Acessibilidade / Reportar erro

Gene-based therapies for neuromuscular disorders

Terapias gênicas nas doenças neuromusculares

Abstract

Neuromuscular diseases (NMD) include a broad group of medical conditions with both acquired and genetic causes. In recent years, important advances have been made in the treatment of genetically caused NMD, and most of these advances are due to the implementation of therapies aimed at gene regulation. Among these therapies, gene replacement, small interfering RNA (siRNA), and antisense antinucleotides are the most promising approaches. More importantly, some of these therapies have already gained regulatory approval or are in the final stages of approval. The review focuses on motor neuron diseases, neuropathies, and Duchenne muscular dystrophy, summarizing the most recent developments in gene-based therapies for these conditions.

Keywords
Gene Therapy; Muscular Atrophy, Spinal; Amyotrophic Lateral Sclerosis; Amyloid Neuropathies; Muscular Dystrophy, Duchenne

Resumo

Doenças neuromusculares (DNM) compõem um grupo amplo de doenças de causa tanto adquiridas quanto genéticas. Nos últimos anos, importantes avanços ocorreram quanto ao tratamento das DNM de causa genética e grande parte desses avanços se deve à implementação de terapias voltadas para a modificação gênica. Dentre essas terapias, destacam-se as terapias de reposição gênica, uso de RNA de interferência, uso de antinucleotídeos antisense, entre outras. E, mais importante, algumas dessas terapias já se tornaram realidade na prática médica e já foram aprovadas, ou estão a poucos passos da aprovação, por órgãos governamentais regulatórios. Esta revisão aborda aspectos mais recentes quanto ao uso das terapias genéticas avançadas para algumas das formas mais comuns de DNM, em especial para doenças do neurônio motor (esclerose lateral amiotrófica e atrofia muscular espinhal), neuropatias e distrofia muscular de Duchenne.

Palavras-chave
Terapia Genética; Atrofia Muscular Espinhal; Esclerose Amiotrófica Lateral; Neuropatias Amiloides; Distrofia Muscular de Duchenne

INTRODUCTION

We have witnessed a real revolution in the therapeutic landscape of neuromuscular disorders (NMD) in the past 5–10 years.11 Landfeldt E. Gene Therapy for Neuromuscular Diseases: Health Economic Challenges and Future Perspectives. J Neuromuscul Dis 2022;9(06):675–688 This is particularly evident for monogenic conditions, where precise disease targets and mechanisms are well known. Diseases such as spinal muscular atrophy and hereditary transthyretin amyloidosis (ATTRv) are now treated effectively with multiple approved disease-modifying agents.22 Mendell JR, Al-Zaidy S, Shell R, et al. Single-Dose Gene-Replacement Therapy for Spinal Muscular Atrophy. N Engl J Med 2017; 377(18):1713–1722

3 Finkel RS, Mercuri E, Darras BT, et al; ENDEAR Study Group. Nusinersen versus Sham Control in Infantile-Onset Spinal Muscular Atrophy. N Engl J Med 2017;377(18):1723–1732

4 Baranello G, Darras BT, Day JW, et al; FIREFISH Working Group. Risdiplam in Type 1 Spinal Muscular Atrophy. N Engl J Med 2021; 384(10):915–923

5 Benson MD, Waddington-Cruz M, Berk JL, et al. Inotersen Treatment for Patients with Hereditary Transthyretin Amyloidosis. N Engl J Med 2018;379(01):22–31
-66 Adams D, Gonzalez-Duarte A, O’Riordan WD, et al. Patisiran, an RNAi Therapeutic, for Hereditary Transthyretin Amyloidosis. N Engl J Med 2018;379(01):11–21 These innovative drugs in NMD are called gene-based therapies and rely upon the modulation of genes, mRNA, and/or proteins.77 Rossor AM, Reilly MM, Sleigh JN. Antisense oligonucleotides and other genetic therapies made simple. Pract Neurol 2018;18(02): 126–131 There are three main classes of such therapies: antisense oligonucleotides (ASOs), small interfering RNAs (RNAi), and gene replacement therapy (GT).

ASOs are synthetic single-stranded nucleic acid sequences containing between 8 and 20 nucleotides, which can bind to specific RNA sequences and thus regulate gene expression. Such ASO-mediated gene regulation can be mediated by activation of RNAse-H, by blocking mRNA transcription at ribosomes, or by modifying gene splicing.88 Goyal N, Narayanaswami P. Making sense of antisense oligonucleotides: A narrative review. Muscle Nerve 2018;57(03):356–370

RNAi-based drugs are small non-coding double-stranded RNA molecules with 21 to 23 nucleotides. These agents are designed to target specific mRNAs, and cause gene silencing through a complex multienzymatic intracellular system (RNA-induced silencing complex).99 Lam JK, Chow MY, Zhang Y, Leung SW. siRNA Versus miRNA as Therapeutics for Gene Silencing. Mol Ther Nucleic Acids 2015;4 (09):e252 So far, RNAi-therapies have primarily been used intravenous (IV) or subcutaneously to treat autosomal dominant gain-of-function NMD (ATTRv and porphyrias).

GT consists of the insertion of genes into an individual's cells or tissues with the aim of treating or preventing a hereditary disease. There are two key components in all GT: the vector (which can be viral or non-viral) and the transgene (which includes the coding sequence of the gene of interest plus regulatory regions).1010 Kay MA. State-of-the-art gene-based therapies: the road ahead. Nat Rev Genet 2011;12(05):316–328 The choice of the transgene and the design of the transgene will depend on the condition treated. For NMDs, this is typically a single shot injection given in vivo either IV or intrathecal (IT).

GENE-BASED THERAPIES FOR AMYOTROPHIC LATERAL SCLEROSIS (ALS)

Amyotrophic lateral sclerosis (ALS) is the prototypical motor neuron disease in adults. The disease is characterized by degeneration of both upper and lower motor neurons, leading to progressive muscle weakness and atrophy.1111 Brown RH Jr, Al-Chalabi A. Amyotrophic Lateral Sclerosis. N Engl J Med 2017;377(16):1602,1212 Akçimen F, Lopez ER, Landers JE, et al. Amyotrophic lateral sclerosis: translating genetic discoveries into therapies. Nat Rev Genet 2023;24(09):642–658 Patients typically die 3 to 5 years after the onset of symptoms due to respiratory failure. Most patients with ALS (90%) have sporadic disease (sALS) with no known family history, whereas the remaining 5-10% are called familial ALS (fALS) because the disease segregates in other affected relatives. The genetic basis of ALS is now well characterized. fALS is considered a monogenic disorder with more than 20 distinct genes already associated. In contrast, monogenic sALS is rare; most patients indeed behave as oligo or polygenic conditions.1313 Chia R, Chiò A, Traynor BJ. Novel genes associated with amyotrophic lateral sclerosis: diagnostic and clinical implications. Lancet Neurol 2018;17(01):94–102 Most monogenic ALS subtypes segregate as autosomal dominant traits and are caused by gain-of-function missense variants (leading to abnormal protein folding and aggregation).1212 Akçimen F, Lopez ER, Landers JE, et al. Amyotrophic lateral sclerosis: translating genetic discoveries into therapies. Nat Rev Genet 2023;24(09):642–658,1313 Chia R, Chiò A, Traynor BJ. Novel genes associated with amyotrophic lateral sclerosis: diagnostic and clinical implications. Lancet Neurol 2018;17(01):94–102

Advanced gene-based therapies are coming to the clinical arena of ALS.1414 Miller TM, Cudkowicz ME, Genge A, et al; VALOR and OLE Working Group. Trial of Antisense Oligonucleotide Tofersen for SOD1 ALS. N Engl J Med 2022;387(12):1099–1110,1515 Mueller C, Berry JD, McKenna-Yasek DM, et al. SOD1 Suppression with Adeno-Associated Virus and MicroRNA in Familial ALS. N Engl J Med 2020;383(02):151–158 Essentially all drugs in development target one of the genes known to be associated with fALS. SOD1 was the first gene identified in fALS back in 1993.1616 Rosen DR, Siddique T, Patterson D, et al. Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature 1993;362(6415):59–62 For this reason, it is the leading gene when it comes to clinical drug development. Other genes such as C9orf72, FUS, and ATXN2, have been also explored lately. Further detail will be given below.

SOD1-ALS

Tofersen is an antisense oligonucleotide (ASO) administered intrathecally every month that targets both wild-type and mutant SOD1 alleles.1414 Miller TM, Cudkowicz ME, Genge A, et al; VALOR and OLE Working Group. Trial of Antisense Oligonucleotide Tofersen for SOD1 ALS. N Engl J Med 2022;387(12):1099–1110 It was tested in a phase 3 clinical trial that recruited 72 patients in a 2:1 ratio treatment versus placebo. They were followed over 24 weeks using the decline in the Amyotrophic Lateral Sclerosis Functional Rating Scale (ALSFRS) score which was the primary efficacy measure. In this time frame, the drug resulted in a significant decline in the cerebrospinal fluid concentrations of SOD1 and in the plasma concentration of neurofilament light chains, a marker of axonal degeneration. Furthermore, severe drug-related side effects were rather infrequent (7%). Despite that, the study did not meet its primary efficacy endpoint in the placebo-controlled phase. Long follow-up data from the open-label extension phase, however, found a clear positive trend towards the treatment. These results in combination enabled regulatory approval for tofersen to treat patients with SOD1-ALS in the US in 2023.1717 https://www.fda.gov/drugs/news-events-human-drugs/fda-approves-treatment-amyotrophic-lateral-sclerosis-associated-mutation-sod1-gene
https://www.fda.gov/drugs/news-events-hu...
There is an ongoing study focusing on presymptomatic carriers of SOD1 mutations treated with tofersen and followed in the long term.1818 Benatar M, Wuu J, Andersen PM, et al. Design of a Randomized, Placebo-Controlled, Phase 3 Trial of Tofersen Initiated in Clinically Presymptomatic SOD1 Variant Carriers: the ATLAS Study. Neurotherapeutics 2022;19(04):1248–1258

GT has been also explored in SOD1-ALS. In 2020, Mueller et reported two patients with SOD1-ALS and fast progression who underwent GT.1515 Mueller C, Berry JD, McKenna-Yasek DM, et al. SOD1 Suppression with Adeno-Associated Virus and MicroRNA in Familial ALS. N Engl J Med 2020;383(02):151–158 The investigational product was administered IT. AAVrh10 was the vector, and the transgene encoded a microRNA targeting SOD1 mRNA. The first treated patient did not receive prior immunosuppression, so he developed severe meningoradiculitis, and no obvious change in the progression was noticed. Post-mortem spinal cord samples from this patient indeed confirmed SOD1 silencing. In contrast, the second patient received IV steroids pre-GT, and the course was strikingly different remaining stable over 70 weeks (ALSFRS scores). These are encouraging results and other groups are now pursuing a similar approach to treat this monogenic subtype of ALS.1919 https://uniqure.gcs-web.com/news-releases/news-release-details/uniqure-and-apic-bio-enter-global-licensing-agreement-apb-102
https://uniqure.gcs-web.com/news-release...

Other genes (C9orf72, FUS, and ATXN2)

C9orf72 is the most frequent genetic cause of fALS in Caucasians.1111 Brown RH Jr, Al-Chalabi A. Amyotrophic Lateral Sclerosis. N Engl J Med 2017;377(16):1602 A phase 1 clinical trial using an IT ASO (BIIB078) targeting this gene was conducted.2020 https://investors.biogen.com/news-releases/news-release-details/biogen-and-ionis-announce-topline-phase-1-study-results
https://investors.biogen.com/news-releas...
The authors recruited and followed 106 patients who were randomized in a 3:1 ratio to treatment versus placebo. Unfortunately, no significant change was noticed between the treatment and placebo groups, which led to the discontinuation of drug development.2020 https://investors.biogen.com/news-releases/news-release-details/biogen-and-ionis-announce-topline-phase-1-study-results
https://investors.biogen.com/news-releas...

FUS causes a very aggressive and early-onset, sometimes juvenile, subtype of ALS.1111 Brown RH Jr, Al-Chalabi A. Amyotrophic Lateral Sclerosis. N Engl J Med 2017;377(16):1602 Jacifusen is a FUS-targeting ASO-administered IT that is currently under investigation at Columbia University. Some patients received the drug from 2019 to 2020 after FDA approval. Clinical benefit was not clear at this point, but post-mortem samples of these individuals confirmed the reduction of FUS expression in the spinal cord. A phase 3 clinical trial named FUSION is currently ongoing to assess the safety and efficacy profile of jacifusen in 64 patients with FUS-ALS over 29 weeks.2121 Boros BD, Schoch KM, Kreple CJ, Miller TM. Antisense Oligonucleotides for the Study and Treatment of ALS. Neurotherapeutics 2022;19(04):1145–1158

Intermediate CAG expansions at ATXN2 have been associated with ALS in multiple populations.2222 Tavares de Andrade HM, Cintra VP, de Albuquerque M, et al. Intermediate-length CAG repeat in ATXN2 is associated with increased risk for amyotrophic lateral sclerosis in Brazilian patients. Neurobiol Aging 2018;69:292.e15–292.e18 In contrast to other previously mentioned genes, this is not a causative gene, but rather a risk gene. On the other hand, its frequency is much higher in the overall ALS public compared to the other monogenic ALS subtypes. In the transgenic TDP43 ALS murine model, anti-ATXN2 ASOs rescued the phenotype and prolonged survival, particularly when given early.2323 Becker LA, Huang B, Bieri G, et al. Therapeutic reduction of ataxin-2 extends lifespan and reduces pathology in TDP-43 mice. Nature 2017;544(7650):367–371 A phase 1 clinical trial is ongoing to assess the safety and efficacy of IT anti-ATXN2 ASOs in patients with sporadic and ATXN2-related ALS.2121 Boros BD, Schoch KM, Kreple CJ, Miller TM. Antisense Oligonucleotides for the Study and Treatment of ALS. Neurotherapeutics 2022;19(04):1145–1158

SPINAL MUSCULAR ATROPHY LINKED TO 5q (SMA-5q)

Spinal muscular atrophy (SMA) is a neurodegenerative disease characterized by progressive muscle weakness, hypotonia, and weakness due to degeneration of motor neurons in the spinal cord and brainstem. The most common form of SMA is caused by recessive mutations in the survival motor neuron 1 (SMN1) gene located at 5q13 (SMA-5q).2424 Lefebvre S, Bürglen L, Reboullet S, et al. Identification and characterization of a spinal muscular atrophy-determining gene. Cell 1995;80(01):155–165 The global incidence of SMA is estimated at 1 in 10,000 live births.2525 Kolb SJ, Kissel JT. Spinal muscular atrophy: a timely review. Arch Neurol 2011;68(08):979–984

SMA has been classified into at least four subtypes depending on the patient's age at the disease's onset and the achievement of motor milestones.2626 Finkel R, Bertini E, Muntoni F, Mercuri EENMC SMA Workshop Study Group. 209th ENMC International Workshop: Outcome Measures and Clinical Trial Readiness in Spinal Muscular Atrophy 7-9 November 2014, Heemskerk, The Netherlands. Neuromuscul Disord 2015;25(07):593–602 In SMA type 1, or infantile form, children do not acquire the ability to sit, and the disease manifests at 0 to 6 months of age. In SMA type 2, clinical manifestations start between 6 and 18 months, and children are unable to walk unassisted. Children with SMA type 3 manifest the disease in the second year of life or later and can walk unaided. Finally, SMA type 4 is the adult form of the disease with the manifestations starting usually after the age of 18.

