Next Article in Journal
Management of Displaced Midshaft Clavicle Fractures with Figure-of-Eight Bandage: The Impact of Residual Shortening on Shoulder Function
Previous Article in Journal
Assessing the Need for Semantic Data Integration for Surgical Biobanks—A Knowledge Representation Perspective
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Advent of Omics Sciences in Clinical Trials of Motor Neuron Diseases

by
Paola Ruffo
1,
Sebastiano Cavallaro
2,† and
Francesca Luisa Conforti
1,*,†
1
Medical Genetics Laboratory, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
2
Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), 95126 Catania, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
J. Pers. Med. 2022, 12(5), 758; https://doi.org/10.3390/jpm12050758
Submission received: 15 April 2022 / Revised: 3 May 2022 / Accepted: 5 May 2022 / Published: 7 May 2022
(This article belongs to the Section Omics/Informatics)

Abstract

:
The “omics revolution” has totally changed the scientific research approach and is contributing to the development of personalized therapies. In motor neuron diseases (MNDs), a set of complex, multifactorial, late-onset and chronic neurodegenerative diseases, the use of multi-omics approaches in clinical trials is providing new opportunities to stratify patients and develop target therapies. To show how omics science is gaining momentum in MNDs, in this work, we review the interventional clinical trials for MNDs based on the application of omics sciences. We analyze a total of 62 clinical trials listed in the ClinicalTrials database where different omics approaches have been applied in an initial phase, for diagnosis or patient selection, or in subsequent stages to cluster subjects, identify molecular signatures or evaluate drugs security or efficacy. The rise of omics sciences in clinical experimentation of MNDs is leading to an upheaval in their diagnosis and therapy that will require significant investments and means to ensure the correct and rapid evolution of personalized medicine.

Graphical Abstract

1. Introduction

The Human Genome Project (HGP), devised in the 1980s, began in 1990 and completed in 2003, made it possible to map and sequence the entire human genome. The rapid advancement of knowledge, together with high-throughput sequencing technologies, have laid the foundation of the omics sciences. Several research areas can be classified as omics sciences: genomics examines the genetic composition, transcriptomics investigates gene expression, proteomics focuses on the final protein product, metabolomics analyses metabolites and epigenomics evaluates epigenetic modifications.
In the last years, the application of omics technologies to clinical research has provided a more comprehensive understanding of the pathogenesis of complex diseases and the identification of biomarkers for patient stratification is simplifying the development of target therapies [1]. The field of precision and personalized medicine is, to date, dominated by oncology where the ability to obtain small biopsies allows to screen for somatic mutations and orient a target therapy [2]. In other fields, such as neurology, the same precision medicine approach is more difficult to achieve. In neurological disorders, neural tissue can be biopsied only post-mortem and the identification of molecular traits is more complex [3]. Nonetheless, the application of omics approaches to neurological disorders is deciphering their molecular signatures, allowing the stratification of patients. This approach is also offering the possibility of repurposing disease-modifying drugs or identifying new personalized solutions, evaluating both the useful and adverse effects of drugs.
MNDs represent a group of neurological progressive and degenerative disorders, which selectively affect motor neurons in brain and spinal cord [4]. They include primary lateral sclerosis (PLS), progressive muscular atrophy (PMA), progressive bulbar palsy (PBP), spinal muscular atrophy (SMA), amyotrophic lateral sclerosis (ALS) and spinobulbar muscular atrophy or Kennedy’s disease (SBMA). Although MNDs have quite different aetiologies, the recent explosion in omics sciences is attracting a great deal of clinical research to this field.
To show how omics science is gaining momentum in MNDs, in this manuscript we highlight the rise of the “omics” approach in clinical trials. Following a brief introduction to the MNDs of interest, we review all omics-based clinical trials performed to date to investigate the diagnostic and therapeutic complexity of these rare diseases.

2. Clinical Trials in Motor Neuron Diseases

The term “clinical trial” defines a human clinical pharmacological, biomedical or health study that follows predefined protocols. Clinical studies can be observational or interventional: the first tends to demonstrate the possible effects of various risk or protective factors on a group of people by observing events that occur without any intervention by the investigator. Interventional studies are research studies that aim to evaluate a new treatment (therapeutic, diagnostic or surgical), with the aim of evaluating if this represents an improvement compared to what is normally practiced.
Once authorized and before its completion or the publication of results, clinical trials can be searched using dedicated databases, such as ClinicalTrials (available online: https://www.clinicaltrials.gov/, 22 April 2022). To date, 62 omics-based clinical trials for MNDs can be found in this database: 41 (66.1%) are for ALS (Table 1), 20 (32.3%) for SMA (Table 2) and one (1.6%) for Kennedy’s disease. The first trials begun in 2006 and, for ALS, there has been a significant surge in recent years (Figure 1).
Among these clinical trials, 15 (24.2%) studies are currently in Phase I-II, in which the drug’s safety and toxicity as well as the effect produced by the treatment on the pathology under study are assessed; 36 (58.1%) studies are in Phase II-III, in which the drug is tested in a wider number of patients; whereas only one (1.61%) study is in Phase IV, called post-marketing surveillance trial. A genomic approach is used in 23 (37.10%) clinical trials for patient stratification; transcriptomics is used in 16 (25.81%) clinical studies and aims to monitoring therapy, while the remaining 23 (37.10%) trials integrate a multi-omics approach to evaluate the effect of treatments. Finally, grouping all trials according to the purpose for which the omics approach was used, we find that 52 (83.87%) studies aim at drug testing using a high-throughput technology, of which 17 (27.42%) trials aim at patient stratification and 35 (56.45%) at the evaluation of pre- or post-treatment clinical outcomes. The remaining 10 (16.13%) studies use different omics technologies to investigate disease molecular signatures, identify new biomarkers or build repositories.
In the next sections, we will discuss these clinical trials mainly focusing on those completed with published results that permit the evaluation of the usefulness of the omics approaches utilized.

3. Amyotrophic Lateral Sclerosis

Amyotrophic Lateral Sclerosis (ALS) is a neurodegenerative disease caused by the progressive loss of motor neurons (MNs) resulting in weakness and paralysis of voluntary muscles. The main clinical feature of ALS is the upper and lower MN involvement. The age of onset is about 60 years, and the incidence is 5 per 100,000 inhabitants [5].
Although important research progress has been made, the etiopathology of ALS is mostly unknown. The mechanisms underlying the development of the disease are multiple, with the involvement of a complex interaction between genetic and molecular characteristics [6]. The major ALS-related genes include superoxide dismutase 1 (SOD1), FUSed in sarcoma (FUS), TAR DNA binding protein (TARDBP) and chromosome 9 open reading frame 72 (C9Orf72) [7,8]. Due to the high complexity of the disease, the diagnosis is made by exclusion and there are no effective drug therapies that can stop or significantly slow down the progression of the disease. To date, the drugs used to slow down the course of the disease are Riluzole [9], which works by reducing excitotoxicity, and Edaravone [10] that decreases oxidative stress.
Neuropathology and medical genetics have led to the discovery that ALS and Frontotemporal Dementia (FTD) are related diseases and form a broad neurodegenerative continuum [11]. Both of these pathologies can be caused by mutations in the same gene that can lead to different clinical pictures [12]. The discovery of hexanucleotide expansion involving the C9Orf72 gene helped to define a genetic basis to explain the spectrum ALS/FTD [13].

3.1. Drug Development

The application of omic sciences has enabled the development of novel approaches to understand the molecular nature of ALS. As described in the following sections, their use has potential clinical implications to stratify patients and identify effective and safe treatments [14,15].

