Abstract
Purpose of Review:
Amyotrophic lateral sclerosis (ALS) is a rapidly fatal disease for which there is currently no effective therapy. This review describes the current progress of existing molecular therapies in the clinical trial pipeline and highlights promising future antisense oligonucleotide and viral therapeutic strategies for treating ALS.
Recent Findings:
The immense progress in the design of clinical trials and generation of ASO therapies directed towards superoxide dismutase-1- (SOD1) and C9orf72 repeat expansion- related disease have been propelled by fundamental work to identify the genetic underpinnings of familial ALS and development of relevant disease models. Preclinical studies have also identified promising targets for sporadic ALS (sALS). Moreover, encouraging results in AAV9-based therapies for spinal muscular atrophy (SMA) provide a roadmap for continued improvement in delivery and design of molecular therapies for ALS.
Summary:
Advances in preclinical and clinical studies of ASO and viral directed approaches to neuromuscular disease, particularly ALS, indicate that these approaches have high specificity and are relatively safe.
Keywords: antisense oligonucleotide, amyotrophic lateral sclerosis, viral therapy, C9orf72, superoxide dismutase 1 (SOD1)
Introduction
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease characterized by motor neuron loss and paralysis. Although the majority of cases are singleton (“sporadic”), genetic advancements in the 10% of patients with familial ALS have provided valuable insights into disease pathomechanisms and pinpointed therapeutic targets in patients with specific gene mutations. Hexanucleotide repeat expansions in chromosome 9 open reading frame 72 (C9orf72) and mutations in superoxide dismutase-1 (SOD1) are the most common forms of hereditary ALS, constituting ~35–50% [1] and ~20% [2] of cases, respectively. For SOD1- and C9orf72-related ALS, the establishment of gain-of-function mechanisms as causes for disease and creation of reliable disease models have catalyzed generation of targeted antisense oligonucleotide [3] therapies that are now or are soon to be in clinical trials. Despite our increasing understanding of ALS pathogenesis, many barriers remain for generating effective therapeutic strategies for sporadic disease including a) the heterogeneity of disease mechanisms, b) incomplete understanding of cell autonomous and non-cell autonomous contributions to disease, and c) limitations in gene delivery methods.
Gene therapy approaches, including ASO and viral-directed gene delivery, have shown enormous potential to enable therapeutic modulation of gene expression in diverse monogenic neurological conditions from Huntington’s disease, myotonic dystrophy, spinal muscular atrophy, and ALS. ASOs are synthetic single-stranded deoxynucleic acid (DNA) sequences, 8 to 50 base pairs in length, that complement selected sequences of target ribonucleic acid (RNA) leading to mRNA degradation through RNase H enzyme recruitment. ASOs can also be modified to promote alternative splicing rather than causing mRNA degradation. Moreover, ASOs distribute widely throughout the central nervous system (CNS) when delivered intrathecally, a crucial feature for treatment of neurological disorders [4*].
On the other hand, viral-mediated gene delivery may offer additional flexibility for drug administration and provide longer-lasting genetic alterations. Viral vectors, such as adeno-associated virus (AAV), can provide greater ease in drug administration since they more readily cross the blood-brain barrier when given systemically and can be manipulated to favor tropism to specific cell types and structures [5, 6]. Furthermore, AAVs express stable extrachromosomal nuclear episomes, ensuring long-lasting gene expression [3].
In recent years, the application of these strategies in a form of motor neuron disease, spinal muscular atrophy (SMA), has led to FDA approval of nusinersen and recent initiation of Phase III trials of an AAV9-based AVXS-101 therapy (AveXis; NCT03461289; NCT03306277; NCT03505099). These successes in tandem with early advances in the development of ASOs in ALS serve as valuable guides for future therapeutic endeavors (Fig. 1).
Figure 1. Cycle of gene therapy development.

