisms of the Epilepsies. 2024
- Perampanel Improves Cortical Myoclonus and Disability in Progressive Myoclonic Epilepsies: A Case Series and a Systematic Review of the Literature.[Front Neurol. 2021]Perampanel Improves Cortical Myoclonus and Disability in Progressive Myoclonic Epilepsies: A Case Series and a Systematic Review of the Literature.Assenza G, Nocerino C, Tombini M, Di Gennaro G, D'Aniello A, Verrotti A, Marrelli A, Ricci L, Lanzone J, Di Lazzaro V, et al. Front Neurol. 2021; 12:630366. Epub 2021 Mar 24.
- Review The best evidence for progressive myoclonic epilepsy: A pathway to precision therapy.[Seizure. 2019]Review The best evidence for progressive myoclonic epilepsy: A pathway to precision therapy.Orsini A, Valetto A, Bertini V, Esposito M, Carli N, Minassian BA, Bonuccelli A, Peroni D, Michelucci R, Striano P. Seizure. 2019 Oct; 71:247-257. Epub 2019 Aug 23.
- Antisense Oligonucleotide Therapy for Progressive Myoclonus Epilepsies - Jasper'...Antisense Oligonucleotide Therapy for Progressive Myoclonus Epilepsies - Jasper's Basic Mechanisms of the Epilepsies
Your browsing activity is empty.
Activity recording is turned off.
See more...