3.1.1. Omics Approach for Patients Stratifications

A genomic approach has been used in different clinical trials to select patients with specific drug-targetable gene mutations. Trial NCT01041222 was the first to use an intrathecally injected antisense oligonucleotide (ASO) designed to inhibit SOD1 expression in SOD1-fALS mutation carriers. The results revealed a successful strategy showing that the drug (ISIS 333611) was well tolerated [16]. A similar genetic stratification was applied in trial NCT00706147, where a genotype-phenotype homogeneous population of SOD1-fALS mutation carriers were used to test the safety, tolerability and efficacy of Arimoclomol, a drug promoting the correct folding of proteins. The study demonstrated drug tolerability and safety but did not show therapeutic efficacy [17]. In trial NCT04494256, subjects with ATXN2 expansion were enrolled to assess the safety, tolerability and pharmacokinetic profile of BIIB105, an ASO designed to bind and degrade the ATXN2 mRNA.

3.1.2. Omics Approach for Monitoring

A transcriptomic approach has been used to evaluate pre- and post-treatment gene expression changes in ALS patients [18]. In trial NCT04632225, RNA sequencing was used to determine and compare transcription profiles in patients receiving Engensis or placebo. Engensis is a gene therapy based on a plasmid that allows direct expression of the hepatocyte growth factor (HGF) in nerve cells of ALS patients. In trial NCT03359538, the effects of rapamycin in addition to riluzole in ALS patients was assessed by evaluating pathways related to immune response in plasma and cerebrospinal fluid (CSF). A transcriptomic and metabolomic approach was used in trial NCT00875446 to test the safety, pharmacokinetics and pharmacodynamics of the monoclonal antibody GSK1223249 (Ozanezumab), targeting the myelin-associate neurite outgrowth inhibitor (NOGO-A) in ALS patients. The results of this study, conducted on muscle biopsy and plasma by DNA microarray technology, demonstrated the drug is well tolerated although no drug-dependent gene expression patterns in muscle or plasma were identified [19]. The same approach was exploited in trial NCT03456882 to identify pharmacodynamic biomarkers following treatment with RNS60, an electrokinetically altered aqueous fluid.
In several clinical trials, the drug’s impact was determined by comparing the expression of different markers in patients exposed to drugs or placebo (NCT01854294, NCT03800524, NCT04505358, NCT03693781) or observing pre- and post-treatment clinical outcomes (NCT01884571, NCT02525471, NCT02469896, NCT05193994, NCT04788745). Notably, the results of trial NCT01854294 showed that the master regulator peptide GM604 altered plasma expression levels of SOD1, TAU and TDP-43 proteins, slowing down disease progression [20]. In trial NCT01884571, analysis of mRNA expression profiles in blood T-cells was used to assess the effect of immunosuppression and showed no disease-modifying effect following this treatment [21]. By evaluating the effects of Tocilizumab, a monoclonal antibody that inhibits Interleukin-6, trial NCT02469896 demonstrated dysregulation of pro-inflammatory genes in peripheral blood mononuclear cells of sporadic ALS patients [22].
Although discoveries related to the molecular basis of a disease may offer unprecedented opportunities to translate into new drugs, their development requires an enormous amount of time, money, and effort. For this reason, some trials have the objective to repurposing existing drugs to new disease areas [23]. Based on this strategy, the pharmacological properties of Enoxacin were assessed in ALS patients in trial NCT04840823. By acting against bacterial DNA topoisomerase II, this antibacterial agent is used in the treatment of urinary tract infections [24]. Since Enoxacin may regulate the expression of miRNAs, this trial aims to evaluate the ability of this drug in modulating miRNA species in CSF and plasma of ALS patients. Another repurposing drug strategy has been exploited in trial NCT02437110 where the combination of Darunavir, Ritonavir, Dolutegravir, and Tenofovir alafenamide (an antiretroviral therapy approved for human immunodeficiency virus infection) has been tested to suppress the activation of Human Endogenous Retrovirus-K (HERV-K) in ALS patients.
Clinical trial NCT04066244 proposed a multi-omics approach, the NCT04066244 to characterize the safety, tolerability and response of BLZ945, a Colony Stimulating Factor 1 (CSF-1) inhibitor. The outcomes of this trial will be based on genotyping cytochrome P4502C8 (CYP2C8), an enzyme involved in metabolism of xenobiotics.

3.1.3. Multi-Omics Approach for Both Stratification and Monitoring

A genomic, metabolomic and transcriptomic approach has been used to select patients and monitoring therapy. In trials NCT03626012 and NCT04288856, C9Orf72 patients were enrolled to evaluate the safety, tolerability and pharmacokinetics of BIIB078, an ASO targeting C9Orf72 mRNA. A similar approach was applied in trials NCT05163886 and NCT05053035, where C9Orf72 patients were recruited to test the safety, tolerability and biological effect of LAM-002A, (apilimod) an inhibitor of the PIKfyve kinase that works by clearing toxic protein aggregates within lysosomes. The outcome of this trial was to evaluate LAM-002 levels, metabolites and neurofilament light chain (NfL) in plasma and CSF.
Clinical trial NCT04993755 aimed to characterize the safety and tolerability of the reverse transcriptase enzyme inhibitor TPN-101 (censavudine) in C9Orf72 patients with ALS/FTD, ALS or FTD. A similar patient’s classification was used in trial NCT04931862 to test the effect of WVE-004, an ASO designed to mediate the degradation of C9Orf72 mRNA.
A genetic stratification was applied in trial NCT02623699, where SOD1-patients were enrolled to test BIIB067 (tofersen), an ASO designed to degrade SOD1 mRNA and to prevent protein synthesis. The results of this study confirmed drug safety and revealed drug-dependent SOD1 expression levels in CSF [25]. The same cohort of patients was used in the extension study NCT03070119 to test long-term treatment with BIIB067. The placebo-controlled trial NCT04856982, proposed to initiate BIIB067 treatment in clinically presymptomatic patients. A similar genetic stratification was applied in trial NCT01083667, where SOD1 mutation carriers were used to test the antiparasitic drug, Pyrimethamine (Daraprim). The results of this study demonstrated a significant reduction of SOD1 protein concentrations in CSF [26].
A genomic and metabolomic approach was used in trial NCT04768972 to evaluate the safety, pharmacokinetics and pharmacodynamics of ION363 in FUS-mutation carriers. This drug is an ASO designed to reduce the production of mutated neurotoxic form of FUS protein. This successful strategy showed a reduction in CSF of NfL concentration. Using a repurposing drug strategy, FUS patients were recruited in trial NCT03707795 to evaluate the effect of Betamethasone by assessing levels of different biomarkers in plasma. A similar strategy was exploited in trial NCT05189106, where the reduction of CSF inflammatory biomarkers was assessed in patients with Alzheimer’s disease and C9Orf72-ALS carriers following treatment with Baricitinib, a JAK inhibitor approved for rheumatoid arthritis and COVID-19 treatment [27].
Clinical trial NCT04220021 used a genomic and transcriptomic approach to evaluate safety and tolerability of Metformin in C9Orf72- patients. Since this drug, approved as a diabetes mellitus remedy, demonstrated beneficial effects through different signalling pathways [28], the trial aim was to evaluate CSF deregulated Repeat-associated non-AUG (RAN) protein levels.