The development of gene therapies for ALS begins with 1) pinpointing promising disease targets which have historically derived from identification of genes in familial ALS and the discovery of aberrant cellular processes in affected patients or animal models. 2) Generation of cellular or animal disease models helps to clarify how gene mutations or pathologic features impact disease. 3) Mechanistic knowledge of disease pathogenesis derived from disease models informs development of a therapeutic strategy. Disease mechanisms governed by gain-of-function (i.e. SOD1-ALS, C9-ALS) may benefit from approaches that enable gene knockdown (i.e. ASOs, AAV-mediated delivery of ASOs, shRNA, or miRNA) while disease caused by loss-of-function could benefit from restoration of the defective gene using AAV. If gene therapies show 4) preclinical efficacy, they may progress to 5) human clinical trials. 6) The cumulative lessons derived from iterations of this cycle will hopefully streamline the process of drug development in ALS.
ASO therapeutics in SOD1-related ALS
The discovery of SOD1 mutations as a cause of familial ALS in 1993 precipitated a molecular revolution in our understanding of the disease. Similar to human SOD1 post-mortem tissue, multiple SOD1 rodent models exhibit SOD1 aggregates, TDP-43 inclusions, and motor neuron degeneration suggesting toxicity through a gain of function mechanism, whereas SOD1 knockout mice fail to develop motor neuron loss [7]. These cumulative findings indicated that SOD1 lowering could be a viable therapeutic strategy. Pioneering studies of SOD1 ASO in rats and non-human primates demonstrated widespread distribution of ASOs in CNS, effective knockdown of SOD1, and extension of survival [8].
A first-in-man Phase I double-blind, placebo-controlled clinical trial of intrathecally-delivered ASO 333611 (ISIS-SOD1RX) in SOD1-related ALS was completed in January 2012 ( NCT01041222). The study employed conservatively low single doses of ASO that did not modify SOD1 expression but established that the treatment approach was safe. Moreover, ISIS-SOD1RX concentrations measured in plasma, CSF, and even spinal cord autopsy tissue of one participant, provided valuable pharmacokinetic data [9].
A more potent second generation SOD1 targeting ASO, BIIB067 (IONIS-SOD1RX) [10**], is currently being tested in an ongoing Phase I A/B randomized, placebo-controlled escalating dose trial that began in late January 2016 and is planned to complete in February 2019 ( NCT02623699). The study includes participants with SOD1-related ALS and FVC > 50% with an enrollment target of 84. In part A, a single infusion of BIIB067 or placebo will be administered, while in part B, SOD1-ALS participants will receive multiple administrations of BIIB067. A Phase I long-term extension study of the effects of BIIB067 in participants who have completed part A/B is also ongoing and estimated to complete in January 2020 ( NCT03070119). The primary goal of these studies is to establish the safety profile and pharmacokinetics of BIIB067. Unlike the initial Phase I study, doses of BIIB067 are anticipated to be sufficiently high to lower SOD1 levels. Thus, an important secondary measure involves detection of SOD1 protein levels in CSF as a marker for target engagement [11]
Human SOD1 has a long half-life in CSF [12], a characteristic that limits how early traditional methods can measure ASO-mediated changes in patients. A promising approach for measuring early changes in SOD1 expression involves monitoring protein kinetics via stable isotope labeling combined with mass spectrometry, a method that provides high specificity and sensitivity [12]. Using this technique, measurements of newly synthesized SOD1 protein may provide earlier assessments of target engagement, a parameter that will serve as a valuable efficacy measure and help determine optimal frequency of dosing for future clinical trials.
As SOD1-targeting ASOs clinical trials advance, a critical consideration will be the challenge of recruiting participants with a rare genetic disorder. A recent natural history study of patients with SOD1-related ALS has provided guidance for design of future clinical trials and estimated enrollment goals needed to adequately assess therapeutic efficacy [13*]. Notably, median survival of SOD1 A4V mutation carriers was only 1.2 years compared to 6.8 years for non-A4V carriers, and 2.7 years for all SOD1 patients. Since A4V constitutes the most common SOD1 mutation in North America, and A4V patients are more clinically homogeneous, future therapeutic efficacy studies may consider stratifying results from these patients.
Gene targeting strategies in ALS associated with C9orf72 repeat expansions
Hexanucleotide repeat expansions in C9orf72 was only discovered to be the most common genetic cause of ALS and frontotemporal dementia (FTD) early this decade, yet tremendous progress has been made to understand how this mutation confers neuronal toxicity. Several mechanisms are thought to play a role. Firstly, the RNA encoding the expansion is transcribed bidirectionally and forms nuclear inclusions that cause gain-of-function toxicity. Secondly, through non-canonical repeat-associated non-ATG mediated (RAN) translation, up to five distinct aggregate-prone dipeptides can be generated from sense and antisense strands. Thirdly, C9orf72 haploinsufficiency has been implicated particularly given its newly appreciated roles in lysosomal maintenance and inflammatory regulation [14, 15].