Exon 7 of the SMN1 is not detectable in approximately 96% of SMA-5q patients, and approximately 4% of patients have a combination of the deletion and an intragenic mutation in the second allele.2424 Lefebvre S, Bürglen L, Reboullet S, et al. Identification and characterization of a spinal muscular atrophy-determining gene. Cell 1995;80(01):155–165,2727 Mendonça RH, Matsui C Jr, Polido GJ, et al. Intragenic variants in the SMN1 gene determine the clinical phenotype in 5q spinal muscular atrophy. Neurol Genet 2020;6(05):e505 SMN2 is a centromeric copy of SMN1 that does not provide the transcription of stable SMN protein due to the lack of exon 7 in most transcripts.2424 Lefebvre S, Bürglen L, Reboullet S, et al. Identification and characterization of a spinal muscular atrophy-determining gene. Cell 1995;80(01):155–165 Several studies have demonstrated a strong inverse correlation between the number of SMN2 copies and SMA severity.2424 Lefebvre S, Bürglen L, Reboullet S, et al. Identification and characterization of a spinal muscular atrophy-determining gene. Cell 1995;80(01):155–165,2828 Calucho M, Bernal S, Alías L, et al. Correlation between SMA type and SMN2 copy number revisited: An analysis of 625 unrelated Spanish patients and a compilation of 2834 reported cases. Neuromuscul Disord 2018;28(03):208–215

Various therapeutic approaches are currently being developed for SMA. SMN-dependent therapies focus on addressing the SMN protein deficiency, such as gene therapy with SNM1 gene replacement (onasemnogene abeparvovec-AVXS101), and inclusion of exon 7 in SMN2 (nusinersen, risdiplam).2929 Chen TH. New and Developing Therapies in Spinal Muscular Atrophy: From Genotype to Phenotype to Treatment and Where Do We Stand? Int J Mol Sci 2020;21(09):3297 These therapies have already been approved by the leading international regulatory agencies and the Brazilian National Surveillance Agency (ANVISA).

Nusinersen (Spinraza®) is an antisense oligonucleotide (ASO) that targets an intronic splicing silencer site within the SMN2 pre-messenger RNA downstream of exon 7.3030 Chiriboga CA, Swoboda KJ, Darras BT, et al. Results from a phase 1 study of nusinersen (ISIS-SMN(Rx)) in children with spinal muscular atrophy. Neurology 2016;86(10):890–897,3131 Finkel RS, Chiriboga CA, Vajsar J, et al. Treatment of infantile-onset spinal muscular atrophy with nusinersen: a phase 2, open-label, dose-escalation study. Lancet 2016;388(10063):3017–3026 This targeting of the splicing silencer allows for increased inclusion of exon 7 during mRNA processing, producing more functional SMN protein from the SMN2 gene. As ASOs do not cross the blood-brain barrier, nusinersen must be administered via the intrathecal route.

The ENDEAR, a placebo-randomized clinical trial, demonstrated that nusinersen-treated SMA type 1 patients had more prolonged survival and significant improvements in motor function compared to those without treatment.33 Finkel RS, Mercuri E, Darras BT, et al; ENDEAR Study Group. Nusinersen versus Sham Control in Infantile-Onset Spinal Muscular Atrophy. N Engl J Med 2017;377(18):1723–1732 The CHERISH trial was a placebo-randomized study that enrolled 126 patients with late-onset SMA aged 2 to 12 years.3232 Mercuri E, Darras BT, Chiriboga CA, et al; CHERISH Study Group. Nusinersen versus sham control in later-onset spinal muscular atrophy. N Engl J Med 2018;378(07):625–635. Doi: 10.1056/ NEJMoa1710504
https://doi.org/10.1056/NEJMoa1710504...
The study demonstrated that patients who received Nusinersen experienced clinically significant improvements in motor function compared to the control group.

The follow-up of approximately three years of patients with SMA types 2 and 3 treated with nusinersen showed improvements in motor function and stabilization of disease activity.3333 Darras BT, Chiriboga CA, Iannaccone ST, et al; ISIS-396443-CS2/ ISIS-396443-CS12 Study Groups. Nusinersen in later-onset spinal muscular atrophy: Long-term results from the phase 1/2 studies. Neurology 2019;92(21):e2492–e2506. Doi: 10.1212/ WNL.0000000000007527
https://doi.org/10.1212/WNL.000000000000...
For SMA type 1, a follow-up of approximately 36.2 months showed a durable clinical response in a significant proportion of patients; 75% of the participants were still alive at the time of study closure.3434 Finkel RS, Chiriboga CA, Vajsar J, et al. Treatment of infantileonset spinal muscular atrophy with nusinersen: final report of a phase 2, open-label, multicentre, dose-escalation study. Lancet Child Adolesc Health 2021;5(07):491–500. Doi: 10.1016/S2352-4642(21)00100-0
https://doi.org/10.1016/S2352-4642(21)00...

Several subsequent real-life studies with SMA patients further confirmed the favorable effects of nusinersen on motor and respiratory function, as well as on the survival of patients with long-term illness and varying respiratory conditions.3535 Hagenacker T, Wurster CD, Günther R, et al. Nusinersen in adults with 5q spinal muscular atrophy: a non-interventional, multicentre, observational cohort study. Lancet Neurol 2020;19(04): 317–325. Doi: 10.1016/S1474-4422(20)30037-5
https://doi.org/10.1016/S1474-4422(20)30...

36 de Holanda Mendonça R, Jorge Polido G, Ciro M, Jorge Fontoura Solla D, Conti Reed U, Zanoteli E. Clinical Outcomes in Patients with Spinal Muscular Atrophy Type 1 Treated with Nusinersen. J Neuromuscul Dis 2021;8(02):217–224. Doi: 10.3233/JND-200533
https://doi.org/10.3233/JND-200533...

37 Mendonça RH, Polido GJ, Matsui C, et al. Real-World Data from Nusinersen Treatment for Patients with Later-Onset Spinal Muscular Atrophy: A Single Center Experience. J Neuromuscul Dis 2021;8(01):101–108. Doi: 10.3233/JND-200551
https://doi.org/10.3233/JND-200551...
-3838 Gavriilaki M, Moschou M, Papaliagkas V, et al. Nusinersen in Adults with 5q Spinal Muscular Atrophy: a Systematic Review and Meta-analysis. Neurotherapeutics 2022;19(02):464–475. Doi: 10.1007/s13311-022-01200-3
https://doi.org/10.1007/s13311-022-01200...

NURTURE is an ongoing phase 2, open-label study to evaluate the efficacy and safety of nusinersen in pre-symptomatic infants.3939 De Vivo DC, Bertini E, Swoboda KJ, et al; NURTURE Study Group. Nusinersen initiated in infants during the presymptomatic stage of spinal muscular atrophy: Interim efficacy and safety results from the Phase 2 NURTURE study. Neuromuscul Disord 2019;29 (11):842–856. Doi: 10.1016/j.nmd.2019.09.007
https://doi.org/10.1016/j.nmd.2019.09.00...
With a median follow-up of 2.9 years, the infants (median of 34.8 months of age) had surpassed the expected age of symptom onset for SMA types 1 or 2, and all of them were alive without the need for tracheostomy or permanent ventilation. Almost all the participants achieved the ability to sit without support (92%), and the majority achieved walking with assistance or independently (88%).3939 De Vivo DC, Bertini E, Swoboda KJ, et al; NURTURE Study Group. Nusinersen initiated in infants during the presymptomatic stage of spinal muscular atrophy: Interim efficacy and safety results from the Phase 2 NURTURE study. Neuromuscul Disord 2019;29 (11):842–856. Doi: 10.1016/j.nmd.2019.09.007
https://doi.org/10.1016/j.nmd.2019.09.00...
After five years of follow-up, all patients were alive, and none discontinued the treatment or utilized respiratory intervention. Children with three SMN2 copies achieved all WHO motor milestones, and all children with two SMN2 copies achieved sitting without support, 4/15 walking with assistance, and 13/15 walking alone.4040 Crawford TO, Swoboda KJ, De Vivo DC, et al; NURTURE Study Group. Continued benefit of nusinersen initiated in the presymptomatic stage of spinal muscular atrophy: 5-year update of the NURTURE study. Muscle Nerve 2023;68(02):157–170. Doi: 10.1002/mus.27853
https://doi.org/10.1002/mus.27853...

Risdiplam (Evrysdi®) is a small oral molecule designed to selectively modify the splicing of SMN2 pre-mRNA and promote the inclusion of exon 7 to increase levels of functional SMN protein from a complete mRNA transcript.4141 Ratni H, Ebeling M, Baird J, et al. Discovery of Risdiplam, a Selective Survival of Motor Neuron-2 ( SMN2) Gene Splicing Modifier for the Treatment of Spinal Muscular Atrophy (SMA). J Med Chem 2018;61(15):6501–6517 Registration approval in Brazil occurred in October 2020.

The FIREFISH study is investigating the safety and efficacy of risdiplam in type 1 SMA versus historical controls.44 Baranello G, Darras BT, Day JW, et al; FIREFISH Working Group. Risdiplam in Type 1 Spinal Muscular Atrophy. N Engl J Med 2021; 384(10):915–923,4242 Darras BT, Masson R, Mazurkiewicz-Bełdzińska M, et al; FIREFISH Working Group. Risdiplam-Treated Infants with Type 1 Spinal Muscular Atrophy versus Historical Controls. N Engl J Med 2021;385(05):427–435. Doi: 10.1056/nejmoa2102047
https://doi.org/10.1056/nejmoa2102047...
After 24 months of treatment, 44% of the patients were sitting without support for at least 30 seconds.4343 Finkel RS, Farrar MA, Servais L, et al. RAINBOWFISH: Primary efficacy and safety data in risdiplam-treated infants with presymptomatic spinal muscular atrophy (SMA). In: 28th International Annual Congress of the World Muscle Society (WMS), Charleston, USA; October 3–7, 2023 The event-free survival at month 12 was 85% and at month 24 was 83%.4242 Darras BT, Masson R, Mazurkiewicz-Bełdzińska M, et al; FIREFISH Working Group. Risdiplam-Treated Infants with Type 1 Spinal Muscular Atrophy versus Historical Controls. N Engl J Med 2021;385(05):427–435. Doi: 10.1056/nejmoa2102047
https://doi.org/10.1056/nejmoa2102047...
,4444 Masson R, Mazurkiewicz-Bełdzińska M, Rose K, et al; FIREFISH Study Group. Safety and efficacy of risdiplam in patients with type 1 spinal muscular atrophy (FIREFISH part 2): secondary analyses from an open-label trial. Lancet Neurol 2022;21(12): 1110–1119. Doi: 10.1016/S1474-4422(22)00339-8
https://doi.org/10.1016/S1474-4422(22)00...
The most frequently reported adverse event was upper respiratory tract infection in 54%.4444 Masson R, Mazurkiewicz-Bełdzińska M, Rose K, et al; FIREFISH Study Group. Safety and efficacy of risdiplam in patients with type 1 spinal muscular atrophy (FIREFISH part 2): secondary analyses from an open-label trial. Lancet Neurol 2022;21(12): 1110–1119. Doi: 10.1016/S1474-4422(22)00339-8
https://doi.org/10.1016/S1474-4422(22)00...
SUNFISH (NCT02908685), a phase 3, randomized, placebo-controlled study, investigates the effects of risdiplam in type 2 and non-ambulant type 3 SMA.4545 Mercuri E, Baranello G, Boespflug-Tanguy O, et al. Risdiplam in types 2 and 3 spinal muscular atrophy: A randomized, placebo-controlled, dose-finding trial followed by 24 months of treatment. Eur J Neurol 2023;30(07):1945–1956. Doi: 10.1111/ene.15499
https://doi.org/10.1111/ene.15499...
Part 1 of the study showed that a median two-fold increase of serum SMN protein was obtained within four weeks of treatment, and it was sustained over 24 months of treatment.4545 Mercuri E, Baranello G, Boespflug-Tanguy O, et al. Risdiplam in types 2 and 3 spinal muscular atrophy: A randomized, placebo-controlled, dose-finding trial followed by 24 months of treatment. Eur J Neurol 2023;30(07):1945–1956. Doi: 10.1111/ene.15499
https://doi.org/10.1111/ene.15499...
In part 2 of the study, an exploratory efficacy showed improved or stabilized motor function. A significantly greater change from baseline in the 32-item Motor Function Measure (MFM32) total score was observed with risdiplam compared with placebo at month 12.4646 Mercuri E, Deconinck N, Mazzone ES, et al; SUNFISH Study Group. Safety and efficacy of once-daily risdiplam in type 2 and non-ambulant type 3 spinal muscular atrophy (SUNFISH part 2): a phase 3, double-blind, randomised, placebo-controlled trial. Lancet Neurol 2022;21(01):42–52. Doi: 10.1016/S1474-4422(21)00367-7
https://doi.org/10.1016/S1474-4422(21)00...
At month 24, 32% of patients demonstrated improvement from baseline in MFM32 total score, and 58% showed stabilization.4747 Oskoui M, Day JW, Deconinck N, et al; SUNFISH Working Group. Two-year efficacy and safety of risdiplam in patients with type 2 or non-ambulant type 3 spinal muscular atrophy (SMA). J Neurol 2023;270(05):2531–2546. Doi: 10.1007/s00415-023-11560-1
https://doi.org/10.1007/s00415-023-11560...

The JEWELFISH is an ongoing, open-label study designed to assess the effects of risdiplam in the broadest population, including patients with SMA types 1–3 (n = 174) with a wide range of ages (1–60 years), disease severities, and who have previously received other therapies (RG7800, 7 nusinersen, olesoxime or onasemnogene abeparvovec).4848 Chiriboga CA, Bruno C, Duong T, et al; JEWELFISH Study Group. Risdiplam in Patients Previously Treated with Other Therapies for Spinal Muscular Atrophy: An Interim Analysis from the JEWELFISH Study. Neurol Ther 2023;12(02):543–557. Doi: 10.1007/s40120-023-00444-1
https://doi.org/10.1007/s40120-023-00444...
The study showed a favorable safety profile and an increase in SMN protein levels after 12 months of treatment.4848 Chiriboga CA, Bruno C, Duong T, et al; JEWELFISH Study Group. Risdiplam in Patients Previously Treated with Other Therapies for Spinal Muscular Atrophy: An Interim Analysis from the JEWELFISH Study. Neurol Ther 2023;12(02):543–557. Doi: 10.1007/s40120-023-00444-1
https://doi.org/10.1007/s40120-023-00444...
An increase in the total distance walked in the 6MWT was observed in ambulant patients over 24 months of treatment with risdiplam.4848 Chiriboga CA, Bruno C, Duong T, et al; JEWELFISH Study Group. Risdiplam in Patients Previously Treated with Other Therapies for Spinal Muscular Atrophy: An Interim Analysis from the JEWELFISH Study. Neurol Ther 2023;12(02):543–557. Doi: 10.1007/s40120-023-00444-1
https://doi.org/10.1007/s40120-023-00444...
Real-world experience with risdiplam has also been published and further supported its beneficial effects on motor function in patients with SMA.4949 Pascual-Morena C, Martínez-Vizcaíno V, Cavero-Redondo I, et al. Efficacy of risdiplam in spinal muscular atrophy: A systematic review and meta-analysis. Pharmacotherapy 2023;•••;. Doi: 10.1002/phar.2866
https://doi.org/10.1002/phar.2866...