3.2. Not Drug Related Clinical Trials

A multi-omics approach has been used in trail NCT02590276 to identify disease signatures in FTD/ALS C9Orf72-carriers [29]. The results implicated dysregulated circulating miRNAs as biomarkers of disease progression [20]. In trial NCT03984708, analysis of metabolic status and associated metabolic pathways in skin biopsy fibroblasts of ALS patients was used to find new therapeutic strategies.
A transcriptomic approach has been used in several clinical studies. In trial NCT01984957, DNA microarray technology was used to identify dysregulated gene expression in muscle biopsies of ALS patients. Combined omics technologies were applied in trial NCT02670226 to identify novel biomarkers in ALS patients by exploration of blood, muscle and satellite cells metabolomes and transcriptomes. A similar approach was applied in clinical trial NCT03851302, where mRNA expression of metabolism genes was assessed in muscle of ALS patients following neuromuscular magnetic stimulation (NMMS).

4. Spinal Muscular Atrophy

SMA is a disease characterized by motor neuron degeneration of the spinal cord anterior horns resulting in muscle atrophy and weakness of the trunk and limbs. Based on the age of onset and the severity of symptoms, four different SMA forms can be distinguished: type 1 SMA (SMA1) is the most severe and appears from 6 months of age; type 2 SMA (SMA2) become visible indicatively, between 6 and 18 months of life; type 3 SMA (SMA3) starts after 12 months of age, while type 4 SMA (SMA4) begins in adulthood and is the least severe form. In 95% of cases, the disease is caused by specific mutations in the SMN1 gene, essential for the function and survival of motor neurons. Patients with SMA have a variable number of second gene copies, SMN2, which encodes a shortened form of the SMN protein, with reduced functionality compared to the full SMN protein encoded by the healthy SMN1 gene. The number of copies of the SMN2 gene is therefore the basis of the great variability of the disease, with more or less severe forms and a very wide range of symptoms [30]. Due to disease’s rarity, the diagnosis is based on the history and clinical examination of the patients and can be confirmed by appropriate genetic tests. Until recently, treatment of SMA was exclusively symptomatic, based on multidisciplinary approaches aimed at improving the quality of life of patients. Today, however, several specific therapies have been approved for this disease [31].

4.1. Drug Development

4.1.1. Omics Approach for Patient Stratifications

A genomic approach for patient stratification has been used in several studies to evaluate the safety, tolerability and pharmacokinetics of a SMN gene-targeted therapy. Trail NCT02122952 was the first to test a replacement gene therapy, based on a self-complementary adeno-associated viral vector-9 (AAV9), to provide a functional copy of the SMN gene in SMA1 patients [32]. Encouraging clinical results showed that the drug Onasemnogene Abeparvovec (formerly known as AVXS-101) rapidly improves motor functions in severe cases [32]. In a global study, infants under 6 months of age were enrolled to evaluate this treatment (United States, NCT03306277 [33]; Europe, NCT03461289 [34]; Pacific countries, NCT03837184 [35]). A similar approach has been used in trial NCT03381729 where drug effect was evaluated on infants and children with deletion of 3 copies of SMN2 and SMN1 genes. [36].
In trial NCT04576494, adults with genetic SMA type 2 and type 3 were enrolled to study the functional effect of Nusinersen (Spinraza), an ASO designed to allow the SMN2 gene to produce the full-length and functionally normal protein. This drug has recently been approved by over 40 countries as a treatment for SMA.

4.1.2. Omics Approach for Monitoring

A transcriptomic approach has been used in trial NCT00439569 to evaluate the effect of sodium phenylbutyrate (a histone deacetylase inhibitor) on SMN-mRNA and -protein levels in children with SMA type 2 and type 3. The study was closed prematurely due to poor compliance with drug administration.

4.1.3. Multi-Omics Approach Both for Stratification and Monitoring

A genomic and transcriptomic strategy has been applied in trials NCT03032172, NCT02908685 and NCT02913482 to characterize the safety, tolerability and response of the splice modifier RO7034067 (Risdiplam) in adult, paediatric or infant SMA patients. In these trials, drug effects were assessed by evaluating changes in SMN mRNA and protein levels in the blood following RO7034067 treatment.
A repurposing drug strategy was exploited in trial NCT00485511 where the efficacy and safety of Hydroxyurea (HU) was tested in children with SMN1-SMN2 mutations. By reducing the need for blood transfusions in patients with sickle cell anaemia, this drug is used for the treatment of chronic myeloid leukaemia as well as in the prevention of painful episodes [37].

4.2. Not Drug Related Clinical Trials

A genomic approach has been applied in trial NCT02550691 to identify specific cis duplication biomarker of SMN1 in SMA families and, in trial NCT04833348, to quantify motor function in infants treated with innovative therapies (gene therapy or pharmacogenomics).

5. Spinal Bulbar Muscular Atrophy

SBMA, or Kennedy disease, is a genetic late onset MND characterized by slowly progressive weakness of the bulbar muscles and limbs with fasciculations, muscle atrophy and gynecomastia [38]. The disorder is clinically similar to, but genetically distinct from classical forms of autosomal SMA. SBMA is caused by a CAG repeat expansion in the first exon of the androgen receptor (AR) gene, on the X chromosome, encoding for a poly-glutamine (polyQ) tract. A repeat number higher than 38 is considered pathogenic. Usually it only affects men, although female carriers may have a mild form of the disease [39]. To date, no disease-modifying treatments for SBMA are available [39].
A genomic approach has been used in clinical trial NCT00303446 to evaluate the pharmacological properties of dutasteride in SBMA patients. By acting as a 5-alpha-reductase I and II inhibitor, this treatment is used against benign prostatic hyperplasia [40]. The results of this study demonstrated drug tolerability but did not show beneficial effect on muscle strength [41].

6. Conclusions

Omics sciences are quickly modifying clinical investigations of MNDs to achieve a more comprehensive view of underlying molecular pathways and allow patients stratification into diagnostic, prognostic and therapeutic subgroups. Today we are at a critical point in the development of personalized medicines for MNDs. The landscape of clinical trials using omics approaches and their surge in recent years are expected to produce an important transformation of clinical management of MNDs. The omics-based development of newly launched therapies is speeding solutions to the clinic and have a profound impact in both diagnostics and therapy. However, the road to translate these solutions to the clinic can be challenging. There is still a wide gap between discovery research and clinical practice. Bridging this gap and allowing an effective and sustainable implementation of personalized medicine will require new investments, technologies as well as the training of health professionals. The lack of these may result in inequities in the access or delivering of person-centred care.