Initial development of ASO-based therapeutics for C9orf72 was ushered by availability of induced pluripotent stem cell (iPSC) and fibroblast models from patients with C9orf72-related ALS. These early studies established design considerations for ASO development and focused on reducing gain-of-function toxicity associated with the repeat expansion. ASOs designed to bind within or immediately upstream of the intronic expansion did not significantly alter C9orf72 mRNA levels while those binding downstream of the expansion led to significant reduction of C9orf72. Regardless of the targeted region, ASOs reduced RNA foci, increased survival from glutamate excitotoxicity, and abrogated aberrant gene expression patterns [16–18]. Several transgenic mouse models expressing human C9orf72 repeat expansions recapitulate pathological disease features [14, 19, 20] and cause neurological deficits [19, 21]. In the first in vivo test of C9orf72 ASO, investigators demonstrated that single dose, intraventricular administration of ASO could attenuate RNA foci and dipeptide aggregates as well as improve the behavioral and cognitive deficits associated with the C9orf72 repeat expansion [19]. Furthermore, poly (GP) dipeptide can be detected in the CSF of C9orf72 expansion carriers and its levels are reduced with ASO treatment in experimental models indicating that it could serve as a marker of target engagement [22, 23]. These encouraging results provided a springboard for the initiation of a Phase I clinical trial of C9-ASO anticipated to start in the next year. These disease models will continue to aid in further refinement of ASO design, safety testing, biomarker development, and practical planning for future therapeutic trials.
Existing ASO strategies preferentially target sense strand transcripts. However, antisense RNA foci have also been identified in C9orf72 patient neuronal tissue and may also contribute to disease [17]. Thus, future ASO strategies may need to target toxic RNA transcribed from both directions in order to adequately treat the condition. One approach is to employ sense- and antisense-directed ASOs in tandem. Alternatively, recent studies suggest that knockdown of Spt4, a transcription elongation factor selectively involved in expression of expanded repeats, inhibits development of sense and antisense RNA foci and dipeptide inclusions in C9orf72 patient fibroblasts [24]. However, knockout of the mouse homolog, Supt4h, causes embryonic lethality [25]. Thus, clarifying the endogenous role of Spt4 will be necessary to determine the clinical viability of this strategy.
Recent studies indicate that both gain- and loss-of-function mechanisms may act in concert to cause pathogenesis in C9orf72-related disease. Arguing against loss-of-function, C9orf72-deficient mice do not display neurodegeneration [14, 15] and lowering of C9orf72 using ASOs in mice is well tolerated [17]. On the other hand, C9orf72 KO mice have a pronounced immune phenotype characterized by splenomegaly and leukocyte, particularly macrophage, abnormalities [15]. Given the prominent neuroinflammation in ALS, these findings suggest an underappreciated link between immune regulation and ALS. A recent study in iMNs suggested more directly that C9orf72 deficiency is related to neurodegeneration. Under conditions of excitotoxic or proteostatic stress, C9orf72-deficient iMNs undergo degeneration stemming from pleiotrophic consequences of perturbed vesicle trafficking including glutamate receptor accumulation and impaired lysosomal function [26**]. Thus, an integrated therapeutic approach to knockdown expansion-related mRNA and protein products as well as restore native C9orf72 expression may be necessary to fully address the cellular deficits in C9orf72-related disease.
ASOs to target TDP-43 pathology
Although ASOs demonstrate therapeutic promise for monogenic gain-of-function conditions such as SOD1- and C9orf72-related ALS, a challenge in developing treatments for the broader ALS population is the heterogeneity of cellular pathways disrupted. Notably, aggregates of the ribosomal protein, transactive response DNA binding protein 43 kDa (TDP-43), are a pathological hallmark of ~97% of ALS cases [27, 28] and mutations in TDP-43 that promote protein aggregation are also sufficient to cause disease [29]. In addition, the abundance of TDP-43 inclusions in mouse models that overexpress wild-type or mutant protein correspond to symptomatic burden [30, 31].