RAINBOWFISH is an ongoing, multicenter, open-label, single-arm study to assess the efficacy and safety of risdiplam in pre-symptomatic SMA. Preliminary data showed that most infants treated with risdiplam could sit independently, and many were standing and walking as assessed by the HINE-2 at month 12.4343 Finkel RS, Farrar MA, Servais L, et al. RAINBOWFISH: Primary efficacy and safety data in risdiplam-treated infants with presymptomatic spinal muscular atrophy (SMA). In: 28th International Annual Congress of the World Muscle Society (WMS), Charleston, USA; October 3–7, 2023 After 12 months of treatment, most infants achieved near-maximum CHOP-INTEND total scores. All infants maintained bulbar function, and none required permanent ventilation after 12 months of treatment.4343 Finkel RS, Farrar MA, Servais L, et al. RAINBOWFISH: Primary efficacy and safety data in risdiplam-treated infants with presymptomatic spinal muscular atrophy (SMA). In: 28th International Annual Congress of the World Muscle Society (WMS), Charleston, USA; October 3–7, 2023

Onasemnogene abeparvovec (Zolgensma®) is a gene replacement therapy based on a self-complementary adeno-associated virus serotype 9 (AAV9) vector that carries a functional copy of the human SMN1.22 Mendell JR, Al-Zaidy S, Shell R, et al. Single-Dose Gene-Replacement Therapy for Spinal Muscular Atrophy. N Engl J Med 2017; 377(18):1713–1722 The administration of onasemnogene abeparvovec is performed intravenously, allowing the AAV9 vector to cross the blood-brain barrier. In the US, the FDA has approved it for the treatment of children with SMA who are under two years of age. In Brazil, the ANVISA has approved onasemnogene abeparvovec for babies younger than two years old.

The START study was a pivotal clinical trial that evaluated the safety and efficacy of onasemnogene abeparvovec in patients with type 1 SMA who had two copies of the SMN2. At 20 months following the treatment, 11 of the 12 children receiving the high dose of gene therapy could sit unassisted and feed unassisted.22 Mendell JR, Al-Zaidy S, Shell R, et al. Single-Dose Gene-Replacement Therapy for Spinal Muscular Atrophy. N Engl J Med 2017; 377(18):1713–1722 Data from the extension study showed maintenance of the effectiveness for at least five years.5050 Mendell JR, Al-Zaidy SA, Lehman KJ, et al. Five-Year Extension Results of the Phase 1 START Trial of Onasemnogene Abeparvovec in Spinal Muscular Atrophy. JAMA Neurol 2021;78(07): 834–841. Doi: 10.1001/jamaneurol.2021.1272
https://doi.org/10.1001/jamaneurol.2021....

The phase 3 studies STR1VE-EU conducted in Europe and STR1VE-US conducted in the US further confirmed the effectiveness of onasemnogene abeparvovec in treating patients with type 1 SMA when administered before six months of age.5151 Mercuri E, Muntoni F, Baranello G, et al; STR1VE-EU study group. Onasemnogene abeparvovec gene therapy for symptomatic infantile-onset spinal muscular atrophy type 1 (STR1VE-EU): an openlabel, single-arm, multicentre, phase 3 trial. Lancet Neurol 2021;20 (10):832–841. Doi: 10.1016/S1474-4422(21)00251-9
https://doi.org/10.1016/S1474-4422(21)00...
,5252 Day JW, Finkel RS, Chiriboga CA, et al. Onasemnogene abeparvovec gene therapy for symptomatic infantile-onset spinal muscular atrophy in patients with two copies of SMN2 (STR1VE): an open-label, single-arm, multicentre, phase 3 trial. Lancet Neurol 2021;20(04):284–293. Doi: 10.1016/S1474-4422(21)00001-6
https://doi.org/10.1016/S1474-4422(21)00...
Between 44% to 59% of treated patients could sit unsupported at 18 months. Furthermore, between 91% to 97% of the infants were alive and free from mechanical ventilation at 14 months of age.5151 Mercuri E, Muntoni F, Baranello G, et al; STR1VE-EU study group. Onasemnogene abeparvovec gene therapy for symptomatic infantile-onset spinal muscular atrophy type 1 (STR1VE-EU): an openlabel, single-arm, multicentre, phase 3 trial. Lancet Neurol 2021;20 (10):832–841. Doi: 10.1016/S1474-4422(21)00251-9
https://doi.org/10.1016/S1474-4422(21)00...
,5252 Day JW, Finkel RS, Chiriboga CA, et al. Onasemnogene abeparvovec gene therapy for symptomatic infantile-onset spinal muscular atrophy in patients with two copies of SMN2 (STR1VE): an open-label, single-arm, multicentre, phase 3 trial. Lancet Neurol 2021;20(04):284–293. Doi: 10.1016/S1474-4422(21)00001-6
https://doi.org/10.1016/S1474-4422(21)00...

Real-world studies have confirmed the efficacy of the gene replacement therapy in an expanded age range of patients eligible for treatment, including those up to two years old, and also patients with three copies of SMN2, regardless of the type of SMA.5353 Weiss C, Ziegler A, Becker -L-L, et al. Gene replacement therapy with onasemnogene abeparvovec in childrenwith spinal muscular atrophy aged 24 months or younger and bodyweight up to 15 kg: an observational cohort study. Lancet Child Adolesc Health 2022;6 (01):17–27. Doi: 10.1016/S2352-4642(21)00287-X
https://doi.org/10.1016/S2352-4642(21)00...

54 Pane M, Berti B, Capasso A, et al; ITASMAc group. Onasemnogene abeparvovec in spinal muscular atrophy: predictors of efficacy and safety in naïve patients with spinal muscular atrophy and following switch from other therapies. EClinicalMedicine 2023; 59:101997. Doi: 10.1016/j.eclinm.2023.101997
https://doi.org/10.1016/j.eclinm.2023.10...
-5555 Yang D, Ruan Y, Chen Y. Safety and efficacy of gene therapy with onasemnogene abeparvovec in the treatment of spinal muscular atrophy: A systematic review and meta-analysis. J Paediatr Child Health 2023;59(03):431–438. Doi: 10.1111/jpc.16340
https://doi.org/10.1111/jpc.16340...
Additionally, some studies have evaluated the use of onasemnogene abeparvovec in patients who had previously been treated with other specific therapies, such as nusinersen or Risdiplam.5353 Weiss C, Ziegler A, Becker -L-L, et al. Gene replacement therapy with onasemnogene abeparvovec in childrenwith spinal muscular atrophy aged 24 months or younger and bodyweight up to 15 kg: an observational cohort study. Lancet Child Adolesc Health 2022;6 (01):17–27. Doi: 10.1016/S2352-4642(21)00287-X
https://doi.org/10.1016/S2352-4642(21)00...
,5454 Pane M, Berti B, Capasso A, et al; ITASMAc group. Onasemnogene abeparvovec in spinal muscular atrophy: predictors of efficacy and safety in naïve patients with spinal muscular atrophy and following switch from other therapies. EClinicalMedicine 2023; 59:101997. Doi: 10.1016/j.eclinm.2023.101997
https://doi.org/10.1016/j.eclinm.2023.10...

The SPR1NT phase 3 study has provided crucial evidence on the efficacy of gene replacement therapy in pre-symptomatic children with SMA.5656 Strauss KA, Farrar MA, Muntoni F, et al. Onasemnogene abeparvovec for presymptomatic infants with two copies of SMN2 at risk for spinal muscular atrophy type 1: the Phase III SPR1NT trial. Nat Med 2022;28(07):1381–1389. Doi: 10.1038/s41591-022-01866-4
https://doi.org/10.1038/s41591-022-01866...
5757 Strauss KA, Farrar MA, Muntoni F, et al. Onasemnogene abeparvovec for presymptomatic infants with three copies of SMN2 at risk for spinal muscular atrophy: the Phase III SPR1NT trial. Nat Med 2022b;28(07):1390–1397. Doi: 10.1038/s41591-022-01867-3
https://doi.org/10.1038/s41591-022-01867...
In children with three SMN2 copies, all 15 participants stood independently before 24 months, within the expected developmental window.5757 Strauss KA, Farrar MA, Muntoni F, et al. Onasemnogene abeparvovec for presymptomatic infants with three copies of SMN2 at risk for spinal muscular atrophy: the Phase III SPR1NT trial. Nat Med 2022b;28(07):1390–1397. Doi: 10.1038/s41591-022-01867-3
https://doi.org/10.1038/s41591-022-01867...
Additionally, 14 of them walked independently within the expected developmental window, and 67% maintained body weight without requiring feeding support through 24 months. For the 14 enrolled infants with two SMN2 copies, all of them achieved the ability to sit independently for ≥30 seconds at any visit before 18 months of age.5656 Strauss KA, Farrar MA, Muntoni F, et al. Onasemnogene abeparvovec for presymptomatic infants with two copies of SMN2 at risk for spinal muscular atrophy type 1: the Phase III SPR1NT trial. Nat Med 2022;28(07):1381–1389. Doi: 10.1038/s41591-022-01866-4
https://doi.org/10.1038/s41591-022-01866...
Importantly, all patients with two or three SMN2 copies in the study survived without permanent ventilation at 14 months, and none of the children required nutritional or respiratory support. At 18 months (children with two copies of SMN2) and 24 months (children with three copies of SMN2), all children swallowed normally and achieved full oral nutrition.5858 Shell RD, McGrattan KE, Hurst-Davis R, et al. Onasemnogene abeparvovec preserves bulbar function in infants with presymptomatic spinal muscular atrophy: a post-hoc analysis of the SPR1NT trial. Neuromuscul Disord 2023;33(08):670–676. Doi: 10.1016/j.nmd.2023.06.005
https://doi.org/10.1016/j.nmd.2023.06.00...

In human clinical trials and real-world studies, treatment-related severe adverse events of onasemnogene abeparvovec have been reported in just over 10% of cases, with the most common being liver function abnormalities and fever.5959 Ogbonmide T, Rathore R, Rangrej SB, et al. Gene Therapy for Spinal Muscular Atrophy (SMA): A Review of Current Challenges and Safety Considerations for Onasemnogene Abeparvovec (Zolgensma). Cureus 2023;15(03):e36197. Doi: 10.7759/cureus.36197
https://doi.org/10.7759/cureus.36197...
Other important adverse events include decreased platelet count and thrombocytopenia. Fatal cases of thrombotic microangiopathy and acute liver failure have been reported,6060 Guillou J, de Pellegars A, Porcheret F, et al. Fatal thrombotic microangiopathy case following adeno-associated viral SMN gene therapy. Blood Adv 2022;6(14):4266–4270. Doi: 10.1182/ bloodadvances.2021006419
https://doi.org/10.1182/bloodadvances.20...
,6161 Philippidis A. Novartis Confirms Deaths of Two Patients Treated with Gene Therapy Zolgensma. Hum Gene Ther 2022;33(17-18):842–844. Doi: 10.1089/hum.2022.29216.bfs
https://doi.org/10.1089/hum.2022.29216.b...
as well as potentially fatal conditions such as hemophagocytic syndrome,6262 Galletta F, Cucinotta U, Marseglia L, et al. Hemophagocytic lymphohistiocytosis following gene replacement therapy in a child with type 1 spinal muscular atrophy. J Clin Pharm Ther 2022;47(09):1478–1481. Doi: 10.1111/jcpt.13733
https://doi.org/10.1111/jcpt.13733...
and necrotizing enterocolitis.6363 Gaillard J, Gu AR, Neil Knierbein EE. Necrotizing Enterocolitis following Onasemnogene Abeparvovec for Spinal Muscular Atrophy: A Case Series. J Pediatr 2023;260:113493. Doi: 10.1016/j.jpeds.2023.113493
https://doi.org/10.1016/j.jpeds.2023.113...

The STRONG study (NCT03381729) is a clinical trial that evaluated the safety and efficacy of an intrathecal single dose of onasemnogene abeparvovec in non-ambulatory patients with SMA who have three copies of the SMN2 and are aged 6 to under 60 months.6464 Finkel RS, Darras BT, Mendell JR, et al. Intrathecal Onasemnogene Abeparvovec for Sitting, Nonambulatory Patients with Spinal Muscular Atrophy: Phase I Ascending-Dose Study (STRONG). J Neuromuscul Dis 2023;10(03):389–404. Doi: 10.3233/JND-221560
https://doi.org/10.3233/JND-221560...
In the younger group (6 to under 24 months) treated with the medium dose, one out of thirteen patients (7.7%) achieved independent standing. For the older group (24 to under 60 months) receiving the medium dose, there was a significant improvement in the change from baseline in the HFMSE compared with the SMA historic control population at month 12.6464 Finkel RS, Darras BT, Mendell JR, et al. Intrathecal Onasemnogene Abeparvovec for Sitting, Nonambulatory Patients with Spinal Muscular Atrophy: Phase I Ascending-Dose Study (STRONG). J Neuromuscul Dis 2023;10(03):389–404. Doi: 10.3233/JND-221560
https://doi.org/10.3233/JND-221560...
Further research is ongoing to explore the use of lower intrathecal doses of onasemnogene abeparvovec in patients aged 2 to under 18 years in a randomized multicenter controlled clinical trial (NCT05089656).

GENE-BASED THERAPIES FOR NEUROPATHIES

Among the inherited peripheral neuropathies, outstanding treatment developments occurred in the last decade. The natural history of patients with transthyretin-associated amyloidosis and acute hepatic porphyria with repetitive crisis has definitely changed due to genetic therapies.

Hereditary transthyretin amyloidosis

Mutations in the transthyretin gene (TTR) result in an amyloidogenic multisystemic disease (ATTRv) that affects mainly the peripheral nervous system (ATTRv-pn) and the heart (ATTRv-h).6565 Poli L, Labella B, Cotti Piccinelli S, et al. Hereditary transthyretin amyloidosis: a comprehensive review with a focus on peripheral neuropathy. Front Neurol 2023;14:1242815. Doi: 10.3389/ fneur.2023.1242815
https://doi.org/10.3389/fneur.2023.12428...
,6666 Adams D, Sekijima Y, Conceição I, et al. Hereditary transthyretin amyloid neuropathies: advances in pathophysiology, biomarkers, and treatment. Lancet Neurol 2023;22(11):1061–1074. Doi: 10.1016/S1474-4422(23)00334-4
https://doi.org/10.1016/S1474-4422(23)00...
Most of the TTR protein has liver production and circulates as a monotetramer, that transports thyroxin and vitamin A.6565 Poli L, Labella B, Cotti Piccinelli S, et al. Hereditary transthyretin amyloidosis: a comprehensive review with a focus on peripheral neuropathy. Front Neurol 2023;14:1242815. Doi: 10.3389/ fneur.2023.1242815
https://doi.org/10.3389/fneur.2023.12428...
,6666 Adams D, Sekijima Y, Conceição I, et al. Hereditary transthyretin amyloid neuropathies: advances in pathophysiology, biomarkers, and treatment. Lancet Neurol 2023;22(11):1061–1074. Doi: 10.1016/S1474-4422(23)00334-4
https://doi.org/10.1016/S1474-4422(23)00...