Author Contributions

Conceptualization, F.L.C. and S.C.; methodology, S.C.; resources, P.R.; writing—original draft preparation, P.R.; writing—review and editing, P.R., F.L.C. and S.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Department of Excellence (Italian Law232/2016) the Department of Pharmacy, Health and Nutritional Sciences of University of Calabria (Italy).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Strafella, C.; Caputo, V.; Galota, M.R.; Zampatti, S.; Marella, G.; Mauriello, S.; Cascella, R.; Giardina, E. Application of precision medicine in neurodegenerative diseases. Front. Neurol. 2018, 9, 701. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Chakraborty, S.; Hosen, M.I.; Ahmed, M.; Shekhar, H.U. Onco-multi-omics approach: A new frontier in cancer research. Biomed. Res. Int. 2018, 2018, 9836256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Gibbs, R.M.; Lipnick, S.; Bateman, J.W.; Chen, L.; Cousins, H.C.; Hubbard, E.G.; Jowett, G.; LaPointe, D.S.; McGredy, M.J.; Odonkor, M.N.; et al. Toward precision medicine for neurological and neuropsychiatric disorders. Cell Stem Cell 2018, 23, 21–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Dharmadasa, T.; Matamala, J.M.; Huynh, W.; Zoing, M.C.; Kiernan, M.C. Motor neurone disease. Handb. Clin. Neurol. 2018, 159, 345–357. [Google Scholar] [CrossRef]
  5. Ungaro, C.; Sprovieri, T.; Morello, G.; Perrone, B.; Spampinato, A.G.; Simone, I.L.; Trojsi, F.; Monsurro, M.R.; Spataro, R.; La Bella, V.; et al. Genetic investigation of amyotrophic lateral sclerosis patients in south Italy: A two-decade analysis. Neurobiol. Aging 2021, 99, 99.e7–99.e14. [Google Scholar] [CrossRef]
  6. Ruffo, P.; Strafella, C.; Cascella, R.; Caputo, V.; Conforti, F.L.; Ando, S.; Giardina, E. Deregulation of ncRNA in neurodegenerative disease: Focus on circRNA, lncRNA and miRNA in amyotrophic lateral sclerosis. Front. Genet. 2021, 12, 784996. [Google Scholar] [CrossRef]
  7. Morello, G.; Guarnaccia, M.; Spampinato, A.G.; La Cognata, V.; D’Agata, V.; Cavallaro, S. Copy number variations in amyotrophic lateral sclerosis: Piecing the mosaic tiles together through a systems biology approach. Mol. Neurobiol. 2018, 55, 1299–1322. [Google Scholar] [CrossRef] [Green Version]
  8. Perrone, B.; Conforti, F.L. Common mutations of interest in the diagnosis of amyotrophic lateral sclerosis: How common are common mutations in ALS genes? Expert Rev. Mol. Diagn. 2020, 20, 703–714. [Google Scholar] [CrossRef]
  9. Miller, R.G.; Mitchell, J.D.; Moore, D.H. Riluzole for amyotrophic lateral sclerosis (ALS)/motor neuron disease (MND). Cochrane Database Syst. Rev. 2012, 3, CD001447. [Google Scholar] [CrossRef]
  10. Yoshino, H. Edaravone for the treatment of amyotrophic lateral sclerosis. Expert Rev. Neurother. 2019, 19, 185–193. [Google Scholar] [CrossRef]
  11. Ahmed, R.M.; Devenney, E.M.; Strikwerda-Brown, C.; Hodges, J.R.; Piguet, O.; Kiernan, M.C. Phenotypic variability in ALS-FTD and effect on survival. Neurology 2020, 94, e2005–e2013. [Google Scholar] [CrossRef] [PubMed]
  12. Weishaupt, J.H.; Hyman, T.; Dikic, I. Common molecular pathways in amyotrophic lateral sclerosis and frontotemporal dementia. Trends Mol. Med. 2016, 22, 769–783. [Google Scholar] [CrossRef] [PubMed]
  13. Van Mossevelde, S.; van der Zee, J.; Cruts, M.; Van Broeckhoven, C. Relationship between C9orf72 repeat size and clinical phenotype. Curr. Opin. Genet. Dev. 2017, 44, 117–124. [Google Scholar] [CrossRef] [PubMed]
  14. Morello, G.; Salomone, S.; D’Agata, V.; Conforti, F.L.; Cavallaro, S. From multi-omics approaches to precision medicine in amyotrophic lateral sclerosis. Front. Neurosci. 2020, 14, 577755. [Google Scholar] [CrossRef] [PubMed]
  15. Morello, G.; Spampinato, A.G.; Cavallaro, S. Molecular Taxonomy of sporadic amyotrophic lateral sclerosis using disease-associated genes. Front. Neurol. 2017, 8, 152. [Google Scholar] [CrossRef] [Green Version]
  16. Miller, T.M.; Pestronk, A.; David, W.; Rothstein, J.; Simpson, E.; Appel, S.H.; Andres, P.L.; Mahoney, K.; Allred, P.; Alexander, K.; 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. Lancet Neurol. 2013, 12, 435–442. [Google Scholar] [CrossRef] [Green Version]
  17. Benatar, M.; Wuu, J.; Andersen, P.M.; Atassi, N.; David, W.; Cudkowicz, M.; Schoenfeld, D. Randomized, double-blind, placebo-controlled trial of arimoclomol in rapidly progressive SOD1 ALS. Neurology 2018, 90, e565–e574. [Google Scholar] [CrossRef] [Green Version]
  18. Brooks, B.R.; Miller, R.G.; Swash, M.; Munsat, T.L. World federation of neurology research group on motor neuron, D. El Escoriall revisited: Revised criteria for the diagnosis of amyotrophic lateral sclerosis. Amyotroph Lateral Scler Other Mot. Neuron. Disord. 2000, 1, 293–299. [Google Scholar] [CrossRef]
  19. Moulignier, A.; Moulonguet, A.; Pialoux, G.; Rozenbaum, W. Reversible ALS-like disorder in HIV infection. Neurology 2001, 57, 995–1001. [Google Scholar] [CrossRef]
  20. Kindy, M.; Lupinacci, P.; Chau, R.; Shum, T.; Ko, D. A Phase 2A randomized, double-blind, placebo-controlled pilot trial of GM604 in patients with Amyotrophic Lateral Sclerosis (ALS Protocol GALS-001) and a single compassionate patient treatment (Protocol GALS-C). F1000Res 2017, 6, 230. [Google Scholar] [CrossRef] [Green Version]
  21. Fournier, C.N.; Schoenfeld, D.; Berry, J.D.; Cudkowicz, M.E.; Chan, J.; Quinn, C.; Brown, R.H.; Salameh, J.S.; Tansey, M.G.; Beers, D.R.; et al. An open label study of a novel immunosuppression intervention for the treatment of amyotrophic lateral sclerosis. Amyotroph Lateral Scler Front. Degener. 2018, 19, 242–249. [Google Scholar] [CrossRef] [PubMed]
  22. Milligan, C.; Atassi, N.; Babu, S.; Barohn, R.J.; Caress, J.B.; Cudkowicz, M.E.; Evora, A.; Hawkins, G.A.; Wosiski-Kuhn, M.; Macklin, E.A.; et al. Tocilizumab is safe and tolerable and reduces C-reactive protein concentrations in the plasma and cerebrospinal fluid of ALS patients. Muscle Nerve 2021, 64, 309–320. [Google Scholar] [CrossRef] [PubMed]
  23. Pushpakom, S.; Iorio, F.; Eyers, P.A.; Escott, K.J.; Hopper, S.; Wells, A.; Doig, A.; Guilliams, T.; Latimer, J.; McNamee, C.; et al. Drug repurposing: Progress, challenges and recommendations. Nat. Rev. Drug Discov. 2019, 18, 41–58. [Google Scholar] [CrossRef] [PubMed]
  24. Jaber, L.A.; Bailey, E.M.; Rybak, M.J. Enoxacin: A new fluoroquinolone. Clin. Pharm 1989, 8, 97–107. [Google Scholar] [PubMed]
  25. Miller, T.; Cudkowicz, M.; Shaw, P.J.; Andersen, P.M.; Atassi, N.; Bucelli, R.C.; Genge, A.; Glass, J.; Ladha, S.; Ludolph, A.L.; et al. Phase 1-2 trial of antisense oligonucleotide tofersen for SOD1 ALS. N. Engl. J. Med. 2020, 383, 109–119. [Google Scholar] [CrossRef]