Recent studies suggest that knockdown of ataxin-2, a protein that regulates stress granule formation and promotes aggregation of TDP-43, may hold therapeutic promise. ASOs targeting ataxin-2 in mice expressing human TDP-43 were shown to have reduced formation of TDP-43 aggregates, slowed disease progression, and markedly prolonged survival [32**]. Ataxin-2 KO adult mice exhibit weight gain but are viable and fertile [33] while removal of TARDBP, the gene encoding TDP-43, causes embryonic lethality [34, 35], suggesting that direct manipulation TDP-43 may not be feasible. A recent study further showed that inhibiting stress granule formation using ataxin-2 ASOs or small molecules suppressed nucleocytoplasmic transport defects and neurodegeneration in C9-ALS iMNs and C9-ALS fly models [36**]. These pre-clinical data indicate that destabilizing stress granules by targeting ataxin-2 or other factors required for their assembly has potential to be beneficial in a broader ALS population.
Emerging viral-based therapeutics in ALS
Recent data using viral delivery in SMA are impressive and may provide lessons for implementation of viral-mediated strategies in ALS. A Phase I trial of a single-dose adenoviral intravenous gene replacement therapy of human survival motor neuron (hSMN) (AVXS-101) in SMA1 (AveXis; NCT02122952) demonstrated striking gains in motor milestones and longer survival compared to a historical cohort [37**]. Side effects included elevated transaminases in a subset of participants though safety and tolerability will continue to be examined in future studies. Enrollment has already been initiated for Phase III open-label, single-dose studies of AVXS-101 in a larger SMA I cohort (AveXis; NCT03461289; NCT03306277) and in a broader pre-symptomatic cohort with SMA types 1–3 (AveXis; NCT03505099) to further assess drug safety and efficacy. The AAV9-mediated therapy has been shown to cross the blood-brain barrier, target neurons diffusely within the spinal cord, and lead to sustained expression of hSMN [37, 38]. Thus, viral-mediated therapies may provide additional ease and versatility for gene delivery as it can be given systemically and may be efficacious with single or infrequent dosing.
Viral-based treatment strategies in ALS have predominantly been explored in pre-clinical studies. Similar to the ability of ASO-based strategies to reduce toxic gene products, viral vectors have also been modified to deliver RNA-based therapeutics to suppress gene expression. Adenoviral or lentiviral vectors carrying SOD1 short hairpin RNA (shRNA) [39–42], SOD1 micro-RNA (miRNA) [43, 44], or SOD1 ASO [45*] have been shown to suppress toxic mutant SOD1, slow disease progression, and/or improve survival in SOD1 G93A mice when given intravenously and/or intrathecally. Notably, AAV-based delivery of SOD1 directed miRNA in non-human primates has been shown to effectively silence SOD1 in the spinal cord setting the stage for development of future clinical trials [44].
A particularly valuable application of viral vectors is that they enable restoration or overexpression of genes. Neurotrophic factors are secreted proteins valuable to the growth and survival of multiple neuronal types and levels of various neurotrophic factors have been shown to decline in ALS patients and animal models [46]. Multiple approaches have been explored to deliver neurotrophic factors in ALS to favorably alter the cellular milieu surrounding motor neurons to impact their survival. This sort of intervention has the potential to modify both sporadic and familial forms of the disease. Over the years, various factors including glial-derived neurotrophic factor (GDNF), insulin-like growth factor 1/2 (IGF-1/2), vascular endothelial growth factor (VEGF) have been shown to provide benefit when virally delivered using various administration routes in pre-clinical models of ALS [47*]. Moreover, the delivery method and cell types targeted have been shown to have a significant impact on the therapeutic benefit gained from this strategy. For instance, although systemic delivery of AAV9-GDNF in G93A rats failed to extend survival [48*], specific induction of GDNF expression in muscle was shown to prolong disease progression and increase survival [49, 50].