ATTRv is a fatal autosomal dominant disease of variable penetrance caused by the deposition of misfolded TTR fibrils whose prognosis changed enormously after liver transplantation. The natural history of the disease was also modified with the introduction of tafamidis, a small molecule that stabilizes the TTR tetramer, avoiding fibril formation.6565 Poli L, Labella B, Cotti Piccinelli S, et al. Hereditary transthyretin amyloidosis: a comprehensive review with a focus on peripheral neuropathy. Front Neurol 2023;14:1242815. Doi: 10.3389/ fneur.2023.1242815
https://doi.org/10.3389/fneur.2023.12428...
,6666 Adams D, Sekijima Y, Conceição I, et al. Hereditary transthyretin amyloid neuropathies: advances in pathophysiology, biomarkers, and treatment. Lancet Neurol 2023;22(11):1061–1074. Doi: 10.1016/S1474-4422(23)00334-4
https://doi.org/10.1016/S1474-4422(23)00...

The possibility of interrupting TTR production through gene silencing became a clinical reality in the second half of the last decade with the use of two different technologies: antisense oligonucleotides (ASO) and small interfering RNAs (siRNA), both effectively suppressing TTR production acting at RNA level.6666 Adams D, Sekijima Y, Conceição I, et al. Hereditary transthyretin amyloid neuropathies: advances in pathophysiology, biomarkers, and treatment. Lancet Neurol 2023;22(11):1061–1074. Doi: 10.1016/S1474-4422(23)00334-4
https://doi.org/10.1016/S1474-4422(23)00...
ASO selectively binds to the complementary RNA, preventing RNA translation and gene expression, which can also be modulated by siRNA which targets and cleaves complementary mRNAs.6767 Carroll A, Dyck PJ, de Carvalho M, et al. Novel approaches to diagnosis and management of hereditary transthyretin amyloidosis. J Neurol Neurosurg Psychiatry 2022;93(06):668–678. Doi: 10.1136/jnnp-2021-327909
https://doi.org/10.1136/jnnp-2021-327909...

The NEURO-TTR trial showed that the antisense oligonucleotide inotersen was effective in stages 1 and 2 of the ATTRv-pn.55 Benson MD, Waddington-Cruz M, Berk JL, et al. Inotersen Treatment for Patients with Hereditary Transthyretin Amyloidosis. N Engl J Med 2018;379(01):22–31 Administered subcutaneously weekly it was well tolerated and effective in stabilizing the disease, but three patients developed glomerulonephritis, and the other three developed thrombocytopenia, with one death. After the introduction of regular monitoring, no more serious complications were reported. The open-label extension study confirmed the efficacy of inotersen in slowing the course of neuropathy and improving quality of life.6868 Brannagan TH, Wang AK, Coelho T, et al; NEURO-TTR open-label extension investigators. Early data on long-term efficacy and safety of inotersen in patients with hereditary transthyretin amyloidosis: a 2-year update from the open-label extension of the NEURO-TTR trial. Eur J Neurol 2020;27(08):1374–1381. Doi: 10.1111/ene.14285
https://doi.org/10.1111/ene.14285...

More recently, in trying to improve the safety and dosing profile of inotersen, the same sequence has been ligand-conjugated to produce a new and potent ASO (eplontersen), which is administered subcutaneously every 4 weeks.6969 Coelho T, Waddington Cruz M, Chao CC, et al. Characteristics of Patients with Hereditary Transthyretin Amyloidosis-Polyneuropathy (ATTRv-PN) in NEURO-TTRansform, an Open-label Phase 3 Study of Eplontersen. Neurol Ther 2023;12(01):267–287. Doi: 10.1007/s40120-022-00414-z
https://doi.org/10.1007/s40120-022-00414...
,7070 Coelho T, Marques W Jr, Dasgupta NR, et al; NEURO-TTRansform Investigators. Eplontersen for Hereditary Transthyretin Amyloidosis With Polyneuropathy. JAMA 2023;330(15):1448–1458 The phase 3 NEURO-TTRansform study showed that the eplontersen treatment group had significantly lowered serum transthyretin concentration, less neuropathy impairment, and better quality of life compared with a historical placebo.6969 Coelho T, Waddington Cruz M, Chao CC, et al. Characteristics of Patients with Hereditary Transthyretin Amyloidosis-Polyneuropathy (ATTRv-PN) in NEURO-TTRansform, an Open-label Phase 3 Study of Eplontersen. Neurol Ther 2023;12(01):267–287. Doi: 10.1007/s40120-022-00414-z
https://doi.org/10.1007/s40120-022-00414...

The phase 3 APOLLO study showed that the siRNA patisiran was effective and safe at the dose of 0.3 mg/kg given intravenously every 3 weeks.66 Adams D, Gonzalez-Duarte A, O’Riordan WD, et al. Patisiran, an RNAi Therapeutic, for Hereditary Transthyretin Amyloidosis. N Engl J Med 2018;379(01):11–21 The open-label extension study confirmed the efficacy and safety of the drug,7171 Adams D, Polydefkis M, González-Duarte A, et al; patisiran Global OLE study group. Long-term safety and efficacy of patisiran for hereditary transthyretin-mediated amyloidosis with polyneuropathy: 12-month results of an open-label extension study. Lancet Neurol 2021;20(01):49–59. Doi: 10.1016/S1474-4422(20)30368-9
https://doi.org/10.1016/S1474-4422(20)30...
and it was shown that patisiran was also efficient in transplanted patients.7272 Schmidt HH, Wixner J, Planté-Bordeneuve V, et al; Patisiran Post- LT Study Group. Patisiran treatment in patients with hereditary transthyretin-mediated amyloidosis with polyneuropathy after liver transplantation. Am J Transplant 2022;22(06):1646–1657 In phase 3 (APOLLO-B, NCT03997383), a double-blind, placebo-controlled, randomized trial, the administration of patisiran over a period of 12 months resulted in preserved functional capacity in patients with ATTR cardiac amyloidosis.7373 Maurer MS, Kale P, Fontana M, et al; APOLLO-B Trial Investigators. Patisiran Treatment in Patients with Transthyretin Cardiac Amyloidosis. N Engl J Med 2023;389(17):1553–1565. Doi: 10.1056/NEJMoa2300757
https://doi.org/10.1056/NEJMoa2300757...
Infusion-related reactions, arthralgia, and muscle spasms occurred more often among patients in the patisiran group than among those in the placebo group.7373 Maurer MS, Kale P, Fontana M, et al; APOLLO-B Trial Investigators. Patisiran Treatment in Patients with Transthyretin Cardiac Amyloidosis. N Engl J Med 2023;389(17):1553–1565. Doi: 10.1056/NEJMoa2300757
https://doi.org/10.1056/NEJMoa2300757...

The HELIOS-A study showed that vutrisiran, a siRNA-GalNAc conjugate, administered subcutaneously (25 mg every 3 months), resulted in significant improvement of the neuropathy impairment.7474 Adams D, Tournev IL, Taylor MS, et al; HELIOS-A Collaborators. Efficacy and safety of vutrisiran for patients with hereditary transthyretin-mediated amyloidosis with polyneuropathy: a randomized clinical trial. Amyloid 2023;30(01):1–9. Doi: 10.1080/13506129.2022.2091985
https://doi.org/10.1080/13506129.2022.20...

Gene editing therapy with the use of clustered, regularly interspaced short palindromic repeats and associated Cas9 endonuclease is also under evaluation to potentially solve the problem permanently. In the initial proof-of-concept study, single doses (0.3 mg/kg) of the NTLA-2001 CRISPR-Cas9 system resulted in a significant reduction of TTR blood levels without significant adverse events.7575 Gillmore JD, Gane E, Taubel J, et al. Crispr-Cas9 in vivo gene editing for transthyretin amyloidosis. N Engl J Med 2021;385 (06):493–502. Doi: 10.1056/NEJMoa2107454
https://doi.org/10.1056/NEJMoa2107454...
Ongoing and larger studies will test the clinical efficacy and safety of this system.7575 Gillmore JD, Gane E, Taubel J, et al. Crispr-Cas9 in vivo gene editing for transthyretin amyloidosis. N Engl J Med 2021;385 (06):493–502. Doi: 10.1056/NEJMoa2107454
https://doi.org/10.1056/NEJMoa2107454...

Acute hepatic porphyria

Acute hepatic porphyria is a group of four diseases, three of autosomal dominant inheritance (acute intermittent porphyria, hereditary coproporphyria, variegate porphyria) and one autosomal recessive (ALA dehydratase) that result from abnormalities in the heme biosynthesis pathway. The penetrance is reduced and most carriers will never manifest the disease, but in those that manifest, the consequences may be serious and even fatal.7676 Gerischer LM, Scheibe F, Nümann A, Köhnlein M, Stölzel U, Meisel A. Acute porphyrias - A neurological perspective. Brain Behav 2021;11(11):e2389. Doi: 10.1002/brb3.2389
https://doi.org/10.1002/brb3.2389...
They usually occur in crises that cause intense abdominal pain, nausea, vomiting, dysautonomia (tachycardia and blood pressure instability), psychiatric problems, seizures, and acute axonal neuropathy.7777 Zhang MM, Bahal R, Rasmussen TP, Manautou JE, Zhong XB. The growth of siRNA-based therapeutics: Updated clinical studies. Biochem Pharmacol 2021;189:114432 Most patients have favorable prognoses with measures to avoid the precipitating factors and use of glucose and hematin at the onset of the attacks.

For those developing repetitive crises (> 4 a year) it has recently been introduced givosiran, a siRNA that binds to a target sequence on ALAS1 mRNA, decreasing the production of ALA and PBG, the molecules thought to be responsible for the porphyria attacks. The phase 3 ENVISION trial showed a 74% decrease in the attack rate, and the secondary endpoints were also met.7878 Balwani M, Sardh E, Ventura P, et al; ENVISION Investigators. Phase 3 Trial of RNAi Therapeutic Givosiran for Acute Intermittent Porphyria. N Engl J Med 2020;382(24):2289–2301 Givosiran was well tolerated, but some patients developed renal and hepatic complications, demanding close surveillance.7777 Zhang MM, Bahal R, Rasmussen TP, Manautou JE, Zhong XB. The growth of siRNA-based therapeutics: Updated clinical studies. Biochem Pharmacol 2021;189:114432 The recommended dose is 2.5 mg/kg once a month. Extension studies are ongoing.

GENE-BASED THERAPIES FOR DUCHENNE MUSCULAR DYSTROPHY

Duchenne muscular dystrophy (DMD) is an X-linked recessive muscular dystrophy caused by mutations in the DMD (dystrophin) gene.7979 Hoffman EP, Brown RH Jr, Kunkel LM. Dystrophin: the protein product of the Duchenne muscular dystrophy locus. Cell 1987;51 (06):919–928. Doi: 10.1016/0092-8674(87)90579-4
https://doi.org/10.1016/0092-8674(87)905...
The disease affects around 1 in 3500 - 5000 boys and causes progressive weakness starting between 3 and 4 years old and loss of ambulation at the age of around 10–12 years.8080 Crisafulli S, Sultana J, Fontana A, Salvo F, Messina S, Trifirò G Global epidemiology of Duchenne muscular dystrophy: an updated systematic review and meta-analysis. Orphanet J Rare Dis 2020;15(01):141. Doi: 10.1186/s13023-020-01430-8
https://doi.org/10.1186/s13023-020-01430...
In addition, DMD patients present respiratory difficulties and progressive dilated cardiomyopathy leading to heart failure and early death.

The DMD is the largest gene identified with 79 exons.8181 Roberts RG, Coffey AJ, Bobrow M, Bentley DR. Exon structure of the human dystrophin gene. Genomics 1993;16(02):536–538. Doi: 10.1006/geno.1993.1225
https://doi.org/10.1006/geno.1993.1225...
DMD mutations are intragenic deletions affecting one or more exons (60-65%), duplications of one or more exons (10%), and nucleotide variants (around 20-25%), including missense and nonsense variants, small insertions/deletions, or splicing alterations.8282 Saad FA, Siciliano G, Angelini C. Advances in Dystrophinopathy Diagnosis and Therapy. Biomolecules 2023;13(09):1319. Doi: 10.3390/biom13091319
https://doi.org/10.3390/biom13091319...
Mutations in DMD that do not disrupt the reading frame, leading to the expression of a truncated yet functional dystrophin cause the milder phenotype of Becker muscular dystrophy.8383 Monaco AP, Bertelson CJ, Liechti-Gallati S, Moser H, Kunkel LM. An explanation for the phenotypic differences between patients bearing partial deletions of the DMD locus. Genomics 1988;2(01):90–95

There is no curative treatment for DMD. Nonetheless, a multidisciplinary approach targeting the symptoms of DMD can change the natural course of the disease. Glucocorticosteroids, such as deflazacort or prednisone, are the current standard of care treatment for DMD patients (Araujo 2023).8484 Araujo APQC, Saute JAM, Fortes CPDD, et al. Update of the Brazilian consensus recommendations on Duchenne muscular dystrophy. Arq Neuropsiquiatr 2023;81(01):81–94. Doi: 10.1055/s-0043-1761466
https://doi.org/10.1055/s-0043-1761466...
Genetic therapies that act by regulating the expression of DMD or administering a transcript capable of encoding a smaller but still functional dystrophin have been the most promising therapies.

Gene replacement therapy with microdystrophin

The large size of the DMD gene does not allow its transfer via an AAV vector. On the other hand, microdystrophin transcripts can be easily transferred to DMD patients via viral vectors.8585 Duan D. Systemic AAV Micro-dystrophin Gene Therapy for Duchenne Muscular Dystrophy. Mol Ther 2018;26(10):2337–2356. Doi: 10.1016/j.ymthe.2018.07.011
https://doi.org/10.1016/j.ymthe.2018.07....
Delandistrogene moxeparvovec is approved in the US for the treatment of ambulatory patients (4-5 years) with DMD. Delandistrogene moxeparvovec (SRP-9001), from Sarepta Therapeutics, is an investigational gene therapy designed for targeted expression of SRP-9001 dystrophin protein, a shortened dystrophin retaining key functional domains of the wild-type protein. An open-label, phase 1/2a, nonrandomized controlled clinical trial (NCT03375164) enrolled four DMD ambulatory males with a mean age of 5.1 years.8686 Mendell JR, Sahenk Z, Lehman K, et al. Assessment of Systemic Delivery of rAAVrh74.MHCK7.micro-dystrophin in Children With Duchenne Muscular Dystrophy: A Nonrandomized Controlled Trial. JAMA Neurol 2020;77(09):1122–1131. Doi: 10.1001/jamaneurol.2020.1484
https://doi.org/10.1001/jamaneurol.2020....
Patients received a single IV infusion (2.0 × 1014 vg/kg) of delandistrogene moxeparvovec and steroids.8686 Mendell JR, Sahenk Z, Lehman K, et al. Assessment of Systemic Delivery of rAAVrh74.MHCK7.micro-dystrophin in Children With Duchenne Muscular Dystrophy: A Nonrandomized Controlled Trial. JAMA Neurol 2020;77(09):1122–1131. Doi: 10.1001/jamaneurol.2020.1484
https://doi.org/10.1001/jamaneurol.2020....
At 12 weeks, immunohistochemistry of gastrocnemius muscle specimens revealed robust transgene expression in all patients, with a mean of 81.2% of muscle fibers expressing micro-dystrophin with a mean intensity of 96% at the sarcolemma. Western blot showed a mean expression of 74.3% of the protein without fat or fibrosis adjustment.8686 Mendell JR, Sahenk Z, Lehman K, et al. Assessment of Systemic Delivery of rAAVrh74.MHCK7.micro-dystrophin in Children With Duchenne Muscular Dystrophy: A Nonrandomized Controlled Trial. JAMA Neurol 2020;77(09):1122–1131. Doi: 10.1001/jamaneurol.2020.1484
https://doi.org/10.1001/jamaneurol.2020....
After 4 years of treatment, there were 18 treatment-related adverse events; all occurred within 70 days posttreatment and were resolved.8787 Mendell JR, Sahenk Z, Lehman KJ, et al. Long-term safety and functional outcomes of delandistrogene moxeparvovec gene therapy in patients with Duchenne muscular dystrophy: A phase 1/2a nonrandomized trial. Muscle Nerve 2023;•••;. Doi: 10.1002/ mus.27955
https://doi.org/10.1002/mus.27955...
The mean North Star Ambulatory Assessment (NSAA) total score increased from 20.5 to 27.5 (+7.0, 2.9), from baseline to year 4.

SRP-9001-102 (NCT03769116) is a phase 2, double-blind, two-part crossover study to evaluate delandistrogene moxeparvovec in DMD patients aged ≥4 to <8 years.8888 Mendell JR, Shieh PB, McDonald CM, et al. Expression of SRP- 9001 dystrophin and stabilization of motor function up to 2 years post-treatment with delandistrogene moxeparvovec gene therapy in individuals with Duchenne muscular dystrophy. Front Cell Dev Biol 2023b;11:1167762. Doi: 10.3389/fcell.2023.1167762
https://doi.org/10.3389/fcell.2023.11677...
Patients were randomized and stratified by age to placebo (n = 21) or delandistrogene moxeparvovec (n = 20) and crossed over for part 2. SRP-9001 dystrophin expression was achieved in all patients: mean change from the baseline to week 12 was 23.82% and 39.64% in parts 1 and 2, respectively.8888 Mendell JR, Shieh PB, McDonald CM, et al. Expression of SRP- 9001 dystrophin and stabilization of motor function up to 2 years post-treatment with delandistrogene moxeparvovec gene therapy in individuals with Duchenne muscular dystrophy. Front Cell Dev Biol 2023b;11:1167762. Doi: 10.3389/fcell.2023.1167762
https://doi.org/10.3389/fcell.2023.11677...
In part 1, the change from the baseline to week 48 in NSAA score was +1.7 for treatment versus +0.9 for placebo.8888 Mendell JR, Shieh PB, McDonald CM, et al. Expression of SRP- 9001 dystrophin and stabilization of motor function up to 2 years post-treatment with delandistrogene moxeparvovec gene therapy in individuals with Duchenne muscular dystrophy. Front Cell Dev Biol 2023b;11:1167762. Doi: 10.3389/fcell.2023.1167762
https://doi.org/10.3389/fcell.2023.11677...
In 4- to 5-year-olds with matched baseline motor function, the change from the baseline to week 48 in NSAA scores was significantly different (+2.5 points), but not significantly different in 6- to 7-year-olds with imbalanced baseline motor function (-0.7 points). For patients treated with delandistrogene moxeparvovec in part 2, the change from the baseline to week 48 in NSAA score was +1.3, whereas, for those treated in part 1, NSAA scores were maintained.8888 Mendell JR, Shieh PB, McDonald CM, et al. Expression of SRP- 9001 dystrophin and stabilization of motor function up to 2 years post-treatment with delandistrogene moxeparvovec gene therapy in individuals with Duchenne muscular dystrophy. Front Cell Dev Biol 2023b;11:1167762. Doi: 10.3389/fcell.2023.1167762
https://doi.org/10.3389/fcell.2023.11677...
The most common adverse events were vomiting, decreased appetite, and nausea.8888 Mendell JR, Shieh PB, McDonald CM, et al. Expression of SRP- 9001 dystrophin and stabilization of motor function up to 2 years post-treatment with delandistrogene moxeparvovec gene therapy in individuals with Duchenne muscular dystrophy. Front Cell Dev Biol 2023b;11:1167762. Doi: 10.3389/fcell.2023.1167762
https://doi.org/10.3389/fcell.2023.11677...

ENDEAVOR (NCT04626674) is a single-arm, open-label study to evaluate delandistrogene moxeparvovec in DMD boys.8989 Zaidman CM, Proud CM, McDonald CM, et al. Delandistrogene Moxeparvovec Gene Therapy in Ambulatory Patients (Aged ≥4 to >8 Years) with Duchenne Muscular Dystrophy: 1-Year Interim Results from Study SRP-9001-103 (ENDEAVOR). Ann Neurol 2023;94(05):955–968. Doi: 10.1002/ana.26755
https://doi.org/10.1002/ana.26755...
In cohort 1 (n = 20), eligible ambulatory males, aged ≥4 to <8 years, received a single IV infusion of delandistrogene moxeparvovec (1.33 × 1014 vg/kg). At week 12, microdystrophin expression had a mean change from the baseline of 54.2% with sarcolemmal localization.8989 Zaidman CM, Proud CM, McDonald CM, et al. Delandistrogene Moxeparvovec Gene Therapy in Ambulatory Patients (Aged ≥4 to >8 Years) with Duchenne Muscular Dystrophy: 1-Year Interim Results from Study SRP-9001-103 (ENDEAVOR). Ann Neurol 2023;94(05):955–968. Doi: 10.1002/ana.26755
https://doi.org/10.1002/ana.26755...
At one year, patients stabilized or improved in NSAA total scores.8989 Zaidman CM, Proud CM, McDonald CM, et al. Delandistrogene Moxeparvovec Gene Therapy in Ambulatory Patients (Aged ≥4 to >8 Years) with Duchenne Muscular Dystrophy: 1-Year Interim Results from Study SRP-9001-103 (ENDEAVOR). Ann Neurol 2023;94(05):955–968. Doi: 10.1002/ana.26755
https://doi.org/10.1002/ana.26755...
Treatment versus a propensity score-weighted external natural history control demonstrated a statistically significant difference in NSAA (+3.2 points).8989 Zaidman CM, Proud CM, McDonald CM, et al. Delandistrogene Moxeparvovec Gene Therapy in Ambulatory Patients (Aged ≥4 to >8 Years) with Duchenne Muscular Dystrophy: 1-Year Interim Results from Study SRP-9001-103 (ENDEAVOR). Ann Neurol 2023;94(05):955–968. Doi: 10.1002/ana.26755
https://doi.org/10.1002/ana.26755...
These positive results advanced SRP-9001 into a double-blind, placebo-controlled, phase 3 clinical trial called EMBARK (NCT05096221).

Fordadistrogene movaparvovec (PF-06939926), from Pfizer, is an AAV9 gene-replacement construct containing a truncated dystrophin transgene. A phase 1b open-label clinical trial (NCT03362502) has been testing the PF-06939926 in males with DMD aged 4 and older.9090 Butterfield R, Shieh P, Yong F, et al. One year data from ambulatory boys in a phase 1b, open-label study of fordadistrogene movaparvovec (PF-06939926) for Duchenne muscular dystrophy (DMD). 2023 MDA Clinical & Scientific Conference. Nineteen ambulatory boys received fordadistrogene movaparvovec (n = 3 low-dose; n = 16 high-dose). Median age at gene therapy was 8.8 years (6.2–13.0 years).9090 Butterfield R, Shieh P, Yong F, et al. One year data from ambulatory boys in a phase 1b, open-label study of fordadistrogene movaparvovec (PF-06939926) for Duchenne muscular dystrophy (DMD). 2023 MDA Clinical & Scientific Conference. Three treatment-related serious adverse events occurred (dehydration, acute kidney injury, thrombocytopenia); all resolved within 15 days. The median change from baseline to 1-year in NSAA total score was +1 point against -4 points for an external control cohort.9090 Butterfield R, Shieh P, Yong F, et al. One year data from ambulatory boys in a phase 1b, open-label study of fordadistrogene movaparvovec (PF-06939926) for Duchenne muscular dystrophy (DMD). 2023 MDA Clinical & Scientific Conference. A randomized, placebo-controlled phase 3 trial, called CIFFREO (NCT04281485), is planned to enroll 99 DMD boys aged 4 to 7.

SGT-001 is a microdystrophin developed by Solid Biosciences that contains a neuronal nitric oxide synthase binding (nNOS) domain responsible for protection against ischemia-induced muscle injury by maintaining nitric oxide (NO) signaling at the muscle sarcolemma. SGT-001 is currently being evaluated in a phase 1/2 study called IGNITE DMD (NCT03368742).9191 Dreghici RD. IGNITE DMD Phase I/II Study of SGT-001 Microdystrophin Gene Therapy for DMD: 2-Year Outcomes Update. 2022 MDA Clinical & Scientific Conference. Interim analysis of 2 years (n = 9 boys) demonstrated common drug-related laboratory abnormalities (thrombocytopenia, anemia), and serious adverse events: systemic inflammatory response syndrome (n = 2), thrombocytopenia (n = 1), and immune hepatitis (n = 1), which were resolved.9191 Dreghici RD. IGNITE DMD Phase I/II Study of SGT-001 Microdystrophin Gene Therapy for DMD: 2-Year Outcomes Update. 2022 MDA Clinical & Scientific Conference. Results showed durable microdystrophin expression and localization on nNOS to the membrane in biopsies collected at time points ranging from 12-24 months post-dosing. In addition, patients had stable Six Minutes Walking Test (6MWT) distances and NSAA scores compared to natural history, and improvements in Forced Vital Capacity (FVC) and Peak Expiratory Flow (PEF) compared to baseline and natural history.9191 Dreghici RD. IGNITE DMD Phase I/II Study of SGT-001 Microdystrophin Gene Therapy for DMD: 2-Year Outcomes Update. 2022 MDA Clinical & Scientific Conference. Up to three years post-dosing, there was durability of treatment effect. Subjects receiving 2E14 vg/kg maintained motor and pulmonary functions compared to expected natural history declines.9292 Dreghici RD, Gonzalez P, Brown K, Morris C, Shieh P, Byrne B. 3- Year Outcomes Update in the IGNITE DMD Phase 1/2 Study of SGT-001 Microdystrophin Gene Therapy. 2023 MDA Clinical & Scientific Conference.

Gene replacement therapy using microdystrophin is a major advance in the treatment of DMD. However, there are important safety concerns. Among the adverse events of therapy, an immune response against protein epitopes encoded by the microdystrophin construct can generate a cytotoxic response mediated by T cells in patients missing these epitopes, which may cause severe myositis and myocarditis.9393 Bönnemann CG, Belluscio BA, Braun S, Morris C, Singh T, Muntoni F. Dystrophin Immunity after Gene Therapy for Duchenne's Muscular Dystrophy. N Engl J Med 2023;388(24):2294–2296. Doi: 10.1056/NEJMc2212912
https://doi.org/10.1056/NEJMc2212912...
This has been particularly observed in patients with mutations encompassing exons 8 to 21. Another important limitation of gene therapy for DMD is that as AAV vectors do not integrate into the muscle fiber genome and do not tend to infect satellite cells, with the process of muscle renewal and regeneration, the effectiveness of the therapy tends to be lost with time.9494 Arnett ALH, Konieczny P, Ramos JN, et al. Adeno-associated viral (AAV) vectors do not efficiently target muscle satellite cells. Mol Ther Methods Clin Dev 2014;1:14038. Doi: 10.1038/mtm.2014.38
https://doi.org/10.1038/mtm.2014.38...

Antisense oligonucleotides (ASOs) therapy for DMD

Antisense oligonucleotides (ASOs), or phosphorodiamidate morpholino oligomers, can induce therapeutic exon skipping during pre-mRNA processing to restore the reading frame of the primary transcript of DMD.9595 Aartsma-Rus A, Fokkema I, Verschuuren J, et al. Theoretic applicability of antisense-mediated exon skipping for Duchenne muscular dystrophy mutations. Hum Mutat 2009;30(03): 293–299. Doi: 10.1002/humu.20918
https://doi.org/10.1002/humu.20918...
As a result, truncated but partially functional dystrophin is produced, potentially slowing down the disease progression and causing a milder Becker muscular dystrophy phenotype.

Eteplirsen is an ASO from Sarepta Therapeutics that binds to a complementary region in the DMD exon 51 pre-mRNA and causes its skipping during the mRNA splicing process.9696 Mendell JR, Rodino-Klapac LR, Sahenk Z, et al; Eteplirsen Study Group. Eteplirsen for the treatment of Duchenne muscular dystrophy. Ann Neurol 2013;74(05):637–647. Doi: 10.1002/ana.23982
https://doi.org/10.1002/ana.23982...
Around 13–14% of DMD cases have mutations that can be potentially treated by skipping exon 51.9797 Charleston JS, Schnell FJ, Dworzak J, et al. Eteplirsen treatment for Duchenne muscular dystrophy: Exon skipping and dystrophin production. Neurology 2018;90(24):e2146–e2154. Doi: 10.1212/WNL.0000000000005680
https://doi.org/10.1212/WNL.000000000000...
Eteplirsen received accelerated approval from the FDA in September 2016. In clinical trials, eteplirsen was shown to be safe, and capable of increasing dystrophin levels in muscle tissue and maintaining the motor function of DMD patients.9696 Mendell JR, Rodino-Klapac LR, Sahenk Z, et al; Eteplirsen Study Group. Eteplirsen for the treatment of Duchenne muscular dystrophy. Ann Neurol 2013;74(05):637–647. Doi: 10.1002/ana.23982
https://doi.org/10.1002/ana.23982...

97 Charleston JS, Schnell FJ, Dworzak J, et al. Eteplirsen treatment for Duchenne muscular dystrophy: Exon skipping and dystrophin production. Neurology 2018;90(24):e2146–e2154. Doi: 10.1212/WNL.0000000000005680
https://doi.org/10.1212/WNL.000000000000...

98 Mendell JR, Goemans N, Lowes LP, et al; Eteplirsen Study Group and Telethon Foundation DMD Italian Network. Longitudinal effect of eteplirsen versus historical control on ambulation in Duchenne muscular dystrophy. Ann Neurol 2016;79(02): 257–271. Doi: 10.1002/ana.24555
https://doi.org/10.1002/ana.24555...

99 Mendell JR, Khan N, Sha N, et al; Eteplirsen Study Group. Comparison of Long-term Ambulatory Function in Patients with Duchenne Muscular Dystrophy Treated with Eteplirsen and Matched Natural History Controls. J Neuromuscul Dis 2021;8(04):469–479. Doi: 10.3233/JND-200548
https://doi.org/10.3233/JND-200548...
-100100 McDonald CM, Shieh PB, Abdel-Hamid HZ, et al; the Italian DMD Telethon Registry Study Group, Leuven NMRC Registry Investigators, CINRG Duchenne Natural History Investigators, and PROMOVI Trial Clinical Investigators. Open-Label Evaluation of Eteplirsen in Patients with Duchenne Muscular Dystrophy Amenable to Exon 51 Skipping: PROMOVI Trial. J Neuromuscul Dis 2021;8(06):989–1001. Doi: 10.3233/JND-210643
https://doi.org/10.3233/JND-210643...

Golodirsen is an ASO from Sarepta Therapeutics approved by the FDA under accelerated review for the treatment of DMD in patients with exon 53 amendable skipping mutations (approximately 8% of all DMD mutations).101101 Anwar S, Yokota T. Golodirsen for Duchenne muscular dystrophy. Drugs Today (Barc) 2020;56(08):491–504. Doi: 10.1358/ dot.2020.56.8.3159186
https://doi.org/10.1358/dot.2020.56.8.31...
In clinical trials A significant increase in exon 53 skipping and dystrophin protein expression was demonstrated.102102 Frank DE, Schnell FJ, Akana C, et al; SKIP-NMD Study Group. Increased dystrophin production with golodirsen in patients with Duchenne muscular dystrophy. Neurology 2020;94(21): e2270–e2282. Doi: 10.1212/WNL.0000000000009233
https://doi.org/10.1212/WNL.000000000000...

103 Servais L, Mercuri E, Straub V, et al; SKIP-NMD Study Group. Long-Term Safety and Efficacy Data of Golodirsen in Ambulatory Patients with Duchenne Muscular Dystrophy Amenable to Exon 53 Skipping: A First-in-human, Multicenter, Two-Part, Open- Label, Phase 1/2 Trial. Nucleic Acid Ther 2022;32(01):29–39. Doi: 10.1089/nat.2021.0043
https://doi.org/10.1089/nat.2021.0043...
-104104 Scaglioni D, Catapano F, Ellis M, et al. The administration ofantisense oligonucleotide golodirsen reduces pathological regeneration in patients with Duchenne muscular dystrophy. Acta Neuropathol Commun 2021;9(01):7. Doi: 10.1186/s40478-020-01106-1
https://doi.org/10.1186/s40478-020-01106...
In addition, mild improvement in 6MWT, and a slowing decline of FVC occurred.103103 Servais L, Mercuri E, Straub V, et al; SKIP-NMD Study Group. Long-Term Safety and Efficacy Data of Golodirsen in Ambulatory Patients with Duchenne Muscular Dystrophy Amenable to Exon 53 Skipping: A First-in-human, Multicenter, Two-Part, Open- Label, Phase 1/2 Trial. Nucleic Acid Ther 2022;32(01):29–39. Doi: 10.1089/nat.2021.0043
https://doi.org/10.1089/nat.2021.0043...

Viltolarsen is an ASO designed to treat DMD in patients with a confirmed mutation of the DMD gene amenable to exon 53 skipping.105105 Clemens PR, Rao VK, Connolly AM, et al; CINRG DNHS Investigators. Safety, tolerability, and efficacy of viltolarsen in boys with Duchenne muscular dystrophy amenable to exon 53 skipping: A phase 2 randomized clinical trial. JAMA Neurol 2020;77(08):982–991,106106 Clemens PR, Rao VK, Connolly AM, et al; CINRG DNHS Investigators. Efficacy and Safety of Viltolarsen in Boys With Duchenne Muscular Dystrophy: Results From the Phase 2, Open- Label, 4-Year Extension Study. J Neuromuscul Dis 2023;10(03): 439–447. Doi: 10.3233/JND-221656
https://doi.org/10.3233/JND-221656...
Viltolarsen is approved in the US by the FDA and in Japan. An increase of dystrophin value of 5.9% was observed,105105 Clemens PR, Rao VK, Connolly AM, et al; CINRG DNHS Investigators. Safety, tolerability, and efficacy of viltolarsen in boys with Duchenne muscular dystrophy amenable to exon 53 skipping: A phase 2 randomized clinical trial. JAMA Neurol 2020;77(08):982–991 and viltolarsen-treated patients presented stabilization of motor function over 4 years of extension study.106106 Clemens PR, Rao VK, Connolly AM, et al; CINRG DNHS Investigators. Efficacy and Safety of Viltolarsen in Boys With Duchenne Muscular Dystrophy: Results From the Phase 2, Open- Label, 4-Year Extension Study. J Neuromuscul Dis 2023;10(03): 439–447. Doi: 10.3233/JND-221656
https://doi.org/10.3233/JND-221656...

Readthrough therapies for DMD

DMD is caused by a nonsense mutation in the DMD in 10–15% of cases. Nonsense mutations result in the production of truncated, non-functional dystrophin. Ataluren (Translarna®) is an oral treatment designed to promote the synthesis of full-length dystrophin through ribosomal readthrough of an in-frame premature stop codon caused by a nonsense mutation in the dystrophin mRNA.107107 Roy B, Friesen WJ, Tomizawa Y, et al. Ataluren stimulates ribosomal selection of near-cognate tRNAs to promote nonsense suppression. Proc Natl Acad Sci U S A 2016;113(44):12508–12513. Doi: 10.1073/pnas.1605336113
https://doi.org/10.1073/pnas.1605336113...

A randomized, double-blind, placebo-controlled study, including males ≥ 5 years with DMD, assessed the safety and efficacy of ataluren (n = 57) or placebo (n = 57). The primary endpoint favored ataluren 10, 10, 20 mg/kg versus placebo.108108 Bushby K, Finkel R, Wong B, et al; PTC124-GD-007-DMD STUDY GROUP. Ataluren treatment of patients with nonsense mutation dystrophinopathy. Muscle Nerve 2014;50(04):477–487. Doi: 10.1002/mus.24332
https://doi.org/10.1002/mus.24332...
A phase 3, randomized, double-blind, placebo-controlled trial, included 228 outpatient boys with DMD aged 7 to 16 years.109109 McDonald CM, Campbell C, Torricelli RE, et al; Clinical Evaluator Training Group. ; ACT DMD Study Group. Ataluren in patients with nonsense mutation Duchenne muscular dystrophy (ACT DMD): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017;390(10101):1489–1498. Doi: 10.1016/S0140-6736(17)31611-2
https://doi.org/10.1016/S0140-6736(17)31...
The study showed that change in 6MWD did not differ significantly between patients in the ataluren group and those in the placebo group.109109 McDonald CM, Campbell C, Torricelli RE, et al; Clinical Evaluator Training Group. ; ACT DMD Study Group. Ataluren in patients with nonsense mutation Duchenne muscular dystrophy (ACT DMD): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017;390(10101):1489–1498. Doi: 10.1016/S0140-6736(17)31611-2
https://doi.org/10.1016/S0140-6736(17)31...
However, a significant effect of ataluren in the prespecified subgroup of patients with a baseline 6MWD of 300 meters or more to less than 400 meters was noted.109109 McDonald CM, Campbell C, Torricelli RE, et al; Clinical Evaluator Training Group. ; ACT DMD Study Group. Ataluren in patients with nonsense mutation Duchenne muscular dystrophy (ACT DMD): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017;390(10101):1489–1498. Doi: 10.1016/S0140-6736(17)31611-2
https://doi.org/10.1016/S0140-6736(17)31...
However, to date, long-term follow-up on study participants (treated and untreated with atalurem) has not been reported.

The safety and effectiveness of ataluren in patients with nonsense mutation DMD in the STRIDE registry (real-world treatment with ataluren) were compared with the CINRG Duchenne Natural History Study.110110 Mercuri E, Osorio AN, Muntoni F, et al; STRIDE and CINRG DNHS investigators. Safety and effectiveness of ataluren in patients with nonsense mutation DMD in the STRIDE Registry compared with the CINRG Duchenne Natural History Study (2015-2022): 2022 interim analysis. J Neurol 2023;270(08):3896–3913. Doi: 10.1007/s00415-023-11687-1
https://doi.org/10.1007/s00415-023-11687...
Ataluren treatment significantly delayed age at loss of ambulation by 4 years and age at decline to predicted FVCof < 60% and < 50% by 1.8 years and 2.3 years, respectively.110110 Mercuri E, Osorio AN, Muntoni F, et al; STRIDE and CINRG DNHS investigators. Safety and effectiveness of ataluren in patients with nonsense mutation DMD in the STRIDE Registry compared with the CINRG Duchenne Natural History Study (2015-2022): 2022 interim analysis. J Neurol 2023;270(08):3896–3913. Doi: 10.1007/s00415-023-11687-1
https://doi.org/10.1007/s00415-023-11687...
However, the major limitation of this study is the fact that the comparator control group included DMD patients with different types of mutations, not just nonsense mutations, which can certainly affect the interpretation of the results.110110 Mercuri E, Osorio AN, Muntoni F, et al; STRIDE and CINRG DNHS investigators. Safety and effectiveness of ataluren in patients with nonsense mutation DMD in the STRIDE Registry compared with the CINRG Duchenne Natural History Study (2015-2022): 2022 interim analysis. J Neurol 2023;270(08):3896–3913. Doi: 10.1007/s00415-023-11687-1
https://doi.org/10.1007/s00415-023-11687...

Ataluren is not approved by the FDA. The Human Medicines Committee (CHMP) of the European Medicines Agency (EMA) gave conditional marketing authorization in 2014. However, in October 2023, in a preliminary analysis, the committee concluded that Ataluren's benefit-risk balance is negative and therefore recommended not renewing the marketing authorization in Europe.111111 https://www.ema.europa.eu/en/news/ema-recommends-non-renewal-authorisation-duchenne-muscular-dystrophy-medicine-translarna
https://www.ema.europa.eu/en/news/ema-re...
In Brazil, ANVISA approved ataluren in 2019 and recently expanded the age range for its use from 2 years onwards.

  • Support

    This study was supported by ABN (Academia Brasileira de Neurologia).

References

  • 1
    Landfeldt E. Gene Therapy for Neuromuscular Diseases: Health Economic Challenges and Future Perspectives. J Neuromuscul Dis 2022;9(06):675–688
  • 2
    Mendell JR, Al-Zaidy S, Shell R, et al. Single-Dose Gene-Replacement Therapy for Spinal Muscular Atrophy. N Engl J Med 2017; 377(18):1713–1722
  • 3
    Finkel RS, Mercuri E, Darras BT, et al; ENDEAR Study Group. Nusinersen versus Sham Control in Infantile-Onset Spinal Muscular Atrophy. N Engl J Med 2017;377(18):1723–1732
  • 4
    Baranello G, Darras BT, Day JW, et al; FIREFISH Working Group. Risdiplam in Type 1 Spinal Muscular Atrophy. N Engl J Med 2021; 384(10):915–923
  • 5
    Benson MD, Waddington-Cruz M, Berk JL, et al. Inotersen Treatment for Patients with Hereditary Transthyretin Amyloidosis. N Engl J Med 2018;379(01):22–31
  • 6
    Adams D, Gonzalez-Duarte A, O’Riordan WD, et al. Patisiran, an RNAi Therapeutic, for Hereditary Transthyretin Amyloidosis. N Engl J Med 2018;379(01):11–21
  • 7
    Rossor AM, Reilly MM, Sleigh JN. Antisense oligonucleotides and other genetic therapies made simple. Pract Neurol 2018;18(02): 126–131
  • 8
    Goyal N, Narayanaswami P. Making sense of antisense oligonucleotides: A narrative review. Muscle Nerve 2018;57(03):356–370
  • 9
    Lam JK, Chow MY, Zhang Y, Leung SW. siRNA Versus miRNA as Therapeutics for Gene Silencing. Mol Ther Nucleic Acids 2015;4 (09):e252
  • 10
    Kay MA. State-of-the-art gene-based therapies: the road ahead. Nat Rev Genet 2011;12(05):316–328
  • 11
    Brown RH Jr, Al-Chalabi A. Amyotrophic Lateral Sclerosis. N Engl J Med 2017;377(16):1602
  • 12
    Akçimen F, Lopez ER, Landers JE, et al. Amyotrophic lateral sclerosis: translating genetic discoveries into therapies. Nat Rev Genet 2023;24(09):642–658
  • 13
    Chia R, Chiò A, Traynor BJ. Novel genes associated with amyotrophic lateral sclerosis: diagnostic and clinical implications. Lancet Neurol 2018;17(01):94–102
  • 14
    Miller TM, Cudkowicz ME, Genge A, et al; VALOR and OLE Working Group. Trial of Antisense Oligonucleotide Tofersen for SOD1 ALS. N Engl J Med 2022;387(12):1099–1110
  • 15
    Mueller C, Berry JD, McKenna-Yasek DM, et al. SOD1 Suppression with Adeno-Associated Virus and MicroRNA in Familial ALS. N Engl J Med 2020;383(02):151–158
  • 16
    Rosen DR, Siddique T, Patterson D, et al. Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature 1993;362(6415):59–62
  • 17
    https://www.fda.gov/drugs/news-events-human-drugs/fda-approves-treatment-amyotrophic-lateral-sclerosis-associated-mutation-sod1-gene
    » https://www.fda.gov/drugs/news-events-human-drugs/fda-approves-treatment-amyotrophic-lateral-sclerosis-associated-mutation-sod1-gene
  • 18
    Benatar M, Wuu J, Andersen PM, et al. Design of a Randomized, Placebo-Controlled, Phase 3 Trial of Tofersen Initiated in Clinically Presymptomatic SOD1 Variant Carriers: the ATLAS Study. Neurotherapeutics 2022;19(04):1248–1258
  • 19
    https://uniqure.gcs-web.com/news-releases/news-release-details/uniqure-and-apic-bio-enter-global-licensing-agreement-apb-102
    » https://uniqure.gcs-web.com/news-releases/news-release-details/uniqure-and-apic-bio-enter-global-licensing-agreement-apb-102
  • 20
    https://investors.biogen.com/news-releases/news-release-details/biogen-and-ionis-announce-topline-phase-1-study-results
    » https://investors.biogen.com/news-releases/news-release-details/biogen-and-ionis-announce-topline-phase-1-study-results
  • 21
    Boros BD, Schoch KM, Kreple CJ, Miller TM. Antisense Oligonucleotides for the Study and Treatment of ALS. Neurotherapeutics 2022;19(04):1145–1158
  • 22
    Tavares de Andrade HM, Cintra VP, de Albuquerque M, et al. Intermediate-length CAG repeat in ATXN2 is associated with increased risk for amyotrophic lateral sclerosis in Brazilian patients. Neurobiol Aging 2018;69:292.e15–292.e18
  • 23
    Becker LA, Huang B, Bieri G, et al. Therapeutic reduction of ataxin-2 extends lifespan and reduces pathology in TDP-43 mice. Nature 2017;544(7650):367–371
  • 24
    Lefebvre S, Bürglen L, Reboullet S, et al. Identification and characterization of a spinal muscular atrophy-determining gene. Cell 1995;80(01):155–165
  • 25
    Kolb SJ, Kissel JT. Spinal muscular atrophy: a timely review. Arch Neurol 2011;68(08):979–984
  • 26
    Finkel R, Bertini E, Muntoni F, Mercuri EENMC SMA Workshop Study Group. 209th ENMC International Workshop: Outcome Measures and Clinical Trial Readiness in Spinal Muscular Atrophy 7-9 November 2014, Heemskerk, The Netherlands. Neuromuscul Disord 2015;25(07):593–602
  • 27
    Mendonça RH, Matsui C Jr, Polido GJ, et al. Intragenic variants in the SMN1 gene determine the clinical phenotype in 5q spinal muscular atrophy. Neurol Genet 2020;6(05):e505
  • 28
    Calucho M, Bernal S, Alías L, et al. Correlation between SMA type and SMN2 copy number revisited: An analysis of 625 unrelated Spanish patients and a compilation of 2834 reported cases. Neuromuscul Disord 2018;28(03):208–215
  • 29
    Chen TH. New and Developing Therapies in Spinal Muscular Atrophy: From Genotype to Phenotype to Treatment and Where Do We Stand? Int J Mol Sci 2020;21(09):3297
  • 30
    Chiriboga CA, Swoboda KJ, Darras BT, et al. Results from a phase 1 study of nusinersen (ISIS-SMN(Rx)) in children with spinal muscular atrophy. Neurology 2016;86(10):890–897
  • 31
    Finkel RS, Chiriboga CA, Vajsar J, et al. Treatment of infantile-onset spinal muscular atrophy with nusinersen: a phase 2, open-label, dose-escalation study. Lancet 2016;388(10063):3017–3026
  • 32
    Mercuri E, Darras BT, Chiriboga CA, et al; CHERISH Study Group. Nusinersen versus sham control in later-onset spinal muscular atrophy. N Engl J Med 2018;378(07):625–635. Doi: 10.1056/ NEJMoa1710504
    » https://doi.org/10.1056/NEJMoa1710504
  • 33
    Darras BT, Chiriboga CA, Iannaccone ST, et al; ISIS-396443-CS2/ ISIS-396443-CS12 Study Groups. Nusinersen in later-onset spinal muscular atrophy: Long-term results from the phase 1/2 studies. Neurology 2019;92(21):e2492–e2506. Doi: 10.1212/ WNL.0000000000007527
    » https://doi.org/10.1212/WNL.0000000000007527
  • 34
    Finkel RS, Chiriboga CA, Vajsar J, et al. Treatment of infantileonset spinal muscular atrophy with nusinersen: final report of a phase 2, open-label, multicentre, dose-escalation study. Lancet Child Adolesc Health 2021;5(07):491–500. Doi: 10.1016/S2352-4642(21)00100-0
    » https://doi.org/10.1016/S2352-4642(21)00100-0
  • 35
    Hagenacker T, Wurster CD, Günther R, et al. Nusinersen in adults with 5q spinal muscular atrophy: a non-interventional, multicentre, observational cohort study. Lancet Neurol 2020;19(04): 317–325. Doi: 10.1016/S1474-4422(20)30037-5
    » https://doi.org/10.1016/S1474-4422(20)30037-5
  • 36
    de Holanda Mendonça R, Jorge Polido G, Ciro M, Jorge Fontoura Solla D, Conti Reed U, Zanoteli E. Clinical Outcomes in Patients with Spinal Muscular Atrophy Type 1 Treated with Nusinersen. J Neuromuscul Dis 2021;8(02):217–224. Doi: 10.3233/JND-200533
    » https://doi.org/10.3233/JND-200533
  • 37
    Mendonça RH, Polido GJ, Matsui C, et al. Real-World Data from Nusinersen Treatment for Patients with Later-Onset Spinal Muscular Atrophy: A Single Center Experience. J Neuromuscul Dis 2021;8(01):101–108. Doi: 10.3233/JND-200551
    » https://doi.org/10.3233/JND-200551
  • 38
    Gavriilaki M, Moschou M, Papaliagkas V, et al. Nusinersen in Adults with 5q Spinal Muscular Atrophy: a Systematic Review and Meta-analysis. Neurotherapeutics 2022;19(02):464–475. Doi: 10.1007/s13311-022-01200-3
    » https://doi.org/10.1007/s13311-022-01200-3
  • 39
    De Vivo DC, Bertini E, Swoboda KJ, et al; NURTURE Study Group. Nusinersen initiated in infants during the presymptomatic stage of spinal muscular atrophy: Interim efficacy and safety results from the Phase 2 NURTURE study. Neuromuscul Disord 2019;29 (11):842–856. Doi: 10.1016/j.nmd.2019.09.007
    » https://doi.org/10.1016/j.nmd.2019.09.007
  • 40
    Crawford TO, Swoboda KJ, De Vivo DC, et al; NURTURE Study Group. Continued benefit of nusinersen initiated in the presymptomatic stage of spinal muscular atrophy: 5-year update of the NURTURE study. Muscle Nerve 2023;68(02):157–170. Doi: 10.1002/mus.27853
    » https://doi.org/10.1002/mus.27853
  • 41
    Ratni H, Ebeling M, Baird J, et al. Discovery of Risdiplam, a Selective Survival of Motor Neuron-2 ( SMN2) Gene Splicing Modifier for the Treatment of Spinal Muscular Atrophy (SMA). J Med Chem 2018;61(15):6501–6517
  • 42
    Darras BT, Masson R, Mazurkiewicz-Bełdzińska M, et al; FIREFISH Working Group. Risdiplam-Treated Infants with Type 1 Spinal Muscular Atrophy versus Historical Controls. N Engl J Med 2021;385(05):427–435. Doi: 10.1056/nejmoa2102047
    » https://doi.org/10.1056/nejmoa2102047
  • 43
    Finkel RS, Farrar MA, Servais L, et al. RAINBOWFISH: Primary efficacy and safety data in risdiplam-treated infants with presymptomatic spinal muscular atrophy (SMA). In: 28th International Annual Congress of the World Muscle Society (WMS), Charleston, USA; October 3–7, 2023
  • 44
    Masson R, Mazurkiewicz-Bełdzińska M, Rose K, et al; FIREFISH Study Group. Safety and efficacy of risdiplam in patients with type 1 spinal muscular atrophy (FIREFISH part 2): secondary analyses from an open-label trial. Lancet Neurol 2022;21(12): 1110–1119. Doi: 10.1016/S1474-4422(22)00339-8
    » https://doi.org/10.1016/S1474-4422(22)00339-8
  • 45
    Mercuri E, Baranello G, Boespflug-Tanguy O, et al. Risdiplam in types 2 and 3 spinal muscular atrophy: A randomized, placebo-controlled, dose-finding trial followed by 24 months of treatment. Eur J Neurol 2023;30(07):1945–1956. Doi: 10.1111/ene.15499
    » https://doi.org/10.1111/ene.15499
  • 46
    Mercuri E, Deconinck N, Mazzone ES, et al; SUNFISH Study Group. Safety and efficacy of once-daily risdiplam in type 2 and non-ambulant type 3 spinal muscular atrophy (SUNFISH part 2): a phase 3, double-blind, randomised, placebo-controlled trial. Lancet Neurol 2022;21(01):42–52. Doi: 10.1016/S1474-4422(21)00367-7
    » https://doi.org/10.1016/S1474-4422(21)00367-7
  • 47
    Oskoui M, Day JW, Deconinck N, et al; SUNFISH Working Group. Two-year efficacy and safety of risdiplam in patients with type 2 or non-ambulant type 3 spinal muscular atrophy (SMA). J Neurol 2023;270(05):2531–2546. Doi: 10.1007/s00415-023-11560-1
    » https://doi.org/10.1007/s00415-023-11560-1
  • 48
    Chiriboga CA, Bruno C, Duong T, et al; JEWELFISH Study Group. Risdiplam in Patients Previously Treated with Other Therapies for Spinal Muscular Atrophy: An Interim Analysis from the JEWELFISH Study. Neurol Ther 2023;12(02):543–557. Doi: 10.1007/s40120-023-00444-1
    » https://doi.org/10.1007/s40120-023-00444-1
  • 49
    Pascual-Morena C, Martínez-Vizcaíno V, Cavero-Redondo I, et al. Efficacy of risdiplam in spinal muscular atrophy: A systematic review and meta-analysis. Pharmacotherapy 2023;•••;. Doi: 10.1002/phar.2866
    » https://doi.org/10.1002/phar.2866
  • 50
    Mendell JR, Al-Zaidy SA, Lehman KJ, et al. Five-Year Extension Results of the Phase 1 START Trial of Onasemnogene Abeparvovec in Spinal Muscular Atrophy. JAMA Neurol 2021;78(07): 834–841. Doi: 10.1001/jamaneurol.2021.1272
    » https://doi.org/10.1001/jamaneurol.2021.1272
  • 51
    Mercuri E, Muntoni F, Baranello G, et al; STR1VE-EU study group. Onasemnogene abeparvovec gene therapy for symptomatic infantile-onset spinal muscular atrophy type 1 (STR1VE-EU): an openlabel, single-arm, multicentre, phase 3 trial. Lancet Neurol 2021;20 (10):832–841. Doi: 10.1016/S1474-4422(21)00251-9
    » https://doi.org/10.1016/S1474-4422(21)00251-9
  • 52
    Day JW, Finkel RS, Chiriboga CA, et al. Onasemnogene abeparvovec gene therapy for symptomatic infantile-onset spinal muscular atrophy in patients with two copies of SMN2 (STR1VE): an open-label, single-arm, multicentre, phase 3 trial. Lancet Neurol 2021;20(04):284–293. Doi: 10.1016/S1474-4422(21)00001-6
    » https://doi.org/10.1016/S1474-4422(21)00001-6
  • 53
    Weiss C, Ziegler A, Becker -L-L, et al. Gene replacement therapy with onasemnogene abeparvovec in childrenwith spinal muscular atrophy aged 24 months or younger and bodyweight up to 15 kg: an observational cohort study. Lancet Child Adolesc Health 2022;6 (01):17–27. Doi: 10.1016/S2352-4642(21)00287-X
    » https://doi.org/10.1016/S2352-4642(21)00287-X
  • 54
    Pane M, Berti B, Capasso A, et al; ITASMAc group. Onasemnogene abeparvovec in spinal muscular atrophy: predictors of efficacy and safety in naïve patients with spinal muscular atrophy and following switch from other therapies. EClinicalMedicine 2023; 59:101997. Doi: 10.1016/j.eclinm.2023.101997
    » https://doi.org/10.1016/j.eclinm.2023.101997
  • 55
    Yang D, Ruan Y, Chen Y. Safety and efficacy of gene therapy with onasemnogene abeparvovec in the treatment of spinal muscular atrophy: A systematic review and meta-analysis. J Paediatr Child Health 2023;59(03):431–438. Doi: 10.1111/jpc.16340
    » https://doi.org/10.1111/jpc.16340
  • 56
    Strauss KA, Farrar MA, Muntoni F, et al. Onasemnogene abeparvovec for presymptomatic infants with two copies of SMN2 at risk for spinal muscular atrophy type 1: the Phase III SPR1NT trial. Nat Med 2022;28(07):1381–1389. Doi: 10.1038/s41591-022-01866-4
    » https://doi.org/10.1038/s41591-022-01866-4
  • 57
    Strauss KA, Farrar MA, Muntoni F, et al. Onasemnogene abeparvovec for presymptomatic infants with three copies of SMN2 at risk for spinal muscular atrophy: the Phase III SPR1NT trial. Nat Med 2022b;28(07):1390–1397. Doi: 10.1038/s41591-022-01867-3
    » https://doi.org/10.1038/s41591-022-01867-3
  • 58
    Shell RD, McGrattan KE, Hurst-Davis R, et al. Onasemnogene abeparvovec preserves bulbar function in infants with presymptomatic spinal muscular atrophy: a post-hoc analysis of the SPR1NT trial. Neuromuscul Disord 2023;33(08):670–676. Doi: 10.1016/j.nmd.2023.06.005
    » https://doi.org/10.1016/j.nmd.2023.06.005
  • 59
    Ogbonmide T, Rathore R, Rangrej SB, et al. Gene Therapy for Spinal Muscular Atrophy (SMA): A Review of Current Challenges and Safety Considerations for Onasemnogene Abeparvovec (Zolgensma). Cureus 2023;15(03):e36197. Doi: 10.7759/cureus.36197
    » https://doi.org/10.7759/cureus.36197
  • 60
    Guillou J, de Pellegars A, Porcheret F, et al. Fatal thrombotic microangiopathy case following adeno-associated viral SMN gene therapy. Blood Adv 2022;6(14):4266–4270. Doi: 10.1182/ bloodadvances.2021006419