  26. Lange, D.J.; Shahbazi, M.; Silani, V.; Ludolph, A.C.; Weishaupt, J.H.; Ajroud-Driss, S.; Fields, K.G.; Remanan, R.; Appel, S.H.; Morelli, C.; et al. Pyrimethamine significantly lowers cerebrospinal fluid Cu/Zn superoxide dismutase in amyotrophic lateral sclerosis patients with SOD1 mutations. Ann. Neurol. 2017, 81, 837–848. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Wohlrab, J.; Stintzing, D.; Schultz, L.; Jugelt, K.; Schroeder, O.H. Influence of janus kinase inhibitors on the neuronal activity as a proof-of-concept model for itch. Ski. Pharm. Physiol. 2022, 35, 94–101. [Google Scholar] [CrossRef]
  28. Lv, Z.; Guo, Y. Metformin and its benefits for various diseases. Front. Endocrinol. 2020, 11, 191. [Google Scholar] [CrossRef]
  29. Querin, G.; Bede, P.; El Mendili, M.M.; Li, M.; Pelegrini-Issac, M.; Rinaldi, D.; Catala, M.; Saracino, D.; Salachas, F.; Camuzat, A.; et al. Presymptomatic spinal cord pathology in c9orf72 mutation carriers: A longitudinal neuroimaging study. Ann. Neurol. 2019, 86, 158–167. [Google Scholar] [CrossRef] [Green Version]
  30. Keinath, M.C.; Prior, D.E.; Prior, T.W. Spinal muscular atrophy: Mutations, testing, and clinical relevance. Appl. Clin. Genet. 2021, 14, 11–25. [Google Scholar] [CrossRef]
  31. Chen, T.H. 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, 3297. [Google Scholar] [CrossRef]
  32. Lowes, L.P.; Alfano, L.N.; Arnold, W.D.; Shell, R.; Prior, T.W.; McColly, M.; Lehman, K.J.; Church, K.; Sproule, D.M.; Nagendran, S.; et al. Impact of age and motor function in a phase 1/2A study of infants with SMA type 1 receiving single-dose gene replacement therapy. Pediatr. Neurol. 2019, 98, 39–45. [Google Scholar] [CrossRef] [Green Version]
  33. Day, J.W.; Finkel, R.S.; Chiriboga, C.A.; Connolly, A.M.; Crawford, T.O.; Darras, B.T.; Iannaccone, S.T.; Kuntz, N.L.; Pena, L.D.M.; Shieh, P.B.; 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, 284–293. [Google Scholar] [CrossRef]
  34. Mercuri, E.; Muntoni, F.; Baranello, G.; Masson, R.; Boespflug-Tanguy, O.; Bruno, C.; Corti, S.; Daron, A.; Deconinck, N.; Servais, L.; et al. Onasemnogene abeparvovec gene therapy for symptomatic infantile-onset spinal muscular atrophy type 1 (STR1VE-EU): An open-label, single-arm, multicentre, phase 3 trial. Lancet Neurol. 2021, 20, 832–841. [Google Scholar] [CrossRef]
  35. Schwartz, M.; Likhite, S.; Meyer, K. Onasemnogene abeparvovec-xioi: A gene replacement strategy for the treatment of infants diagnosed with spinal muscular atrophy. Drugs Today (Barc) 2021, 57, 387–399. [Google Scholar] [CrossRef] [PubMed]
  36. Meyer, K.; Ferraiuolo, L.; Schmelzer, L.; Braun, L.; McGovern, V.; Likhite, S.; Michels, O.; Govoni, A.; Fitzgerald, J.; Morales, P.; et al. Improving single injection CSF delivery of AAV9-mediated gene therapy for SMA: A dose-response study in mice and nonhuman primates. Mol. Ther. 2015, 23, 477–487. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Musialek, M.W.; Rybaczek, D. Hydroxyurea—The good, the bad and the ugly. Genes 2021, 12, 1096. [Google Scholar] [CrossRef]
  38. Harding, A.E.; Thomas, P.K.; Baraitser, M.; Bradbury, P.G.; Morgan-Hughes, J.A.; Ponsford, J.R. X-linked recessive bulbospinal neuronopathy: A report of ten cases. J. Neurol. Neurosurg. Psychiatry 1982, 45, 1012–1019. [Google Scholar] [CrossRef] [Green Version]
  39. Breza, M.; Koutsis, G. Kennedy’s disease (spinal and bulbar muscular atrophy): A clinically oriented review of a rare disease. J. Neurol. 2019, 266, 565–573. [Google Scholar] [CrossRef]
  40. Dolder, C.R. Dutasteride: A dual 5-alpha reductase inhibitor for the treatment of symptomatic benign prostatic hyperplasia. Ann. Pharm. 2006, 40, 658–665. [Google Scholar] [CrossRef]
  41. Fernandez-Rhodes, L.E.; Kokkinis, A.D.; White, M.J.; Watts, C.A.; Auh, S.; Jeffries, N.O.; Shrader, J.A.; Lehky, T.J.; Li, L.; Ryder, J.E.; et al. Efficacy and safety of dutasteride in patients with spinal and bulbar muscular atrophy: A randomised placebo-controlled trial. Lancet Neurol. 2011, 10, 140–147. [Google Scholar] [CrossRef] [Green Version]
Figure 1. The evolution of interventional clinical trials in ALS and SMA using omics approaches.
Figure 1. The evolution of interventional clinical trials in ALS and SMA using omics approaches.
Jpm 12 00758 g001
Table 1. List of clinical trials carried out on ALS using different omics approaches.
Table 1. List of clinical trials carried out on ALS using different omics approaches.
Amyotrophic Lateral Sclerosis
Clinical Trials IdentifierDatePhaseStatusTreatmentApproachTitleStatePhase
StartCompletion NameDrug
NCT01041222January 2010January 2012CompletedfALS diagnosis: SOD1 gene mutation carriesISIS 333611ASO designed to inhibit SOD1 expressionGenomicsA Phase 1, Double-Blind, Placebo-Controlled, Dose-Escalation Study of the Safety, Tolerability, and Pharmacokinetics of ISIS 333,611 Administered Intrathecally to Patients with Familial Amyotrophic Lateral Sclerosis Due to Superoxide Dismutase 1 Gene MutationsUnited StatesI
NCT00706147January 2009December 2014CompletedSOD1 genetic mutationArimoclomolHSP response inductorGenomicsPhase II/III Randomized, Placebo-Controlled Trial of Arimoclomol in SOD1 Positive Familial Amyotrophic Lateral Sclerosis (ALS).United StatesII-III
NCT0449425628 September 2020 RecruitingGenetic diagnosis (SOD1, FUS, ATXN2)BIIB105ASO is designed to bind ATXN2 mRNA and mediate its degradationGenomicsA Phase 1 Multiple-Ascending-Dose Study to Assess the Safety, Tolerability, and Pharmacokinetics of BIIB105 Administered Intrathecally to Adults with Amyotrophic Lateral Sclerosis with or without Poly-CAG Expansion in the Ataxin-2 Gene.United StatesI
NCT046322259 February 2021 Active, not recruitingALS diagnosis according to the El Escorial CriteriaEngensisGene therapy using plasmid to deliver the HGF gene directly to nerve cellsTranscriptomicsA Phase 2a, Double-Blind, Randomized, Placebo-Controlled, Multicenter Study to Assess the Safety of Engensis in Participants with Amyotrophic Lateral SclerosisUnited StatesII
NCT0335953819 September 2017 Active, not recruitingALS diagnosis according to the El Escorial CriteriaRapamycin—SirolimusImmunomodulatory effects and improves protein degradationTranscriptomicsRapamycin (Sirolimus) Treatment for Amyotrophic Lateral SclerosisItalyII
NCT0087544613 May 20099 September 2011CompletedALS diagnosis according to the Gold Coast
Criteria
GSK1223249-
Ozanezumab