Adenoviral gene therapy has also provided a tool for clarifying the therapeutic potential of targeting molecular and pathological derangements observed in sporadic ALS. For instance, the “dying back” of motor nerve terminals is a common pathological feature observed in ALS model mice and patients. In a recent study, SOD1 G93A mice were given systemic gene therapy to express Dok7, a regulator of synaptogenesis. Systemic viral delivery of the gene was found to reduce motor nerve denervation and prolonged lifespan, suggesting that stabilizing the neuromuscular junction (NMJ) in ALS may provide additional therapeutic benefit, particularly if combined with other treatment strategies [51*]. Glutamatergic excititoxicity is another pathway widely implicated in sporadic and familial ALS. Accumulation of D-serine, an NMDA receptor co-agonist that can facilitate receptor overactivation, has been seen in sporadic ALS and SOD1 model mice. D-serine is degraded by D-amino acid oxidase (DAO), an enzyme that has lowered expression in ALS model mice. Increasing DAO expression in G93A mice using a single intrathecal injection of ssAAV9-DAO reduced D-serine in the spinal cord, lessened motor neuron loss, and extended survival [52*]. The durability and versatility of viral-based gene therapies motivates further exploration of these strategies in ALS pre-clinical and clinical trials.
Conclusion
Considerable strides in developing gene therapy for ALS and SMA have been made in recent years. ASO-based therapeutics for familial ALS caused by SOD1 and C9orf72 mutations have shown promise in pre-clinical studies and continue to advance in the clinical pipeline. Success of viral-based gene delivery in SMA highlights its potential to improve ease of drug administration and expand possibilities for gene modification. The complexity and heterogeneity of cellular processes disrupted in sporadic ALS is a significant challenge for therapeutic development. However, targeting pathological features broadly observed in ALS, such as TDP-43 inclusions, provides an intriguing approach. In addition, combining gene therapies to target independent processes could provide synergistic benefit [53*].
Key Points:
ASO therapies for SOD1 and C9orf72-related famililal ALS have shown promise in pre-clinical studies and have progressed to early clinical trials
Advances in viral-based gene therapy for SMA suggests that this approach may enable durable and versatile gene manipulation in other motor neuron diseases such as ALS.
Pre-clinical gene therapy studies targeting multiple processes commonly disrupted in sporadic ALS show benefit.
Financial Support and sponsorship:
This work is supported by a Clinical Research Training Fellowship funded by the ALS Association to C.V.L. and NIH NINDS R01 NS078398-07 to T.M.M.
Footnotes
Conflicts of Interest:
Dr. Ly has no conflicts to disclose. Washington University has equity ownership interest in C2N Diagnostics and may receive royalty income based on technology licensed by Washington University to C2N Diagnostics. C2N has licensed IP regarding protein kinetics measurements. Washington University has submitted a nonprovisional patent application “SOD1 kinetics measurements,” (T.M.M.). Dr. Miller has served on a medical advisory board for Biogen. Ionis pharmaceuticals provided support for research. Dr. Miller serves as a consultant for Cytokinetics.
References:
- 1.Majounie E, Renton AE, Mok K, et al. Frequency of the C9orf72 hexanucleotide repeat expansion in patients with amyotrophic lateral sclerosis and frontotemporal dementia: a cross-sectional study. The Lancet Neurology. 2012;11(4):323–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Pasinelli P, Brown RH. Molecular biology of amyotrophic lateral sclerosis: insights from genetics. Nature reviews Neuroscience. 2006;7(9):710–23. [DOI] [PubMed] [Google Scholar]
- 3.Murlidharan G, Samulski RJ, Asokan A. Biology of adeno-associated viral vectors in the central nervous system. Frontiers in molecular neuroscience. 2014;7:76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.*.Schoch KM, Miller TM. Antisense Oligonucleotides: Translation from Mouse Models to Human Neurodegenerative Diseases. Neuron. 2017;94(6):1056–70. [DOI] [PMC free article] [PubMed] [Google Scholar]; This review provides a thorough overview of ASO chemistry, mechanisms of action, and application in neurodegenerative disease.