    » https://doi.org/10.1182/bloodadvances.2021006419
  • 61
    Philippidis A. Novartis Confirms Deaths of Two Patients Treated with Gene Therapy Zolgensma. Hum Gene Ther 2022;33(17-18):842–844. Doi: 10.1089/hum.2022.29216.bfs
    » https://doi.org/10.1089/hum.2022.29216.bfs
  • 62
    Galletta F, Cucinotta U, Marseglia L, et al. Hemophagocytic lymphohistiocytosis following gene replacement therapy in a child with type 1 spinal muscular atrophy. J Clin Pharm Ther 2022;47(09):1478–1481. Doi: 10.1111/jcpt.13733
    » https://doi.org/10.1111/jcpt.13733
  • 63
    Gaillard J, Gu AR, Neil Knierbein EE. Necrotizing Enterocolitis following Onasemnogene Abeparvovec for Spinal Muscular Atrophy: A Case Series. J Pediatr 2023;260:113493. Doi: 10.1016/j.jpeds.2023.113493
    » https://doi.org/10.1016/j.jpeds.2023.113493
  • 64
    Finkel RS, Darras BT, Mendell JR, et al. Intrathecal Onasemnogene Abeparvovec for Sitting, Nonambulatory Patients with Spinal Muscular Atrophy: Phase I Ascending-Dose Study (STRONG). J Neuromuscul Dis 2023;10(03):389–404. Doi: 10.3233/JND-221560
    » https://doi.org/10.3233/JND-221560
  • 65
    Poli L, Labella B, Cotti Piccinelli S, et al. Hereditary transthyretin amyloidosis: a comprehensive review with a focus on peripheral neuropathy. Front Neurol 2023;14:1242815. Doi: 10.3389/ fneur.2023.1242815
    » https://doi.org/10.3389/fneur.2023.1242815
  • 66
    Adams D, Sekijima Y, Conceição I, et al. Hereditary transthyretin amyloid neuropathies: advances in pathophysiology, biomarkers, and treatment. Lancet Neurol 2023;22(11):1061–1074. Doi: 10.1016/S1474-4422(23)00334-4
    » https://doi.org/10.1016/S1474-4422(23)00334-4
  • 67
    Carroll A, Dyck PJ, de Carvalho M, et al. Novel approaches to diagnosis and management of hereditary transthyretin amyloidosis. J Neurol Neurosurg Psychiatry 2022;93(06):668–678. Doi: 10.1136/jnnp-2021-327909
    » https://doi.org/10.1136/jnnp-2021-327909
  • 68
    Brannagan TH, Wang AK, Coelho T, et al; NEURO-TTR open-label extension investigators. Early data on long-term efficacy and safety of inotersen in patients with hereditary transthyretin amyloidosis: a 2-year update from the open-label extension of the NEURO-TTR trial. Eur J Neurol 2020;27(08):1374–1381. Doi: 10.1111/ene.14285
    » https://doi.org/10.1111/ene.14285
  • 69
    Coelho T, Waddington Cruz M, Chao CC, et al. Characteristics of Patients with Hereditary Transthyretin Amyloidosis-Polyneuropathy (ATTRv-PN) in NEURO-TTRansform, an Open-label Phase 3 Study of Eplontersen. Neurol Ther 2023;12(01):267–287. Doi: 10.1007/s40120-022-00414-z
    » https://doi.org/10.1007/s40120-022-00414-z
  • 70
    Coelho T, Marques W Jr, Dasgupta NR, et al; NEURO-TTRansform Investigators. Eplontersen for Hereditary Transthyretin Amyloidosis With Polyneuropathy. JAMA 2023;330(15):1448–1458
  • 71
    Adams D, Polydefkis M, González-Duarte A, et al; patisiran Global OLE study group. Long-term safety and efficacy of patisiran for hereditary transthyretin-mediated amyloidosis with polyneuropathy: 12-month results of an open-label extension study. Lancet Neurol 2021;20(01):49–59. Doi: 10.1016/S1474-4422(20)30368-9
    » https://doi.org/10.1016/S1474-4422(20)30368-9
  • 72
    Schmidt HH, Wixner J, Planté-Bordeneuve V, et al; Patisiran Post- LT Study Group. Patisiran treatment in patients with hereditary transthyretin-mediated amyloidosis with polyneuropathy after liver transplantation. Am J Transplant 2022;22(06):1646–1657
  • 73
    Maurer MS, Kale P, Fontana M, et al; APOLLO-B Trial Investigators. Patisiran Treatment in Patients with Transthyretin Cardiac Amyloidosis. N Engl J Med 2023;389(17):1553–1565. Doi: 10.1056/NEJMoa2300757
    » https://doi.org/10.1056/NEJMoa2300757
  • 74
    Adams D, Tournev IL, Taylor MS, et al; HELIOS-A Collaborators. Efficacy and safety of vutrisiran for patients with hereditary transthyretin-mediated amyloidosis with polyneuropathy: a randomized clinical trial. Amyloid 2023;30(01):1–9. Doi: 10.1080/13506129.2022.2091985
    » https://doi.org/10.1080/13506129.2022.2091985
  • 75
    Gillmore JD, Gane E, Taubel J, et al. Crispr-Cas9 in vivo gene editing for transthyretin amyloidosis. N Engl J Med 2021;385 (06):493–502. Doi: 10.1056/NEJMoa2107454
    » https://doi.org/10.1056/NEJMoa2107454
  • 76
    Gerischer LM, Scheibe F, Nümann A, Köhnlein M, Stölzel U, Meisel A. Acute porphyrias - A neurological perspective. Brain Behav 2021;11(11):e2389. Doi: 10.1002/brb3.2389
    » https://doi.org/10.1002/brb3.2389
  • 77
    Zhang MM, Bahal R, Rasmussen TP, Manautou JE, Zhong XB. The growth of siRNA-based therapeutics: Updated clinical studies. Biochem Pharmacol 2021;189:114432
  • 78
    Balwani M, Sardh E, Ventura P, et al; ENVISION Investigators. Phase 3 Trial of RNAi Therapeutic Givosiran for Acute Intermittent Porphyria. N Engl J Med 2020;382(24):2289–2301
  • 79
    Hoffman EP, Brown RH Jr, Kunkel LM. Dystrophin: the protein product of the Duchenne muscular dystrophy locus. Cell 1987;51 (06):919–928. Doi: 10.1016/0092-8674(87)90579-4
    » https://doi.org/10.1016/0092-8674(87)90579-4
  • 80
    Crisafulli S, Sultana J, Fontana A, Salvo F, Messina S, Trifirò G Global epidemiology of Duchenne muscular dystrophy: an updated systematic review and meta-analysis. Orphanet J Rare Dis 2020;15(01):141. Doi: 10.1186/s13023-020-01430-8
    » https://doi.org/10.1186/s13023-020-01430-8
  • 81
    Roberts RG, Coffey AJ, Bobrow M, Bentley DR. Exon structure of the human dystrophin gene. Genomics 1993;16(02):536–538. Doi: 10.1006/geno.1993.1225
    » https://doi.org/10.1006/geno.1993.1225
  • 82
    Saad FA, Siciliano G, Angelini C. Advances in Dystrophinopathy Diagnosis and Therapy. Biomolecules 2023;13(09):1319. Doi: 10.3390/biom13091319
    » https://doi.org/10.3390/biom13091319
  • 83
    Monaco AP, Bertelson CJ, Liechti-Gallati S, Moser H, Kunkel LM. An explanation for the phenotypic differences between patients bearing partial deletions of the DMD locus. Genomics 1988;2(01):90–95
  • 84
    Araujo APQC, Saute JAM, Fortes CPDD, et al. Update of the Brazilian consensus recommendations on Duchenne muscular dystrophy. Arq Neuropsiquiatr 2023;81(01):81–94. Doi: 10.1055/s-0043-1761466
    » https://doi.org/10.1055/s-0043-1761466
  • 85
    Duan D. Systemic AAV Micro-dystrophin Gene Therapy for Duchenne Muscular Dystrophy. Mol Ther 2018;26(10):2337–2356. Doi: 10.1016/j.ymthe.2018.07.011
    » https://doi.org/10.1016/j.ymthe.2018.07.011
  • 86
    Mendell JR, Sahenk Z, Lehman K, et al. Assessment of Systemic Delivery of rAAVrh74.MHCK7.micro-dystrophin in Children With Duchenne Muscular Dystrophy: A Nonrandomized Controlled Trial. JAMA Neurol 2020;77(09):1122–1131. Doi: 10.1001/jamaneurol.2020.1484
    » https://doi.org/10.1001/jamaneurol.2020.1484
  • 87
    Mendell JR, Sahenk Z, Lehman KJ, et al. Long-term safety and functional outcomes of delandistrogene moxeparvovec gene therapy in patients with Duchenne muscular dystrophy: A phase 1/2a nonrandomized trial. Muscle Nerve 2023;•••;. Doi: 10.1002/ mus.27955
    » https://doi.org/10.1002/mus.27955
  • 88
    Mendell JR, Shieh PB, McDonald CM, et al. Expression of SRP- 9001 dystrophin and stabilization of motor function up to 2 years post-treatment with delandistrogene moxeparvovec gene therapy in individuals with Duchenne muscular dystrophy. Front Cell Dev Biol 2023b;11:1167762. Doi: 10.3389/fcell.2023.1167762
    » https://doi.org/10.3389/fcell.2023.1167762
  • 89
    Zaidman CM, Proud CM, McDonald CM, et al. Delandistrogene Moxeparvovec Gene Therapy in Ambulatory Patients (Aged ≥4 to >8 Years) with Duchenne Muscular Dystrophy: 1-Year Interim Results from Study SRP-9001-103 (ENDEAVOR). Ann Neurol 2023;94(05):955–968. Doi: 10.1002/ana.26755
    » https://doi.org/10.1002/ana.26755
  • 90
    Butterfield R, Shieh P, Yong F, et al. One year data from ambulatory boys in a phase 1b, open-label study of fordadistrogene movaparvovec (PF-06939926) for Duchenne muscular dystrophy (DMD). 2023 MDA Clinical & Scientific Conference.
  • 91
    Dreghici RD. IGNITE DMD Phase I/II Study of SGT-001 Microdystrophin Gene Therapy for DMD: 2-Year Outcomes Update. 2022 MDA Clinical & Scientific Conference.
  • 92
    Dreghici RD, Gonzalez P, Brown K, Morris C, Shieh P, Byrne B. 3- Year Outcomes Update in the IGNITE DMD Phase 1/2 Study of SGT-001 Microdystrophin Gene Therapy. 2023 MDA Clinical & Scientific Conference.
  • 93
    Bönnemann CG, Belluscio BA, Braun S, Morris C, Singh T, Muntoni F. Dystrophin Immunity after Gene Therapy for Duchenne's Muscular Dystrophy. N Engl J Med 2023;388(24):2294–2296. Doi: 10.1056/NEJMc2212912
    » https://doi.org/10.1056/NEJMc2212912
  • 94
    Arnett ALH, Konieczny P, Ramos JN, et al. Adeno-associated viral (AAV) vectors do not efficiently target muscle satellite cells. Mol Ther Methods Clin Dev 2014;1:14038. Doi: 10.1038/mtm.2014.38
    » https://doi.org/10.1038/mtm.2014.38
  • 95
    Aartsma-Rus A, Fokkema I, Verschuuren J, et al. Theoretic applicability of antisense-mediated exon skipping for Duchenne muscular dystrophy mutations. Hum Mutat 2009;30(03): 293–299. Doi: 10.1002/humu.20918
    » https://doi.org/10.1002/humu.20918
  • 96
    Mendell JR, Rodino-Klapac LR, Sahenk Z, et al; Eteplirsen Study Group. Eteplirsen for the treatment of Duchenne muscular dystrophy. Ann Neurol 2013;74(05):637–647. Doi: 10.1002/ana.23982
    » https://doi.org/10.1002/ana.23982
  • 97
    Charleston JS, Schnell FJ, Dworzak J, et al. Eteplirsen treatment for Duchenne muscular dystrophy: Exon skipping and dystrophin production. Neurology 2018;90(24):e2146–e2154. Doi: 10.1212/WNL.0000000000005680
    » https://doi.org/10.1212/WNL.0000000000005680
  • 98
    Mendell JR, Goemans N, Lowes LP, et al; Eteplirsen Study Group and Telethon Foundation DMD Italian Network. Longitudinal effect of eteplirsen versus historical control on ambulation in Duchenne muscular dystrophy. Ann Neurol 2016;79(02): 257–271. Doi: 10.1002/ana.24555
    » https://doi.org/10.1002/ana.24555
  • 99
    Mendell JR, Khan N, Sha N, et al; Eteplirsen Study Group. Comparison of Long-term Ambulatory Function in Patients with Duchenne Muscular Dystrophy Treated with Eteplirsen and Matched Natural History Controls. J Neuromuscul Dis 2021;8(04):469–479. Doi: 10.3233/JND-200548
    » https://doi.org/10.3233/JND-200548
  • 100
    McDonald CM, Shieh PB, Abdel-Hamid HZ, et al; the Italian DMD Telethon Registry Study Group, Leuven NMRC Registry Investigators, CINRG Duchenne Natural History Investigators, and PROMOVI Trial Clinical Investigators. Open-Label Evaluation of Eteplirsen in Patients with Duchenne Muscular Dystrophy Amenable to Exon 51 Skipping: PROMOVI Trial. J Neuromuscul Dis 2021;8(06):989–1001. Doi: 10.3233/JND-210643
    » https://doi.org/10.3233/JND-210643
  • 101
    Anwar S, Yokota T. Golodirsen for Duchenne muscular dystrophy. Drugs Today (Barc) 2020;56(08):491–504. Doi: 10.1358/ dot.2020.56.8.3159186
    » https://doi.org/10.1358/dot.2020.56.8.3159186
  • 102
    Frank DE, Schnell FJ, Akana C, et al; SKIP-NMD Study Group. Increased dystrophin production with golodirsen in patients with Duchenne muscular dystrophy. Neurology 2020;94(21): e2270–e2282. Doi: 10.1212/WNL.0000000000009233
    » https://doi.org/10.1212/WNL.0000000000009233
  • 103
    Servais L, Mercuri E, Straub V, et al; SKIP-NMD Study Group. Long-Term Safety and Efficacy Data of Golodirsen in Ambulatory Patients with Duchenne Muscular Dystrophy Amenable to Exon 53 Skipping: A First-in-human, Multicenter, Two-Part, Open- Label, Phase 1/2 Trial. Nucleic Acid Ther 2022;32(01):29–39. Doi: 10.1089/nat.2021.0043
    » https://doi.org/10.1089/nat.2021.0043
  • 104
    Scaglioni D, Catapano F, Ellis M, et al. The administration ofantisense oligonucleotide golodirsen reduces pathological regeneration in patients with Duchenne muscular dystrophy. Acta Neuropathol Commun 2021;9(01):7. Doi: 10.1186/s40478-020-01106-1
    » https://doi.org/10.1186/s40478-020-01106-1
  • 105
    Clemens PR, Rao VK, Connolly AM, et al; CINRG DNHS Investigators. Safety, tolerability, and efficacy of viltolarsen in boys with Duchenne muscular dystrophy amenable to exon 53 skipping: A phase 2 randomized clinical trial. JAMA Neurol 2020;77(08):982–991
  • 106
    Clemens PR, Rao VK, Connolly AM, et al; CINRG DNHS Investigators. Efficacy and Safety of Viltolarsen in Boys With Duchenne Muscular Dystrophy: Results From the Phase 2, Open- Label, 4-Year Extension Study. J Neuromuscul Dis 2023;10(03): 439–447. Doi: 10.3233/JND-221656
    » https://doi.org/10.3233/JND-221656
  • 107
    Roy B, Friesen WJ, Tomizawa Y, et al. Ataluren stimulates ribosomal selection of near-cognate tRNAs to promote nonsense suppression. Proc Natl Acad Sci U S A 2016;113(44):12508–12513. Doi: 10.1073/pnas.1605336113
    » https://doi.org/10.1073/pnas.1605336113
  • 108
    Bushby K, Finkel R, Wong B, et al; PTC124-GD-007-DMD STUDY GROUP. Ataluren treatment of patients with nonsense mutation dystrophinopathy. Muscle Nerve 2014;50(04):477–487. Doi: 10.1002/mus.24332
    » https://doi.org/10.1002/mus.24332
  • 109
    McDonald CM, Campbell C, Torricelli RE, et al; Clinical Evaluator Training Group. ; ACT DMD Study Group. Ataluren in patients with nonsense mutation Duchenne muscular dystrophy (ACT DMD): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017;390(10101):1489–1498. Doi: 10.1016/S0140-6736(17)31611-2
    » https://doi.org/10.1016/S0140-6736(17)31611-2
  • 110
    Mercuri E, Osorio AN, Muntoni F, et al; STRIDE and CINRG DNHS investigators. Safety and effectiveness of ataluren in patients with nonsense mutation DMD in the STRIDE Registry compared with the CINRG Duchenne Natural History Study (2015-2022): 2022 interim analysis. J Neurol 2023;270(08):3896–3913. Doi: 10.1007/s00415-023-11687-1
    » https://doi.org/10.1007/s00415-023-11687-1
  • 111
    https://www.ema.europa.eu/en/news/ema-recommends-non-renewal-authorisation-duchenne-muscular-dystrophy-medicine-translarna
    » https://www.ema.europa.eu/en/news/ema-recommends-non-renewal-authorisation-duchenne-muscular-dystrophy-medicine-translarna

Publication Dates

  • Publication in this collection
    28 June 2024
  • Date of issue
    2024

History

  • Received
    08 Nov 2023
  • Accepted
    22 Nov 2023
Academia Brasileira de Neurologia - ABNEURO R. Vergueiro, 1353 sl.1404 - Ed. Top Towers Offices Torre Norte, 04101-000 São Paulo SP Brazil, Tel.: +55 11 5084-9463 | +55 11 5083-3876 - São Paulo - SP - Brazil
E-mail: revista.arquivos@abneuro.org