Monoclonal antibody targeting NOGO-A proteinTranscriptomics and metabolomicsA Single and Repeat Dose Escalation Study of the Safety, Pharmacokinetics and Pharmacodynamics of GSK1223249 in ALS PatientsUnited StatesI
NCT0345688218 November 201623 November 2020CompletedALS diagnosis according to the El Escorial CriteriaRNS60Saline solution with charged oxygenated nanobubblesMetabolomics and transcriptomicsThe Effect of RNS60 on ALS Biomarkers.ItalyII
NCT01854294August 2013April 2014CompletedALS diagnosis according to the El Escorial CriteriaGM604PeptideTranscriptomicsGM604 Phase 2A Randomized Double-blind Placebo Controlled Pilot Trial in Amyotrophic Lateral Disease (ALS)United StatesII
NCT0380052422 February 2019 RecruitingALS diagnosis according to the El Escorial CriteriaTauroursodeoxycholicAntiapoptotic and ER stress response damping effectsTranscriptomicsSafety and Efficacy of Tauroursodeoxycholic (TUDCA) as add-on Treatment in Patients Affected by Amyotrophic Lateral Sclerosis (ALS)Belgium, France and othersIII
NCT0450535830 December 2021 Not yet recruitingALS diagnosis according to the El Escorial CriteriaPU-AD—
Icapamespib
Brain permeable Hsp90 protein inhibitorGenomics and transcriptomeA Randomized, Double-blind, Placebo-controlled Pilot Study to Evaluate the Biological Activity, Safety, and Pharmacokinetics of PU-AD in Subjects with Amyotrophic Lateral Sclerosis (ALS) II
NCT0369378110 April 2019 Active, not recruitingALS diagnosis according to the El Escorial CriteriaColchicine TranscriptomicsColchicine for Amyotrophic Lateral Sclerosis: A Phase II, Randomized, Double Blind, Placebo Controlled, Multicenter Clinical TrialItalyII
NCT01884571October 2013January 2016CompletedALS diagnosis according to the El Escorial CriteriaBasiliximab, Methylprednisolone, Prednisone, Tacrolimus, Mycophenolate mofetilImmunosuppression treatmentTranscriptomicsA Novel Immunosuppression Intervention for the Treatment of Amyotrophic Lateral Sclerosis (ALS)United StatesII
NCT02525471October 201521 June 2017CompletedALS diagnosis according to the El Escorial CriteriaRNS60Saline solution with charged oxygenated nanobubblesTranscriptomicsA Pilot Study of RNS60 in Amyotrophic Lateral Sclerosis (ALS)United StatesI
NCT02469896November 201511 July 2018CompletedALS diagnosis according to the El Escorial CriteriaTocilizumabASO designed to inhibit interleukin 6
(IL-6)
TranscriptomicsA Phase 2 Randomized, Placebo Controlled Trial of Tocilizumab in ALS SubjectsUnited StatesII
NCT0519399418 January 2022 Not yet recruitingALS diagnosis according to the Gold Coast CriteriaTriumeqCombined treatment of: dolutegravir, abacavir, lamivudineGenomics and transcriptomicsRandomised Double-Blind Placebo-Controlled Phase 3 Trial of Triumeq in Amyotrophic Lateral SclerosisAustraliaIII
NCT0478874529 June 2021 RecruitingALS diagnosis according to the El Escorial CriteriaTrimetazidine Dihydrochloride TranscriptomicsTargeting Metabolic Flexibility in ALS (MetFlex); Safety and Tolerability of Trimetazidine for the Treatment of ALSAustralia, United Kindom and othersII
NCT0484082326 March 2021 RecruitingALS diagnosis according to the El Escorial CriteriaEnoxacinQuinolone/fluoroquinolone antibioticTranscriptomicsA Randomized, Double-blind, Parallel Group, Single Centre, Phase 1b/2 Study to Assess the Safety, Tolerability, Pharmacokinetics and Pharmacodynamics of Three Orally Administered Doses of Enoxacin (200 mg Twice Daily, 400 mg Twice Daily and 600 mg Twice Daily) in Adults with Amyotrophic Lateral SclerosisCanadaI-II
NCT024371101 April 2019 RecruitingALS diagnosis according to the El Escorial CriteriaDarunavir, Ritonavir, Dolutegravir, Tenofovir alafenamide (TAF)Darunavir and Ritonavir = protease inhibitor; Dolutegravir = integrase inhibitor; TAF = nucleoside reverse transcriptase inhibitorTranscriptomicsHERV-K Suppression Using Antiretroviral Therapy in Volunteers with Amyotrophic Lateral Sclerosis (ALS)United StatesI
NCT0406624430 December 2019 RecruitingALS diagnosis according to the El Escorial CriteriaBLZ945CSF-1 InhibitorGenomics Metabolomics and transcriptomicsAn Open-label, Adaptive Design Study in Patients with Amyotrophic Lateral Sclerosis (ALS) to Characterize Safety, Tolerability and Brain Microglia Response, as Measured by TSPO Binding, Following Multiple Doses of BLZ945 Using Positron Emission Tomography (PET) With the Radioligand [11C]-PBR28United StatesII
NCT0362601210 September 201817 November 2021CompletedC9Orf72 genetic
mutation
BIIB078ASO designed to target C9Orf72 mRNAGenomics and metabolomicsA Phase 1 Multiple-Ascending-Dose Study to Assess the Safety, Tolerability, and Pharmacokinetics of BIIB078 Administered Intrathecally to Adults with C9ORF72-Associated Amyotrophic Lateral SclerosisUnited States, Belgium and othersI
NCT0428885628 April 2020 Active, not recruitingC9Orf72 genetic
mutation
BIIB078ASO designed to target C9Orf72 mRNAGenomics and metabolomicsAn Extension Study to Assess the Long-Term Safety, Tolerability, Pharmacokinetics, and Effect on Disease Progression of BIIB078 Administered to Previously Treated Adults with C9ORF72-Associated Amyotrophic Lateral SclerosisUnited StatesI
NCT0516388623 December 2021 RecruitingC9Orf72 genetic
mutation
LAM-002APIKfyve kinase inhibitor that activates the transcription factor EB (TFEB)Genomics and transcriptomeA Phase IIa Trial to Evaluate the Safety, Tolerability, and Biological Activity of LAM-002A (Apilimod Dimesylate Capsules) in C9ORF72-Associated Amyotrophic Lateral SclerosisUnited StatesII
NCT050530352 September 2021 RecruitingC9Orf72 genetic
mutation
LAM-002APIKfyve kinase inhibitor that activates the transcription factor EB (TFEB)Genomics and transcriptomeA Phase 2, Multicenter, Randomized, Double-Blind, Placebo-Controlled Study to Evaluate the Safety, Tolerability, Pharmacokinetics, and Pharmacodynamics of AL001 in C9orf72-Associated Amyotrophic Lateral SclerosisUnited StatesII
NCT049937551 October 2021 RecruitingC9Orf72
genetic mutation
TPN-101,
censavudine
Inhibitor of the reverse transcriptase enzymeGenomics and metabolomicsA Phase 2a Study of TPN-101 in Patients with Amyotrophic Lateral Sclerosis (ALS) and/or Frontotemporal Dementia (FTD) Associated with Hexanucleotide Repeat Expansion in the C9Orf72 Gene (C9ORF72 ALS/FTD)United StatesII
NCT0493186228 June 2021 RecruitingC9Orf72 genetic
mutation
WVE-004ASO is designed to mediate the degradation of C9ORF72 mRNAsGenomicsA Multicenter, Randomized, Double-blind, Placebo-controlled, Phase 1b/2a Study of WVE-004 Administered Intrathecally to Patients with C9orf72-associated Amyotrophic Lateral Sclerosis (ALS) or Frontotemporal Dementia (FTD)AustraliaI-II