- 5.Bedbrook CN, Deverman BE, Gradinaru V. Viral Strategies for Targeting the Central and Peripheral Nervous Systems. Annual review of neuroscience. 2018. [DOI] [PubMed] [Google Scholar]
- 6.Srivastava A In vivo tissue-tropism of adeno-associated viral vectors. Current opinion in virology. 2016;21:75–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Picher-Martel V, Valdmanis PN, Gould PV, et al. From animal models to human disease: a genetic approach for personalized medicine in ALS. Acta neuropathologica communications. 2016;4(1):70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Smith RA, Miller TM, Yamanaka K, et al. Antisense oligonucleotide therapy for neurodegenerative disease. The Journal of clinical investigation. 2006;116(8):2290–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Miller TM, Pestronk A, David W, et al. An antisense oligonucleotide against SOD1 delivered intrathecally for patients with SOD1 familial amyotrophic lateral sclerosis: a phase 1, randomised, first-in-man study. The Lancet Neurology. 2013;12(5):435–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.**.McCampbell A, Cole T, Wegener A, et al. Antisense Oligonucleotides Extend Survival and Reverse Decrement in Muscle Response in ALS Models. Journal of Clinical Investigation. 2018. (in press). [DOI] [PMC free article] [PubMed] [Google Scholar]; This study shows that next generation SOD1 ASOs used in current clinical trials more potently reduce SOD1 expression, extend survival, and show evidence of disease reversal in SOD1 G93A rodent models.
- 11.Winer L, Srinivasan D, Chun S, et al. SOD1 in cerebral spinal fluid as a pharmacodynamic marker for antisense oligonucleotide therapy. JAMA neurology. 2013;70(2):201–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Crisp MJ, Mawuenyega KG, Patterson BW, et al. In vivo kinetic approach reveals slow SOD1 turnover in the CNS. The Journal of clinical investigation. 2015;125(7):2772–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.*.Bali T, Self W, Liu J, et al. Defining SOD1 ALS natural history to guide therapeutic clinical trial design. Journal of neurology, neurosurgery, and psychiatry. 2017;88(2):99–105. [DOI] [PMC free article] [PubMed] [Google Scholar]; This article provides evidence that A4V SOD1 is a relatively homogenous population with a short median survival, a finding pertinent to design and implementation of treatment trials in the SOD1-ALS population.
- 14.O’Rourke JG, Bogdanik L, Yanez A, et al. C9orf72 is required for proper macrophage and microglial function in mice. Science. 2016;351(6279):1324–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Burberry A, Suzuki N, Wang JY, et al. Loss-of-function mutations in the C9ORF72 mouse ortholog cause fatal autoimmune disease. Science translational medicine. 2016;8(347):347ra93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Donnelly CJ, Zhang PW, Pham JT, et al. RNA toxicity from the ALS/FTD C9ORF72 expansion is mitigated by antisense intervention. Neuron. 2013;80(2):415–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Lagier-Tourenne C, Baughn M, Rigo F, et al. Targeted degradation of sense and antisense C9orf72 RNA foci as therapy for ALS and frontotemporal degeneration. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(47):E4530–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Sareen D, O’Rourke JG, Meera P, et al. Targeting RNA foci in iPSC-derived motor neurons from ALS patients with a C9ORF72 repeat expansion. Science translational medicine. 2013;5(208):208ra149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Jiang J, Zhu Q, Gendron TF, et al. Gain of Toxicity from ALS/FTD-Linked Repeat Expansions in C9ORF72 Is Alleviated by Antisense Oligonucleotides Targeting GGGGCC-Containing RNAs. Neuron. 2016;90(3):535–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Peters OM, Cabrera GT, Tran H, et al. Human C9ORF72 Hexanucleotide Expansion Reproduces RNA Foci and Dipeptide Repeat Proteins but Not Neurodegeneration in BAC Transgenic Mice. Neuron. 2015;88(5):902–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Liu Y, Pattamatta A, Zu T, et al. C9orf72 BAC Mouse Model with Motor Deficits and Neurodegenerative Features of ALS/FTD. Neuron. 2016;90(3):521–34. [DOI] [PubMed] [Google Scholar]
- 22.**.Gendron TF, Chew J, Stankowski JN, et al. Poly(GP) proteins are a useful pharmacodynamic marker for C9ORF72-associated amyotrophic lateral sclerosis. Science translational medicine. 2017;9(383). [DOI] [PMC free article] [PubMed] [Google Scholar]; This study determined that poly (GP) dipeptide could be detected in the CSF of C9orf72 expansion carriers and had potential as a pharmacodynamics biomarker for the disease.
- 23.Ash PE, Bieniek KF, Gendron TF, et al. Unconventional translation of C9ORF72 GGGGCC expansion generates insoluble polypeptides specific to c9FTD/ALS. Neuron. 2013;77(4):639–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Kramer NJ, Carlomagno Y, Zhang YJ, et al. Spt4 selectively regulates the expression of C9orf72 sense and antisense mutant transcripts. Science. 2016;353(6300):708–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Cheng HM, Chern Y, Chen IH, et al. Effects on murine behavior and lifespan of selectively decreasing expression of mutant huntingtin allele by supt4h knockdown. PLoS genetics. 2015;11(3):e1005043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.**.Shi Y, Lin S, Staats KA, et al. Haploinsufficiency leads to neurodegeneration in C9ORF72 ALS/FTD human induced motor neurons. Nature medicine. 2018;24(3):313–25. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study indicates that C9orf72 haploinsufficiency impairs vesicle trafficking and renders neurons vulnerable to excitotoxic and proteostatic stressors, suggesting that loss-of-function mechnisms also play a role in disease.
- 27.Neumann M, Sampathu DM, Kwong LK, et al. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science. 2006;314(5796):130–3. [DOI] [PubMed] [Google Scholar]
- 28.Ling SC, Polymenidou M, Cleveland DW. Converging mechanisms in ALS and FTD: disrupted RNA and protein homeostasis. Neuron. 2013;79(3):416–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Sreedharan J, Blair IP, Tripathi VB, et al. TDP-43 mutations in familial and sporadic amyotrophic lateral sclerosis. Science. 2008;319(5870):1668–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Wils H, Kleinberger G, Janssens J, et al. TDP-43 transgenic mice develop spastic paralysis and neuronal inclusions characteristic of ALS and frontotemporal lobar degeneration. Proceedings of the National Academy of Sciences of the United States of America. 2010;107(8):3858–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Janssens J, Wils H, Kleinberger G, et al. Overexpression of ALS-associated p.M337V human TDP-43 in mice worsens disease features compared to wild-type human TDP-43 mice. Molecular neurobiology. 2013;48(1):22–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.**.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):367–71. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study provides pre-clinical evidence that ASO-mediated knockdown of ataxin-2 in a TDP-43 mouse model reduces TDP-43 inclusions and prolongs survival.
- 33.Kiehl TR, Nechiporuk A, Figueroa KP, et al. Generation and characterization of Sca2 (ataxin-2) knockout mice. Biochemical and biophysical research communications. 2006;339(1):17–24. [DOI] [PubMed] [Google Scholar]
- 34.Sephton CF, Good SK, Atkin S, et al. TDP-43 is a developmentally regulated protein essential for early embryonic development. The Journal of biological chemistry. 2010;285(9):6826–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Wu LS, Cheng WC, Hou SC, et al. TDP-43, a neuro-pathosignature factor, is essential for early mouse embryogenesis. Genesis. 2010;48(1):56–62. [DOI] [PubMed] [Google Scholar]
- 36.**.Zhang K, Daigle JG, Cunningham KM, et al. Stress Granule Assembly Disrupts Nucleocytoplasmic Transport. Cell. 2018;173(4):958–71. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study showed that inhibiting stress granule formation in C9orf72 disease models using small molecule inhibitors or ataxin ASO improved nucleocytoplasmic transport deficits and neurodegeneration.
- 37.**.Mendell JR, Al-Zaidy S, Shell R, et al. Single-Dose Gene-Replacement Therapy for Spinal Muscular Atrophy. The New England journal of medicine. 2017;377(18):1713–22. [DOI] [PubMed] [Google Scholar]; This article details the initial clinical trial of single-dose scAAV-SMN therapy in patients with SMA1 that showed that these patients had improved motor outcomes and survival compared to a historic cohort.
- 38.Foust KD, Nurre E, Montgomery CL, et al. Intravascular AAV9 preferentially targets neonatal neurons and adult astrocytes. Nature biotechnology. 2009;27(1):59–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Foust KD, Salazar DL, Likhite S, et al. Therapeutic AAV9-mediated suppression of mutant SOD1 slows disease progression and extends survival in models of inherited ALS. Molecular therapy : the journal of the American Society of Gene Therapy. 2013;21(12):2148–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Miller TM, Kaspar BK, Kops GJ, et al. Virus-delivered small RNA silencing sustains strength in amyotrophic lateral sclerosis. Annals of neurology. 2005;57(5):773–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Ralph GS, Radcliffe PA, Day DM, et al. Silencing mutant SOD1 using RNAi protects against neurodegeneration and extends survival in an ALS model. Nature medicine. 2005;11(4):429–33. [DOI] [PubMed] [Google Scholar]
- 42.Raoul C, Abbas-Terki T, Bensadoun JC, et al. Lentiviral-mediated silencing of SOD1 through RNA interference retards disease onset and progression in a mouse model of ALS. Nature medicine. 2005;11(4):423–8. [DOI] [PubMed] [Google Scholar]
- 43.Wang H, Yang B, Qiu L, et al. Widespread spinal cord transduction by intrathecal injection of rAAV delivers efficacious RNAi therapy for amyotrophic lateral sclerosis. Human molecular genetics. 2014;23(3):668–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Borel F, Gernoux G, Cardozo B, et al. Therapeutic rAAVrh10 Mediated SOD1 Silencing in Adult SOD1(G93A) Mice and Nonhuman Primates. Human gene therapy. 2016;27(1):19–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.*.Biferi MG, Cohen-Tannoudji M, Cappelletto A, et al. A New AAV10-U7-Mediated Gene Therapy Prolongs Survival and Restores Function in an ALS Mouse Model. Molecular therapy : the journal of the American Society of Gene Therapy. 2017;25(9):2038–52. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study shows that antisense sequences to SOD1 embedded in a U7 small nuclear RNA backbone and delivered by AAV10 can be used to reduce SOD1 levels and incraese survival of SOD1 G93A mice.
- 46.Krakora D, Macrander C, Suzuki M. Neuromuscular junction protection for the potential treatment of amyotrophic lateral sclerosis. Neurology research international. 2012;2012:379657. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.*.Tosolini AP, Sleigh JN. Motor Neuron Gene Therapy: Lessons from Spinal Muscular Atrophy for Amyotrophic Lateral Sclerosis. Frontiers in molecular neuroscience. 2017;10:405. [DOI] [PMC free article] [PubMed] [Google Scholar]; This review provides a thorough historical context for gene therapy development in motor neuron diseases and extracts practical considerations for future drug development.
- 48.*.Thomsen GM, Alkaslasi M, Vit JP, et al. Systemic injection of AAV9-GDNF provides modest functional improvements in the SOD1(G93A) ALS rat but has adverse side effects. Gene therapy. 2017;24(4):245–52. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study provides evidence that systemic delivery of AAV9-GDNF fails to extend survival and is associated with slower weight gain and cognitive deficits.
- 49.Acsadi G, Anguelov RA, Yang H, et al. Increased survival and function of SOD1 mice after glial cell-derived neurotrophic factor gene therapy. Human gene therapy. 2002;13(9):1047–59. [DOI] [PubMed] [Google Scholar]
- 50.Wang LJ, Lu YY, Muramatsu S, et al. Neuroprotective effects of glial cell line-derived neurotrophic factor mediated by an adeno-associated virus vector in a transgenic animal model of amyotrophic lateral sclerosis. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2002;22(16):6920–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.*.Miyoshi S, Tezuka T, Arimura S, et al. DOK7 gene therapy enhances motor activity and life span in ALS model mice. EMBO molecular medicine. 2017;9(7):880–9. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study demonstrates that stabilizing neuromuscular junctions via adminstration of AAV9-DOK7 improves motor performance and survival in SOD1 G93A mice.
- 52.*.Wang W, Duan W, Wang Y, et al. Intrathecal Delivery of ssAAV9-DAO Extends Survival in SOD1(G93A) ALS Mice. Neurochemical research. 2017;42(4):986–96. [DOI] [PubMed] [Google Scholar]; This study shows that administration of ssAAV9-DAO reduced motor neuron loss and extended survival in SOD1 G93A mice.
- 53.*.Frakes AE, Braun L, Ferraiuolo L, et al. Additive amelioration of ALS by co-targeting independent pathogenic mechanisms. Annals of clinical and translational neurology. 2017;4(2):76–86. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study demonstrated that using a combinatorial therapeutic approach to target independent mechanisms and cell populations in SOD1 G93A mice can provide additive benefit.