NCT0262369920 January 201616 July 2021CompletedSOD1 genetic mutationBIIB067—TofersenASO designed to degrade SOD1 mRNA to prevent protein synthesis and reduce levels of harmful proteins.Genomics and metabolomicsA Study to Evaluate the Efficacy, Safety, Tolerability, Pharmacokinetics, and Pharmacodynamics of BIIB067 Administered to Adult Subjects with Amyotrophic Lateral Sclerosis and Confirmed Superoxide Dismutase 1 MutationUnited StatesIII
NCT030701198 March 2017 Active, not recruitingSOD1 genetic mutationBIIB067—TofersenASO designed to degrade SOD1 mRNA to prevent protein synthesis and reduce levels of harmful proteins.GenomicsAn Extension Study to Assess the Long-Term Safety, Tolerability, Pharmacokinetics, and Effect on Disease Progression of BIIB067 Administered to Previously Treated Adults with Amyotrophic Lateral Sclerosis Caused by Superoxide Dismutase 1 MutationUnited StatesIII
NCT0485698217 May 2021 RecruitingSOD1 genetic mutationBIIB067—TofersenASO designed to degrade SOD1 mRNA to prevent protein synthesis and reduce levels of harmful proteins.Genomics and metabolomicsA Phase 3 Randomized, Placebo-Controlled Trial with a Longitudinal Natural History Run-In and Open-Label Extension to Evaluate BIIB067 Initiated in Clinically Presymptomatic Adults with a Confirmed Superoxide Dismutase 1 MutationUnited StatesIII
NCT01083667November 2009December 2014CompletedSOD1 genetic mutationPyrimethamine –
Daraprim
Genomics and metabolomicsPhase I/II Study of SOD1 Inhibition by Pyrimethamine in Familial ALSUnited States, Germany and othersI-II
NCT0476897214 June 2021 RecruitingFUS genetic
mutation
ION363—JaciFUS enASO designed to reduce the production of a mutated neurotoxic form of the FUS proteinGenomics and metabolomicsA Phase 1–3 Study to Evaluate the Efficacy, Safety, Pharmacokinetics and Pharmacodynamics of Intrathecally Administered ION363 in Amyotrophic Lateral Sclerosis Patients with FUS ed in Sarcoma Mutations (FUS -ALS)United StatesIII
NCT0370779521 August 201710 January 2019CompletedFUS genetic
mutation and
fALS diagnosis
BetamethasoneCorticosteroid, reducing inflammation and changing the body’s immune responseGenomics and metabolomicsTreatment of FUS -Related ALS With Betamethasone—The TRANSLATE StudyUnited StatesEarly I
NCT051891061 February 2022 Not yet
recruiting
ALS diagnosis according to the El Escorial CriteriaBaricitinib—
Olumiant
Immunosuppressant—JAK inhibitorsGenomics,
proteomics and metabolomics
Neurodegenerative Alzheimer’s Disease and Amyotrophic Lateral Sclerosis (NADALS) Basket Proof of Concept Trial Including Asymptomatic Individuals Using BaricitinibUnited StatesI-II
NCT0422002110 January 2020 RecruitingC9Orf72 genetic
mutation
Metformin Genomics and metabolomicsA Single-Center, Open Label Study to Assess the Safety and Tolerability of Metformin in Subjects with C9orf72 Amyotrophic Lateral Sclerosis Over 24 Weeks of TreatmentUnited StatesII
NCT025902768 October 201527 October 2020CompletedC9Orf72 genetic
mutation
Genomics, Metabolomics and transcriptomicsPredict to Prevent Frontotemporal Lobar Degeneration and Amyotrophic Lateral SclerosisFranceNot Applicable
NCT0398470827 January 2020 RecruitingALS diagnosis according to the El Escorial Criteria Metabolomics, lipidpomics and transcriptomicsNew Therapeutic Strategy in ALS Based on Metabolic Status and Associated Metabolic Pathways.FranceNot Applicable
NCT01984957January 2013January 2015CompletedALS diagnosis according to the El Escorial Criteria TranscriptomicsDifferential Study of Muscle Transcriptome in Patients with Neuromuscular Disease and Control SubjectsFranceNot Applicable
NCT0267022629 March 20169 December 2019CompletedALS diagnosis according to the El Escorial Criteria Metabolomics and transcriptomicsMetabolomics and Transcriptomics Approaches to Identify Muscular Biomarkers in Amyotrophic Lateral SclerosisFranceNot Applicable
NCT0385130228 October 2019 RecruitingALS diagnosis according to the El Escorial Criteria TranscriptomicsEffects of Remote Ischemic Conditioning on Hand Use in Individuals with Spinal Cord Injury and Amyotrophic Lateral Sclerosis: A Preliminary StudyUnited StatesNot Applicable
NCT036189661 November 20141 May 2016CompletedALS diagnosis according to the El Escorial Criteria TranscriptomicsNeuromuscular Magnetic Stimulation Counteracts Muscle Decline in ALS Patients II
NCT0336765013 May 2014 RecruitingALS diagnosis according to the El Escorial Criteria GenomicsEpidemiology and Genetics of the Amyotrophic Lateral Sclerosis in the French West IndiesFranceNot Applicable
NCT0357346610 April 2019 Active, not recruitingSOD1- C9Orf72
genetic mutation
GenomicsPresymptomatic Neuromuscular Junction Defects and Compensatory Mechanisms in Amyotrophic Lateral Sclerosis (ALS)FranceNot Applicable
The search for clinical trials in the ClinicalTrials.gov database was carried out using “Amyotrophic Lateral Sclerosis” and “mutation”, “mutational”, “gene expression”, “genotype”, “gene”, “transcriptome”, “transcriptomics”, “C9Orf72”, “SOD1”, “FUS”, “TARDBP”, “DNA”, “RNA”, “sequencing” as keywords. The names of genes are in italics.
Table 2. List of clinical trials carried out on SMA and SBMA using different omics approaches.
Table 2. List of clinical trials carried out on SMA and SBMA using different omics approaches.
Spinal Muscular Atrophy and Spinal-Bulbar Muscular Atrophy
Clinical Trials IdentifierDatePhaseStatusTreatmentApproachTitleStatePhase
StartCompletion NameDrug
NCT021229525 May 201415 December 2017CompletedSMN1SMN2
genetic diagnosis
AVXS-101—Onasemnogene AbeparvovecSMN gene
therapy
GenomicsPhase I Gene Transfer Clinical Trial for Spinal Muscular Atrophy Type 1 Delivering AVXS-101United StatesI
NCT0330627724 October 201712 November 2019CompletedSMN1SMN2
genetic diagnosis
AVXS-101—Onasemnogene AbeparvovecSMN gene
therapy
GenomicsPhase 3, Open-Label, Single-Arm, Single-Dose Gene Replacement Therapy Clinical Trial for Patients with Spinal Muscular Atrophy Type 1 With One or Two SMN2 Copies Delivering AVXS-101 by Intravenous InFUS ionUnited StatesIII
NCT0346128916 August 201811 September 2020CompletedSMN1SMN2
genetic diagnosis
AVXS-101—Onasemnogene AbeparvovecSMN gene
therapy
GenomicsPhase 3, Open-Label, Single-Arm, Single-Dose Gene Replacement Therapy Clinical Trial for Patients with Spinal Muscular Atrophy Type 1 With One or Two SMN2 Copies Delivering AVXS-101 by Intravenous InFUS ionBelgium, France and othersIII
NCT0383718431 May 201929 June 2021CompletedSMN1SMN2
genetic diagnosis
AVXS-101—Onasemnogene AbeparvovecSMN gene
therapy
GenomicsPhase 3, Open-Label, Single-Arm, Single-Dose Gene Replacement Therapy Clinical Trial for Patients with Spinal Muscular Atrophy Type 1 With One or Two SMN2 Copies Delivering AVXS-101 by Intravenous InFUS ionJapan, Korea and TaiwanIII
NCT0338172914 December 201718 November 2021CompletedSMN2 genetic
diagnosis
AVXS-101—Onasemnogene AbeparvovecSMN gene
therapy
GenomicsPhase I, Open-Label, Dose Comparison Study of AVXS-101 for Sitting but Non-ambulatory Patients with Spinal Muscular AtrophyUnited StatesI
NCT035050992 April 201815 June 2021CompletedSMN2 genetic
diagnosis
AVXS-101—Onasemnogene AbeparvovecSMN gene
therapy
GenomicsA Global Study of a Single, One-Time Dose of AVXS-101 Delivered to Infants with Genetically Diagnosed and Pre-symptomatic Spinal Muscular Atrophy with Multiple Copies of SMN2United StatesIII
NCT0404202510 February 2020 Enrolling by invitationSMA clinical and genetic diagnosisAVXS-101—Onasemnogene AbeparvovecSMN gene
therapy
GenomicsA Long-term Follow-up Study of Patients in the Clinical Trials for Spinal Muscular Atrophy Receiving AVXS-101United StatesIV
NCT0262874320 January 201618 December 2018CompletedSMN2 genetic
diagnosis
AVXS-101—Onasemnogene AbeparvovecSMN gene
therapy
GenomicsMulticenter, Open-Label, Single-Arm Study to Evaluate Long-Term Safety, Tolerability, and Effectiveness of 10 mg/kg BID Olesoxime in Patients with Spinal Muscular AtrophyBelgium, France and others.II
NCT0457649424
January 2022
RecruitingSMA genetic
diagnosis
Nusinersen—SpinrazaASO designed to allow the SMN2 gene to produce the full-length protein that can function normallyGenomicsStudy of the Functional Effects of Nusinersen in 5q-spinal Muscular Amyotrophy Adults (SMA Type 2 or 3 Forms): a Multicenter Single-case Experimental Design in Multiple Baselines Across Subjects, Randomized, Single-blinded EvaluationFranceNot Applicable
NCT048518738
September 2021
RecruitingSMN1SMN2
genetic diagnosis
OAV101 (AVXS-101)SMN gene
therapy
GenomicsA Phase IIIb, Open-label, Single-arm, Single-dose, Multicenter Study to Evaluate the Safety, Tolerability and Efficacy of Gene Replacement Therapy with Intravenous OAV101 (AVXS-101) in Pediatric Patients With Spinal Muscular Atrophy (SMA)AustraliaIII
NCT050896562 February 2022 RecruitingSMN1SMN2
genetic diagnosis
OAV101 (AVXS-101)SMN gene
therapy
GenomicsA Randomized, Sham-controlled, Double-blind Study to Evaluate the Efficacy and Safety of Intrathecal OAV101 in Patients Type 2 Spinal Muscular Atrophy (SMA) Who Are ≥ 2 to < 18 Years of Age, Treatment Naive, Sitting, and Never AmbulatoryUnited StatesIII
NCT037793347
August 2019
Active, not recruitingSMN2 genetic
diagnosis
RO7034067- RisdiplamA splice modifier of the pre-mRNA of the SMN2 geneGenomicsAn Open-Label Study of Risdiplam in Infants with Genetically Diagnosed and Presymptomatic Spinal Muscular AtrophyUnited StatesII
NCT01671384August 2013 UnknownSMN1 genetic
diagnosis
Valproate and levocarnitineValproic acid (VPA) = a histone deacetylase inhibitor (HDAC)GenomicsRandomized Placebo Controlled Trial of Valproate and Levocarnitine in Children with Spinal Muscular Atrophy Aged 2–15 YearsIndiaIII
NCT00439569January 2008August 2008TerminatedSMA clinical
diagnosis
Sodium phenylbutyrateHistone deacetylase inhibitor and a chemical chaperoneTranscriptomicsPhase I/IIa Clinical Trial of Sodium Phenylbutyrate in Pediatric Subjects with Type II/III Spinal Muscular AtrophyUnited StatesI-II
NCT030321723 March 2017 Active, not recruitingSMN2 genetic
diagnosis
RO7034067- RisdiplamA splice modifier of the pre-mRNA of the SMN2 geneTranscriptomicsAn Open-Label Study to Investigate the Safety, Tolerability, and Pharmacokinetics/Pharmacodynamics of Risdiplam (RO7034067) in Adult and Pediatric Patients with Spinal Muscular AtrophyUnited StatesII
NCT0290868520
October 2016
Active, not recruitingSMA genetic
diagnosis
RO7034067- RisdiplamA splice modifier of the pre-mRNA of the SMN2 geneGenomics and transcriptomicsA Two Part Seamless, Multi-Center Randomized, Placebo-Controlled, Double-Blind Study to Investigate the Safety, Tolerability, Pharmacokinetics, Pharmacodynamics and Efficacy of Risdiplam (RO7034067) in Type 2 and 3 Spinal Muscular Atrophy PatientsUnited StatesII-III
NCT0291348223 December 2016 Active, not recruitingSMA genetic
diagnosis
RO7034067- RisdiplamA splice modifier of the pre-mRNA of the SMN2 geneGenomics and transcriptomicsA Two Part Seamless, Open-label, Multicenter Study to Investigate the Safety, Tolerability, Pharmacokinetics, Pharmacodynamics and Efficacy of Risdiplam (RO7034067) in Infants with Type 1 Spinal Muscular AtrophyUnited StatesII-III
NCT00485511June 2007June 2009CompletedSMN1SMN2
genetic diagnosis
HydroxyureaSMN2 transcription pattern
modified
Genomics and transcriptomicsA Randomized, Double-Blind, Placebo-Controlled Trial of Hydroxyurea in Spinal Muscular AtrophyTaiwanII-III
NCT0255069115 December 20154 July 2016TerminatedSMN1SMN2
genetic diagnosis
GenomicsIdentification of a Biomarker Associated with Cis-duplication of the SMN1 Gene Aiming at Improving the Genetic Counseling in Spinal Muscular Atrophy FamiliesFranceNot Applicable
NCT04833348August 2021 Not yet recruitingSMN1 genetic
diagnosis
GenomicsQuantification of Motor Function in Infants with Spinal Muscular Atrophy Treated with Innovative Therapies, IMUSMA ProjectFranceNot Applicable
NCT00303446March 2006December 2009Completed
SBMA genetically confirmed
DutasterideInhibitor of 5-alpha-reductase I and IIGenomicsDutasteride to Treat Spinal and Bulbar Muscular Atrophy (SBMA)
United StatesII
The search for clinical trials in the ClinicalTrials.gov database was carried out using the “Spinal Muscular Atrophy” or “Spinal-Bulbar Muscular Atrophy” and “mutation”, “mutational”, “gene expression”, “genotype”, “gene”, “transcriptome”, “transcriptomics”, “C9Orf72”, “SOD1”, “FUS”, “TARDBP”, “DNA”, “RNA”, “sequencing” as keywords. The names of genes are in italics.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ruffo, P.; Cavallaro, S.; Conforti, F.L. The Advent of Omics Sciences in Clinical Trials of Motor Neuron Diseases. J. Pers. Med. 2022, 12, 758. https://doi.org/10.3390/jpm12050758

AMA Style

Ruffo P, Cavallaro S, Conforti FL. The Advent of Omics Sciences in Clinical Trials of Motor Neuron Diseases. Journal of Personalized Medicine. 2022; 12(5):758. https://doi.org/10.3390/jpm12050758

Chicago/Turabian Style

Ruffo, Paola, Sebastiano Cavallaro, and Francesca Luisa Conforti. 2022. "The Advent of Omics Sciences in Clinical Trials of Motor Neuron Diseases" Journal of Personalized Medicine 12, no. 5: 758. https://doi.org/10.3390/jpm12050758

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop