1
|
Shefner JM, Cudkowicz ME, Genge A, Hardiman O, Al-Chalabi A, Andrews JA, Chio A, Corcia P, Couratier P, de Carvalho M, Heiman-Patterson T, Henderson RD, Ingre C, Johnston W, Ludolph A, Maragakis NJ, Miller TM, Mora JS, Petri S, Simmons Z, van den Berg LH, Zinman L, Kupfer S, Malik FI, Meng L, Simkins TJ, Wei J, Wolff AA, Rudnicki SA. Reldesemtiv in Amyotrophic Lateral Sclerosis: Results From the COURAGE-ALS Randomized Clinical Trial. JAMA Neurol 2025:2831901. [PMID: 40126464 PMCID: PMC11933997 DOI: 10.1001/jamaneurol.2025.0241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 11/09/2024] [Indexed: 03/25/2025]
Abstract
Importance Treatment options for amyotrophic lateral sclerosis (ALS) remain suboptimal. Results from a phase 2 study of reldesemtiv in ALS suggested that it may slow disease progression. Objective To assess the effect of reldesemtiv vs placebo on functional outcomes in ALS. Design, Setting, and Participants A Study to Evaluate the Efficacy and Safety of Reldesemtiv in Patients With Amyotrophic Lateral Sclerosis (COURAGE-ALS) was a double-blind, placebo-controlled phase 3 randomized clinical trial conducted at 83 ALS centers in 16 countries from August 2021 to July 2023. The first 24-week period was placebo controlled vs reldesemtiv. All participants received reldesemtiv during the second 24-week period with a 4-week follow-up. Two interim analyses were planned, the first for futility and the second for futility and possible resizing. This was a hybrid decentralized trial with approximately half the trial visits performed remotely and the remaining visits in the clinic. Eligible participants met criteria for definite, probable, or possible ALS with lower motor neuron signs by modified El Escorial Criteria, ALS symptoms for 24 months or less, ALS Functional Rating Scale-Revised (ALSFRS-R) total score of 44 or less, and forced vital capacity of greater than or equal to 65% of predicted. Interventions Oral reldesemtiv, 300 mg, or placebo twice daily. Main Outcomes and Measures The primary end point was change in ALSFRS-R total score from baseline to week 24. Results Of the 696 participants screened, 207 were screen failures. A total of 486 participants (mean [SD] age, 59.4 [10.9] years; 309 male [63.6%]) were randomized to reldesemtiv (n = 325) or placebo (n = 161); 3 randomized patients were not dosed. The second interim analysis at 24 weeks after randomization included 256 participants. The data monitoring committee recommended that the trial should end due to futility, and the sponsor agreed. The mean (SE) group difference in the ALSFRS-R score from baseline to week 24 was -1.1 (0.53; 95% CI, -2.17 to -0.08; P = .04, favoring placebo). Given excess missing data from early termination, the combined assessment assumed greater importance; it, too, failed to show a benefit from treatment with reldesemtiv (win probability was 0.44 for reldesemtiv and 0.49 for placebo, with a win ratio of 0.91; 95% CI of win ratio, 0.77-1.10; P = .11). Conclusions and Relevance This randomized clinical trial failed to demonstrate efficacy for reldesemtiv in slowing functional decline in ALS. Trial Registration ClinicalTrials.gov Identifier: NCT04944784.
Collapse
Affiliation(s)
- Jeremy M. Shefner
- Department of Neurology, Barrow Neurological Institute, University of Arizona, Phoenix
- Creighton University, Phoenix, Arizona
| | | | - Angela Genge
- Montreal Neurological Institute, Montréal, Québec, Canada
| | | | | | - Jinsy A. Andrews
- The Neurological Institute of New York, Columbia University Irving Medical Center, New York
| | - Adriano Chio
- Department of Neuroscience, University of Turin, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, Turin, Italy
| | | | | | - Mamede de Carvalho
- Faculty of Medicine-University of Lisbon, Hospital de Santa Maria-CHULN, Lisbon, Portugal
| | - Terry Heiman-Patterson
- Neurology Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | | | | | | | - Albert Ludolph
- University of, Ulm, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen e. V. (DZNE), Ulm, Germany
| | | | | | | | | | - Zachary Simmons
- Penn State Health Milton S. Hershey Medical Center, Hershey, Pennsylvania
| | | | - Lorne Zinman
- ALS/Neuromuscular Clinic, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Stuart Kupfer
- Cytokinetics Incorporated, South San Francisco, California
| | - Fady I. Malik
- Cytokinetics Incorporated, South San Francisco, California
| | - Lisa Meng
- Cytokinetics Incorporated, South San Francisco, California
| | | | - Jenny Wei
- Cytokinetics Incorporated, South San Francisco, California
| | | | | |
Collapse
|
2
|
Bogomolova AP, Katrukha IA. Troponins and Skeletal Muscle Pathologies. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:2083-2106. [PMID: 39865025 DOI: 10.1134/s0006297924120010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 11/19/2024] [Accepted: 12/01/2024] [Indexed: 01/28/2025]
Abstract
Skeletal muscles account for ~30-40% of the total weight of human body and are responsible for its most important functions, including movement, respiration, thermogenesis, and glucose and protein metabolism. Skeletal muscle damage negatively impacts the whole-body functioning, leading to deterioration of the quality of life and, in severe cases, death. Therefore, timely diagnosis and therapy for skeletal muscle dysfunction are important goals of modern medicine. In this review, we focused on the skeletal troponins that are proteins in the thin filaments of muscle fibers. Skeletal troponins play a key role in regulation of muscle contraction. Biochemical properties of these proteins and their use as biomarkers of skeletal muscle damage are described in this review. One of the most convenient and sensitive methods of protein biomarker measurement in biological liquids is immunochemical analysis; hence, we examined the factors that influence immunochemical detection of skeletal troponins and should be taken into account when developing diagnostic test systems. Also, we reviewed the available data on the skeletal troponin mutations that are considered to be associated with pathologies leading to the development of diseases and discussed utilization of troponins as drug targets for treatment of the skeletal muscle disorders.
Collapse
Affiliation(s)
- Agnessa P Bogomolova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia.
- Hytest Ltd., Turku, Finland
| | - Ivan A Katrukha
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
- Hytest Ltd., Turku, Finland
| |
Collapse
|
3
|
Giannakou M, Akrani I, Tsoka A, Myrianthopoulos V, Mikros E, Vorgias C, Hatzinikolaou DG. Discovery of Novel Inhibitors against ALS-Related SOD1(A4V) Aggregation through the Screening of a Chemical Library Using Differential Scanning Fluorimetry (DSF). Pharmaceuticals (Basel) 2024; 17:1286. [PMID: 39458929 PMCID: PMC11510448 DOI: 10.3390/ph17101286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 07/26/2024] [Accepted: 08/01/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Cu/Zn Superoxide Dismutase 1 (SOD1) is a 32 kDa cytosolic dimeric metalloenzyme that neutralizes superoxide anions into oxygen and hydrogen peroxide. Mutations in SOD1 are associated with ALS, a disease causing motor neuron atrophy and subsequent mortality. These mutations exert their harmful effects through a gain of function mechanism, rather than a loss of function. Despite extensive research, the mechanism causing selective motor neuron death still remains unclear. A defining feature of ALS pathogenesis is protein misfolding and aggregation, evidenced by ubiquitinated protein inclusions containing SOD1 in affected motor neurons. This work aims to identify compounds countering SOD1(A4V) misfolding and aggregation, which could potentially aid in ALS treatment. METHODS The approach employed was in vitro screening of a library comprising 1280 pharmacologically active compounds (LOPAC®) in the context of drug repurposing. Using differential scanning fluorimetry (DSF), these compounds were tested for their impact on SOD1(A4V) thermal stability. RESULTS AND CONCLUSIONS Dimer stability was the parameter chosen as the criterion for screening, since the dissociation of the native SOD1 dimer is the step prior to its in vitro aggregation. The screening revealed one compound raising protein-ligand Tm by 6 °C, eleven inducing a higher second Tm, suggesting a stabilization effect, and fourteen reducing Tm from 10 up to 26 °C, suggesting possible interactions or non-specific binding.
Collapse
Affiliation(s)
- Maria Giannakou
- Biochemistry and Molecular Biology Unit, Department of Biology, National and Kapodistrian University of Athens, 15784 Zografou, Greece
- Enzyme and Microbial Biotechnology Unit, Department of Biology, National and Kapodistrian University of Athens, 15784 Zografou, Greece
| | - Ifigeneia Akrani
- Laboratory of Pharmaceutical Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, 15784 Zografou, Greece; (I.A.)
| | - Angeliki Tsoka
- Biochemistry and Molecular Biology Unit, Department of Biology, National and Kapodistrian University of Athens, 15784 Zografou, Greece
| | - Vassilios Myrianthopoulos
- Laboratory of Pharmaceutical Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, 15784 Zografou, Greece; (I.A.)
| | - Emmanuel Mikros
- Laboratory of Pharmaceutical Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, 15784 Zografou, Greece; (I.A.)
| | - Constantinos Vorgias
- Biochemistry and Molecular Biology Unit, Department of Biology, National and Kapodistrian University of Athens, 15784 Zografou, Greece
| | - Dimitris G. Hatzinikolaou
- Enzyme and Microbial Biotechnology Unit, Department of Biology, National and Kapodistrian University of Athens, 15784 Zografou, Greece
| |
Collapse
|
4
|
Niazi SK. Bioavailability as Proof to Authorize the Clinical Testing of Neurodegenerative Drugs-Protocols and Advice for the FDA to Meet the ALS Act Vision. Int J Mol Sci 2024; 25:10211. [PMID: 39337696 PMCID: PMC11432374 DOI: 10.3390/ijms251810211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
Although decades of intensive drug discovery efforts to treat neurodegenerative disorders (NDs) have failed, around half a million patients in more than 2000 studies continue being tested, costing over USD 100 billion, despite the conclusion that even those drugs which have been approved have no better effect than a placebo. The US Food and Drug Administration (FDA) has established multiple programs to innovate the treatment of rare diseases, particularly NDs, providing millions of USD in funding primarily by encouraging novel clinical trials to account for issues related to study sizes and adopting multi-arm studies to account for patient dropouts. Instead, the FDA should focus on the primary reason for failure: the poor bioavailability of drugs reaching the brain (generally 0.1% at most) due to the blood-brain barrier (BBB). There are several solutions to enhance entry into the brain, and the FDA must require proof of significant entry into the brain as the prerequisite to approving Investigational New Drug (IND) applications. The FDA should also rely on factors other than biomarkers to confirm efficacy, as these are rarely relevant to clinical use. This study summarizes how the drugs used to treat NDs can be made effective and how the FDA should change its guidelines for IND approval of these drugs.
Collapse
Affiliation(s)
- Sarfaraz K Niazi
- College of Pharmacy, University of Illinois, Chicago, IL 60612, USA
| |
Collapse
|
5
|
Rizea RE, Corlatescu AD, Costin HP, Dumitru A, Ciurea AV. Understanding Amyotrophic Lateral Sclerosis: Pathophysiology, Diagnosis, and Therapeutic Advances. Int J Mol Sci 2024; 25:9966. [PMID: 39337454 PMCID: PMC11432652 DOI: 10.3390/ijms25189966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
This review offers an in-depth examination of amyotrophic lateral sclerosis (ALS), addressing its epidemiology, pathophysiology, clinical presentation, diagnostic techniques, and current as well as emerging treatments. The purpose is to condense key findings and illustrate the complexity of ALS, which is shaped by both genetic and environmental influences. We reviewed the literature to discuss recent advancements in understanding molecular mechanisms such as protein misfolding, mitochondrial dysfunction, oxidative stress, and axonal transport defects, which are critical for identifying potential therapeutic targets. Significant progress has been made in refining diagnostic criteria and identifying biomarkers, leading to earlier and more precise diagnoses. Although current drug treatments provide some benefits, there is a clear need for more effective therapies. Emerging treatments, such as gene therapy and stem cell therapy, show potential in modifying disease progression and improving the quality of life for ALS patients. The review emphasizes the importance of continued research to address challenges such as disease variability and the limited effectiveness of existing treatments. Future research should concentrate on further exploring the molecular foundations of ALS and developing new therapeutic approaches. The implications for clinical practice include ensuring the accessibility of new treatments and that healthcare systems are equipped to support ongoing research and patient care.
Collapse
Affiliation(s)
- Radu Eugen Rizea
- Department of Neurosurgery, University of Medicine and Pharmacy, "Carol Davila", 020021 Bucharest, Romania
- Department of Neurosurgery, "Bagdasar-Arseni" Clinical Emergency Hospital, 041915 Bucharest, Romania
| | - Antonio-Daniel Corlatescu
- Department of Neurosurgery, University of Medicine and Pharmacy, "Carol Davila", 020021 Bucharest, Romania
| | - Horia Petre Costin
- Department of Neurosurgery, University of Medicine and Pharmacy, "Carol Davila", 020021 Bucharest, Romania
| | - Adrian Dumitru
- Department of Neurosurgery, University of Medicine and Pharmacy, "Carol Davila", 020021 Bucharest, Romania
- Department of Morphopathology, University of Medicine and Pharmacy, "Carol Davila", 020021 Bucharest, Romania
- Emergency University Hospital Bucharest, 050098 Bucharest, Romania
| | - Alexandru Vlad Ciurea
- Department of Neurosurgery, University of Medicine and Pharmacy, "Carol Davila", 020021 Bucharest, Romania
- Sanador Clinical Hospital, 010991 Bucharest, Romania
| |
Collapse
|
6
|
Galli RA, Borsboom TC, Gineste C, Brocca L, Rossi M, Hwee DT, Malik FI, Bottinelli R, Gondin J, Pellegrino MA, de Winter JM, Ottenheijm CA. Tirasemtiv enhances submaximal muscle tension in an Acta1:p.Asp286Gly mouse model of nemaline myopathy. J Gen Physiol 2024; 156:e202313471. [PMID: 38376469 PMCID: PMC10876480 DOI: 10.1085/jgp.202313471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 12/01/2023] [Accepted: 01/30/2024] [Indexed: 02/21/2024] Open
Abstract
Nemaline myopathies are the most common form of congenital myopathies. Variants in ACTA1 (NEM3) comprise 15-25% of all nemaline myopathy cases. Patients harboring variants in ACTA1 present with a heterogeneous disease course characterized by stable or progressive muscle weakness and, in severe cases, respiratory failure and death. To date, no specific treatments are available. Since NEM3 is an actin-based thin filament disease, we tested the ability of tirasemtiv, a fast skeletal muscle troponin activator, to improve skeletal muscle function in a mouse model of NEM3, harboring the patient-based p.Asp286Gly variant in Acta1. Acute and long-term tirasemtiv treatment significantly increased muscle contractile capacity at submaximal stimulation frequencies in both fast-twitch extensor digitorum longus and gastrocnemius muscle, and intermediate-twitch diaphragm muscle in vitro and in vivo. Additionally, long-term tirasemtiv treatment in NEM3 mice resulted in a decreased respiratory rate with preserved minute volume, suggesting more efficient respiration. Altogether, our data support the therapeutic potential of fast skeletal muscle troponin activators in alleviating skeletal muscle weakness in a mouse model of NEM3 caused by the Acta1:p.Asp286Gly variant.
Collapse
Affiliation(s)
- Ricardo A. Galli
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, Amsterdam, The Netherlands
- Amsterdam Movement Sciences, Musculoskeletal Health and Tissue Function and Regeneration, Amsterdam, The Netherlands
| | - Tamara C. Borsboom
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, Amsterdam, The Netherlands
| | | | - Lorenza Brocca
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Maira Rossi
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Darren T. Hwee
- Research and Early Development, Cytokinetics Inc., South San Francisco, CA, USA
| | - Fady I. Malik
- Research and Early Development, Cytokinetics Inc., South San Francisco, CA, USA
| | - Roberto Bottinelli
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- IRCCS Mondino Foundation, Pavia, Italy
| | - Julien Gondin
- Aix-Marseille University, CNRS, CRMBM, Marseille, France
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, INSERM U1315, Université Lyon, Lyon, France
| | | | - Josine M. de Winter
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, Amsterdam, The Netherlands
- Amsterdam Movement Sciences, Musculoskeletal Health and Tissue Function and Regeneration, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam, The Netherlands
| | - Coen A.C. Ottenheijm
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Atherosclerosis, Amsterdam, The Netherlands
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
7
|
Bahat G, Ozkok S. The Current Landscape of Pharmacotherapies for Sarcopenia. Drugs Aging 2024; 41:83-112. [PMID: 38315328 DOI: 10.1007/s40266-023-01093-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2023] [Indexed: 02/07/2024]
Abstract
Sarcopenia is a skeletal muscle disorder characterized by progressive and generalized decline in muscle mass and function. Although it is mostly known as an age-related disorder, it can also occur secondary to systemic diseases such as malignancy or organ failure. It has demonstrated a significant relationship with adverse outcomes, e.g., falls, disabilities, and even mortality. Several breakthroughs have been made to find a pharmaceutical therapy for sarcopenia over the years, and some have come up with promising findings. Yet still no drug has been approved for its treatment. The key factor that makes finding an effective pharmacotherapy so challenging is the general paradigm of standalone/single diseases, traditionally adopted in medicine. Today, it is well known that sarcopenia is a complex disorder caused by multiple factors, e.g., imbalance in protein turnover, satellite cell and mitochondrial dysfunction, hormonal changes, low-grade inflammation, senescence, anorexia of aging, and behavioral factors such as low physical activity. Therefore, pharmaceuticals, either alone or combined, that exhibit multiple actions on these factors simultaneously will likely be the drug of choice to manage sarcopenia. Among various drug options explored throughout the years, testosterone still has the most cumulated evidence regarding its effects on muscle health and its safety. A mas receptor agonist, BIO101, stands out as a recent promising pharmaceutical. In addition to the conventional strategies (i.e., nutritional support and physical exercise), therapeutics with multiple targets of action or combination of multiple therapeutics with different targets/modes of action appear to promise greater benefit for the prevention and treatment of sarcopenia.
Collapse
Affiliation(s)
- Gulistan Bahat
- Division of Geriatrics, Department of Internal Medicine, Istanbul Medical School, Istanbul University, Capa, 34390, Istanbul, Turkey.
| | - Serdar Ozkok
- Division of Geriatrics, Department of Internal Medicine, Hatay Training and Research Hospital, Hatay, 31040, Turkey
| |
Collapse
|
8
|
Theme 10 - Disease Stratification and Phenotyping of Patients. Amyotroph Lateral Scler Frontotemporal Degener 2023; 24:230-244. [PMID: 37966327 DOI: 10.1080/21678421.2023.2260202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
|
9
|
Maragakis NJ, de Carvalho M, Weiss MD. Therapeutic targeting of ALS pathways: Refocusing an incomplete picture. Ann Clin Transl Neurol 2023; 10:1948-1971. [PMID: 37641443 PMCID: PMC10647018 DOI: 10.1002/acn3.51887] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/04/2023] [Accepted: 08/14/2023] [Indexed: 08/31/2023] Open
Abstract
Numerous potential amyotrophic lateral sclerosis (ALS)-relevant pathways have been hypothesized and studied preclinically, with subsequent translation to clinical trial. However, few successes have been observed with only modest effects. Along with an improved but incomplete understanding of ALS as a neurodegenerative disease is the evolution of more sophisticated and diverse in vitro and in vivo preclinical modeling platforms, as well as clinical trial designs. We highlight proposed pathological pathways that have been major therapeutic targets for investigational compounds. It is likely that the failures of so many of these therapeutic compounds may not have occurred because of lack of efficacy but rather because of a lack of preclinical modeling that would help define an appropriate disease pathway, as well as a failure to establish target engagement. These challenges are compounded by shortcomings in clinical trial design, including lack of biomarkers that could predict clinical success and studies that are underpowered. Although research investments have provided abundant insights into new ALS-relevant pathways, most have not yet been developed more fully to result in clinical study. In this review, we detail some of the important, well-established pathways, the therapeutics targeting them, and the subsequent clinical design. With an understanding of some of the shortcomings in translational efforts over the last three decades of ALS investigation, we propose that scientists and clinicians may choose to revisit some of these therapeutic pathways reviewed here with an eye toward improving preclinical modeling, biomarker development, and the investment in more sophisticated clinical trial designs.
Collapse
Affiliation(s)
| | - Mamede de Carvalho
- Faculdade de MedicinaInsqatituto de Medicina Molecular João Lobo Antunes, Centro Académico de Medicina de Lisboa, Universidade de LisboaLisbonPortugal
| | - Michael D. Weiss
- Department of NeurologyUniversity of WashingtonSeattleWashingtonUSA
| |
Collapse
|
10
|
Claassen WJ, Baelde RJ, Galli RA, de Winter JM, Ottenheijm CAC. Small molecule drugs to improve sarcomere function in those with acquired and inherited myopathies. Am J Physiol Cell Physiol 2023; 325:C60-C68. [PMID: 37212548 PMCID: PMC10281779 DOI: 10.1152/ajpcell.00047.2023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/15/2023] [Accepted: 05/15/2023] [Indexed: 05/23/2023]
Abstract
Muscle weakness is a hallmark of inherited or acquired myopathies. It is a major cause of functional impairment and can advance to life-threatening respiratory insufficiency. During the past decade, several small-molecule drugs that improve the contractility of skeletal muscle fibers have been developed. In this review, we provide an overview of the available literature and the mechanisms of action of small-molecule drugs that modulate the contractility of sarcomeres, the smallest contractile units in striated muscle, by acting on myosin and troponin. We also discuss their use in the treatment of skeletal myopathies. The first of three classes of drugs discussed here increase contractility by decreasing the dissociation rate of calcium from troponin and thereby sensitizing the muscle to calcium. The second two classes of drugs directly act on myosin and stimulate or inhibit the kinetics of myosin-actin interactions, which may be useful in patients with muscle weakness or stiffness.NEW & NOTEWORTHY During the past decade, several small molecule drugs that improve the contractility of skeletal muscle fibers have been developed. In this review, we provide an overview of the available literature and the mechanisms of action of small molecule drugs that modulate the contractility of sarcomeres, the smallest contractile units in striated muscle, by acting on myosin and troponin.
Collapse
Affiliation(s)
- Wout J Claassen
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan, Amsterdam, Netherlands
| | - Rianne J Baelde
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan, Amsterdam, Netherlands
| | - Ricardo A Galli
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan, Amsterdam, Netherlands
| | - Josine M de Winter
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan, Amsterdam, Netherlands
| | - Coen A C Ottenheijm
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan, Amsterdam, Netherlands
| |
Collapse
|
11
|
Issahaku AR, Ibrahim MAA, Mukelabai N, Soliman MES. Intermolecular And Dynamic Investigation of The Mechanism of Action of Reldesemtiv on Fast Skeletal Muscle Troponin Complex Toward the Treatment of Impaired Muscle Function. Protein J 2023:10.1007/s10930-023-10091-y. [PMID: 36959428 DOI: 10.1007/s10930-023-10091-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2023] [Indexed: 03/25/2023]
Abstract
Muscle weakness as a secondary feature of attenuated neuronal input often leads to disability and sometimes death in patients with neurogenic neuromuscular diseases. These impaired muscle function has been observed in several diseases including amyotrophic lateral sclerosis, Charcot-Marie-Tooth, spinal muscular atrophy and Myasthenia gravis. This has spurred the search for small molecules which could activate fast skeletal muscle troponin complex as a means to increase muscle strength. Discovered small molecules have however been punctuated by off-target and side effects leading to the development of the second-generation small molecule, Reldesemtiv. In this study, we investigated the impact of Reldesemtiv binding to the fast skeletal troponin complex and the molecular determinants that condition the therapeutic prowess of Redesemtiv through computational techniques. It was revealed that Reldesemtiv binding possibly potentiates troponin C compacting characterized by reduced exposure to solvent molecules which could favor the slow release of calcium ions and the resultant sensitization of the subunit to calcium. These conformational changes were underscored by conventional and carbon hydrogen bonds, pi-alkyl, pi-sulfur and halogen interactions between Reldesemtiv the binding site residues. Arg113 (-3.96 kcal/mol), Met116 (-2.23 kcal/mol), Val114 (-1.28 kcal/mol) and Met121 (-0.63 kcal/mol) of the switch region of the inhibitory subunit were among the residues that contributed the most to the total free binding energy of Reldesemtiv highlighting their importance. These findings present useful insights which could lay the foundation for the development of fast skeletal muscle small molecule activators with high specificity and potency.
Collapse
Affiliation(s)
- Abdul Rashid Issahaku
- Molecular Bio-Computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa
- West African Centre for Computational Research and Innovation, Accra, Ghana
| | - Mahmoud A A Ibrahim
- CompChem Research Group, Chemistry Department, Faculty of Science, Minia University, Minia, 61519, Egypt
| | - Namutula Mukelabai
- Department of Physiotherapy, School of Health Sciences, University of KwaZulu- Natal, Westville Campus, Durban, 4001, South Africa
| | - Mahmoud E S Soliman
- Molecular Bio-Computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa.
| |
Collapse
|
12
|
Shefner JM, Bedlack R, Andrews JA, Berry JD, Bowser R, Brown R, Glass JD, Maragakis NJ, Miller TM, Rothstein JD, Cudkowicz ME. Amyotrophic Lateral Sclerosis Clinical Trials and Interpretation of Functional End Points and Fluid Biomarkers: A Review. JAMA Neurol 2022; 79:1312-1318. [PMID: 36251310 DOI: 10.1001/jamaneurol.2022.3282] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Importance Clinical trial activity in amyotrophic lateral sclerosis (ALS) is dramatically increasing; as a result, trial modifications have been introduced to improve efficiency, outcome measures have been reassessed, and considerable discussion about the level of data necessary to advance a drug to approval has occurred. This review discusses what recent pivotal studies can teach the community about these topics. Observations By restricting inclusion and exclusion criteria, recent trials have enrolled populations distinct from previous studies. This has led to efficacy signals being observed in studies that are smaller and shorter than was thought feasible previously. However, such trials raise questions about generalizability of results. Small trials with equivocal clinical results also raise questions about the data necessary to lead to regulatory approval. The ALS Functional Rating Scale-Revised remains the most commonly used primary outcome measure; this review discusses innovations in its use. Blood neurofilament levels can predict prognosis in ALS and may be a sensitive indicator of biologic effect; current knowledge does not yet support its use as a primary outcome. Conclusions and Relevance It is now possible to use specific inclusion criteria to recruit a homogeneous patient population progressing at a specific rate; this will likely impact trials in the future. Generalizability of results on limited populations remains a concern. Although clinical outcomes remain the most appropriate primary outcome measures, fluid markers reflecting biologically important processes will assume more importance as more is learned about the association between such markers and clinical end points. The benefit of use of analytic strategies, such as responder analyses, is still uncertain.
Collapse
Affiliation(s)
| | | | - Jinsy A Andrews
- The Neurological Institute, Columbia University, New York, New York
| | - James D Berry
- Healey & AMG Center ALS, Massachusetts General Hospital, Boston
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Martin LJ, Adams DA, Niedzwiecki MV, Wong M. Aberrant DNA and RNA Methylation Occur in Spinal Cord and Skeletal Muscle of Human SOD1 Mouse Models of ALS and in Human ALS: Targeting DNA Methylation Is Therapeutic. Cells 2022; 11:3448. [PMID: 36359844 PMCID: PMC9657572 DOI: 10.3390/cells11213448] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 10/24/2022] [Accepted: 10/30/2022] [Indexed: 11/06/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal disease. Skeletal muscles and motor neurons (MNs) degenerate. ALS is a complex disease involving many genes in multiple tissues, the environment, cellular metabolism, and lifestyles. We hypothesized that epigenetic anomalies in DNA and RNA occur in ALS and examined this idea in: (1) mouse models of ALS, (2) human ALS, and (3) mouse ALS with therapeutic targeting of DNA methylation. Human superoxide dismutase-1 (hSOD1) transgenic (tg) mice were used. They expressed nonconditionally wildtype (WT) and the G93A and G37R mutant variants or skeletal muscle-restricted WT and G93A and G37R mutated forms. Age-matched non-tg mice were controls. hSOD1 mutant mice had increased DNA methyltransferase enzyme activity in spinal cord and skeletal muscle and increased 5-methylcytosine (5mC) levels. Genome-wide promoter CpG DNA methylation profiling in skeletal muscle of ALS mice identified hypermethylation notably in cytoskeletal genes. 5mC accumulated in spinal cord MNs and skeletal muscle satellite cells in mice. Significant increases in DNA methyltransferase-1 (DNMT1) and DNA methyltransferase-3A (DNMT3A) levels occurred in spinal cord nuclear and chromatin bound extracts of the different hSOD1 mouse lines. Mutant hSOD1 interacted with DNMT3A in skeletal muscle. 6-methyladenosine (6mA) RNA methylation was markedly increased or decreased in mouse spinal cord depending on hSOD1-G93A model, while fat mass and obesity associated protein was depleted and methyltransferase-like protein 3 was increased in spinal cord and skeletal muscle. Human ALS spinal cord had increased numbers of MNs and interneurons with nuclear 5mC, motor cortex had increased 5mC-positive neurons, while 6mA was severely depleted. Treatment of hSOD1-G93A mice with DNMT inhibitor improved motor function and extended lifespan by 25%. We conclude that DNA and RNA epigenetic anomalies are prominent in mouse and human ALS and are potentially targetable for disease-modifying therapeutics.
Collapse
Affiliation(s)
- Lee J. Martin
- Department of Pathology, Division of Neuropathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Pathobiology Graduate Training Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Danya A. Adams
- Department of Pathology, Division of Neuropathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Mark V. Niedzwiecki
- Department of Pathology, Division of Neuropathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Margaret Wong
- Department of Pathology, Division of Neuropathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
14
|
Khamaysa M, Pradat PF. Status of ALS Treatment, Insights into Therapeutic Challenges and Dilemmas. J Pers Med 2022; 12:1601. [PMID: 36294741 PMCID: PMC9605458 DOI: 10.3390/jpm12101601] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/14/2022] [Accepted: 09/23/2022] [Indexed: 12/18/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an extremely heterogeneous disease of motor neurons that eventually leads to death. Despite impressive advances in understanding the genetic, molecular, and pathological mechanisms of the disease, the only drug approved to date by both the FDA and EMA is riluzole, with a modest effect on survival. In this opinion view paper, we will discuss how to address some challenges for drug development in ALS at the conceptual, technological, and methodological levels. In addition, socioeconomic and ethical issues related to the legitimate need of patients to benefit quickly from new treatments will also be addressed. In conclusion, this brief review takes a more optimistic view, given the recent approval of two new drugs in some countries and the development of targeted gene therapies.
Collapse
Affiliation(s)
- Mohammed Khamaysa
- Laboratoire d’Imagerie Biomédicale, Sorbonne Université, CNRS, INSERM, 75006 Paris, France
| | - Pierre-François Pradat
- Laboratoire d’Imagerie Biomédicale, Sorbonne Université, CNRS, INSERM, 75006 Paris, France
- Centre Référent SLA, Département de Neurologie, AP-HP, Hôpital Pitié-Salpêtrière, 75013 Paris, France
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, Ulster University, C-TRIC, Altnagelvin Hospital, Derry-Londonderry BT47 6SB, UK
| |
Collapse
|
15
|
Morelli C, Ingrasciotta G, Jacoby D, Masri A, Olivotto I. Sarcomere protein modulation: The new frontier in cardiovascular medicine and beyond. Eur J Intern Med 2022; 102:1-7. [PMID: 35534374 DOI: 10.1016/j.ejim.2022.04.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/14/2022] [Accepted: 04/17/2022] [Indexed: 01/10/2023]
Abstract
Over the past decade, the constant progress in science and technologies has provided innovative drug molecules that address specific disease mechanisms thus opening the era of drugs targeting the underlying pathophysiology of the disease. In this scenario, a new paradigm of modulation has emerged, following the development of small molecules capable of interfering with sarcomere contractile proteins. Potential applications include heart muscle disease and various forms of heart failure, although promising targets also include conditions affecting the skeletal muscle, such as degenerative neuromuscular diseases. In cardiac patients, a cardiac myosin stimulator, omecamtiv mecarbil, has shown efficacy in heart failure with reduced systolic function, lowering heart failure related events or cardiovascular death, while two inhibitors, mavacamten and aficamten, in randomized trials targeting hypertrophic cardiomyopathy, have been shown to reduce hypercontractility and left ventricular outflow obstruction improving functional capacity. Based on years of intensive basic and translational research, these agents are the prototypes of active pipelines promising to deliver an array of molecules in the near future. We here review the available evidence and future perspectives of myosin modulation in cardiovascular medicine.
Collapse
Affiliation(s)
- Cristina Morelli
- Azienda Ospedaliera Universitaria Careggi and University of Florence, Florence, Italy
| | - Gessica Ingrasciotta
- Azienda Ospedaliera Universitaria Careggi and University of Florence, Florence, Italy
| | - Daniel Jacoby
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale University, New Haven, CT, USA
| | - Ahmad Masri
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| | - Iacopo Olivotto
- Azienda Ospedaliera Universitaria Careggi and University of Florence, Florence, Italy.
| |
Collapse
|
16
|
Fournier CN. Considerations for Amyotrophic Lateral Sclerosis (ALS) Clinical Trial Design. Neurotherapeutics 2022; 19:1180-1192. [PMID: 35819713 PMCID: PMC9275386 DOI: 10.1007/s13311-022-01271-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2022] [Indexed: 11/20/2022] Open
Abstract
Thoughtful clinical trial design is critical for efficient therapeutic development, particularly in the field of amyotrophic lateral sclerosis (ALS), where trials often aim to detect modest treatment effects among a population with heterogeneous disease progression. Appropriate outcome measure selection is necessary for trials to provide decisive and informative results. Investigators must consider the outcome measure's reliability, responsiveness to detect change when change has actually occurred, clinical relevance, and psychometric performance. ALS clinical trials can also be performed more efficiently by utilizing statistical enrichment techniques. Innovations in ALS prediction models allow for selection of participants with less heterogeneity in disease progression rates without requiring a lead-in period, or participants can be stratified according to predicted progression. Statistical enrichment can reduce the needed sample size and improve study power, but investigators must find a balance between optimizing statistical efficiency and retaining generalizability of study findings to the broader ALS population. Additional progress is still needed for biomarker development and validation to confirm target engagement in ALS treatment trials. Selection of an appropriate biofluid biomarker depends on the treatment mechanism of interest, and biomarker studies should be incorporated into early phase trials. Inclusion of patients with ALS as advisors and advocates can strengthen clinical trial design and study retention, but more engagement efforts are needed to improve diversity and equity in ALS research studies. Another challenge for ALS therapeutic development is identifying ways to respect patient autonomy and improve access to experimental treatment, something that is strongly desired by many patients with ALS and ALS advocacy organizations. Expanded access programs that run concurrently to well-designed and adequately powered randomized controlled trials may provide an opportunity to broaden access to promising therapeutics without compromising scientific integrity or rushing regulatory approval of therapies without adequate proof of efficacy.
Collapse
Affiliation(s)
- Christina N Fournier
- Department of Neurology, Emory University, Atlanta, GA, USA.
- Department of Veterans Affairs, Atlanta, GA, USA.
| |
Collapse
|
17
|
Mahmud Z, Tikunova S, Belevych N, Wagg CS, Zhabyeyev P, Liu PB, Rasicci DV, Yengo CM, Oudit GY, Lopaschuk GD, Reiser PJ, Davis JP, Hwang PM. Small Molecule RPI-194 Stabilizes Activated Troponin to Increase the Calcium Sensitivity of Striated Muscle Contraction. Front Physiol 2022; 13:892979. [PMID: 35755445 PMCID: PMC9213791 DOI: 10.3389/fphys.2022.892979] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/10/2022] [Indexed: 11/13/2022] Open
Abstract
Small molecule cardiac troponin activators could potentially enhance cardiac muscle contraction in the treatment of systolic heart failure. We designed a small molecule, RPI-194, to bind cardiac/slow skeletal muscle troponin (Cardiac muscle and slow skeletal muscle share a common isoform of the troponin C subunit.) Using solution NMR and stopped flow fluorescence spectroscopy, we determined that RPI-194 binds to cardiac troponin with a dissociation constant KD of 6-24 μM, stabilizing the activated complex between troponin C and the switch region of troponin I. The interaction between RPI-194 and troponin C is weak (KD 311 μM) in the absence of the switch region. RPI-194 acts as a calcium sensitizer, shifting the pCa50 of isometric contraction from 6.28 to 6.99 in mouse slow skeletal muscle fibers and from 5.68 to 5.96 in skinned cardiac trabeculae at 100 μM concentration. There is also some cross-reactivity with fast skeletal muscle fibers (pCa50 increases from 6.27 to 6.52). In the slack test performed on the same skinned skeletal muscle fibers, RPI-194 slowed the velocity of unloaded shortening at saturating calcium concentrations, suggesting that it slows the rate of actin-myosin cross-bridge cycling under these conditions. However, RPI-194 had no effect on the ATPase activity of purified actin-myosin. In isolated unloaded mouse cardiomyocytes, RPI-194 markedly decreased the velocity and amplitude of contractions. In contrast, cardiac function was preserved in mouse isolated perfused working hearts. In summary, the novel troponin activator RPI-194 acts as a calcium sensitizer in all striated muscle types. Surprisingly, it also slows the velocity of unloaded contraction, but the cause and significance of this is uncertain at this time. RPI-194 represents a new class of non-specific troponin activator that could potentially be used either to enhance cardiac muscle contractility in the setting of systolic heart failure or to enhance skeletal muscle contraction in neuromuscular disorders.
Collapse
Affiliation(s)
- Zabed Mahmud
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Svetlana Tikunova
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, United States
| | - Natalya Belevych
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, United States
| | - Cory S Wagg
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Pavel Zhabyeyev
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Philip B Liu
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - David V Rasicci
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, University Park, PA, United States
| | - Christopher M Yengo
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, University Park, PA, United States
| | - Gavin Y Oudit
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Gary D Lopaschuk
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Peter J Reiser
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, United States
| | - Jonathan P Davis
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, United States
| | - Peter M Hwang
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada.,Department of Medicine, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
18
|
Collibee SE, Bergnes G, Chuang C, Ashcraft L, Gardina J, Garard M, Jamison CR, Lu K, Lu PP, Muci A, Romero A, Valkevich E, Wang W, Warrington J, Yao B, Durham N, Hartman J, Marquez A, Hinken A, Schaletzky J, Xu D, Hwee DT, Morgans D, Malik FI, Morgan BP. Discovery of Reldesemtiv, a Fast Skeletal Muscle Troponin Activator for the Treatment of Impaired Muscle Function. J Med Chem 2021; 64:14930-14941. [PMID: 34636234 DOI: 10.1021/acs.jmedchem.1c01067] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The discovery of reldesemtiv, a second-generation fast skeletal muscle troponin activator (FSTA) that increases force production at submaximal stimulation frequencies, is reported. Property-based optimization of high throughput screening hit 1 led to compounds with improved free exposure and in vivo muscle activation potency compared to the first-generation FSTA, tirasemtiv. Reldesemtiv demonstrated increased muscle force generation in a phase 1 clinical trial and is currently being evaluated in clinical trials for the treatment of amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Scott E Collibee
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Gustave Bergnes
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Chihyuan Chuang
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Luke Ashcraft
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Jeffrey Gardina
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Marc Garard
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Chris R Jamison
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Kevin Lu
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Pu-Ping Lu
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Alexander Muci
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Antonio Romero
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Ellen Valkevich
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Wenyue Wang
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Jeffrey Warrington
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Bing Yao
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Nickie Durham
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - James Hartman
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Anna Marquez
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Aaron Hinken
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Julia Schaletzky
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Donghong Xu
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Darren T Hwee
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - David Morgans
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Fady I Malik
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Bradley P Morgan
- Cytokinetics, Inc., 280 East Grand Avenue, South San Francisco, California 94080, United States
| |
Collapse
|
19
|
Wong C, Stavrou M, Elliott E, Gregory JM, Leigh N, Pinto AA, Williams TL, Chataway J, Swingler R, Parmar MKB, Stallard N, Weir CJ, Parker RA, Chaouch A, Hamdalla H, Ealing J, Gorrie G, Morrison I, Duncan C, Connelly P, Carod-Artal FJ, Davenport R, Reitboeck PG, Radunovic A, Srinivasan V, Preston J, Mehta AR, Leighton D, Glasmacher S, Beswick E, Williamson J, Stenson A, Weaver C, Newton J, Lyle D, Dakin R, Macleod M, Pal S, Chandran S. Clinical trials in amyotrophic lateral sclerosis: a systematic review and perspective. Brain Commun 2021; 3:fcab242. [PMID: 34901853 PMCID: PMC8659356 DOI: 10.1093/braincomms/fcab242] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 09/07/2021] [Accepted: 09/13/2021] [Indexed: 12/15/2022] Open
Abstract
Amyotrophic lateral sclerosis is a progressive and devastating neurodegenerative disease. Despite decades of clinical trials, effective disease-modifying drugs remain scarce. To understand the challenges of trial design and delivery, we performed a systematic review of Phase II, Phase II/III and Phase III amyotrophic lateral sclerosis clinical drug trials on trial registries and PubMed between 2008 and 2019. We identified 125 trials, investigating 76 drugs and recruiting more than 15 000 people with amyotrophic lateral sclerosis. About 90% of trials used traditional fixed designs. The limitations in understanding of disease biology, outcome measures, resources and barriers to trial participation in a rapidly progressive, disabling and heterogenous disease hindered timely and definitive evaluation of drugs in two-arm trials. Innovative trial designs, especially adaptive platform trials may offer significant efficiency gains to this end. We propose a flexible and scalable multi-arm, multi-stage trial platform where opportunities to participate in a clinical trial can become the default for people with amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Charis Wong
- Centre for Clinical Brain Sciences, Chancellor's Building, 49 Little France Crescent, The University of Edinburgh, Edinburgh, EH16 4SB, UK
- Anne Rowling Regenerative Neurology Clinic, Chancellor's Building, 49 Little France Crescent, The University of Edinburgh, Edinburgh, EH16 4SB, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, FU303F, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Maria Stavrou
- Centre for Clinical Brain Sciences, Chancellor's Building, 49 Little France Crescent, The University of Edinburgh, Edinburgh, EH16 4SB, UK
- Anne Rowling Regenerative Neurology Clinic, Chancellor's Building, 49 Little France Crescent, The University of Edinburgh, Edinburgh, EH16 4SB, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, FU303F, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
- UK Dementia Research Institute, Chancellor’s Building, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Elizabeth Elliott
- Centre for Clinical Brain Sciences, Chancellor's Building, 49 Little France Crescent, The University of Edinburgh, Edinburgh, EH16 4SB, UK
- Anne Rowling Regenerative Neurology Clinic, Chancellor's Building, 49 Little France Crescent, The University of Edinburgh, Edinburgh, EH16 4SB, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, FU303F, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
- UK Dementia Research Institute, Chancellor’s Building, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Jenna M Gregory
- Centre for Clinical Brain Sciences, Chancellor's Building, 49 Little France Crescent, The University of Edinburgh, Edinburgh, EH16 4SB, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, FU303F, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
- UK Dementia Research Institute, Chancellor’s Building, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Nigel Leigh
- Department of Neuroscience, Brighton and Sussex Medical School, University of Sussex, Brighton, BN1 9PX, UK
| | - Ashwin A Pinto
- Neurology Department, Wessex Neurosciences Centre, Southampton General Hospital, Southampton, SO16 6YD, UK
| | - Timothy L Williams
- Department of Neurology, Royal Victoria Infirmary, Newcastle upon Tyne NE1 4LP, UK
| | - Jeremy Chataway
- Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London WC1B 5EH, UK
- National Institute for Health Research, University College London Hospitals, Biomedical Research Centre, London, W1T 7DN, UK
- MRC CTU at UCL, Institute of Clinical Trials and Methodology, University College London, London, WC1V 6LJ, UK
| | - Robert Swingler
- Euan MacDonald Centre for MND Research, University of Edinburgh, FU303F, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Mahesh K B Parmar
- MRC CTU at UCL, Institute of Clinical Trials and Methodology, University College London, London, WC1V 6LJ, UK
| | - Nigel Stallard
- Statistics and Epidemiology, Division of Health Sciences, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
| | - Christopher J Weir
- Edinburgh Clinical Trials Unit, Usher Institute, Level 2, NINE Edinburgh BioQuarter, 9 Little France Road, Edinburgh EH16 4UX, UK
| | - Richard A Parker
- Edinburgh Clinical Trials Unit, Usher Institute, Level 2, NINE Edinburgh BioQuarter, 9 Little France Road, Edinburgh EH16 4UX, UK
| | - Amina Chaouch
- Motor Neurone Disease Care Centre, Manchester Centre for Clinical Neurosciences, Salford, M6 8HD, UK
| | - Hisham Hamdalla
- Motor Neurone Disease Care Centre, Manchester Centre for Clinical Neurosciences, Salford, M6 8HD, UK
| | - John Ealing
- Motor Neurone Disease Care Centre, Manchester Centre for Clinical Neurosciences, Salford, M6 8HD, UK
| | - George Gorrie
- Department of Neurology, Institute of Neurological Sciences, Queen Elizabeth University Hospital, NHS Greater Glasgow and Clyde, Glasgow, G51 4TF, UK
| | - Ian Morrison
- Department of Neurology, NHS Tayside, Dundee, DD2 1UB, UK
| | - Callum Duncan
- Department of Neurology, Aberdeen Royal Infirmary, Aberdeen, AB25 2ZN, UK
| | - Peter Connelly
- NHS Research Scotland Neuroprogressive Disorders and Dementia Network, Ninewells Hospital, Dundee, DD1 9SY, UK
| | | | - Richard Davenport
- Anne Rowling Regenerative Neurology Clinic, Chancellor's Building, 49 Little France Crescent, The University of Edinburgh, Edinburgh, EH16 4SB, UK
- Department of Clinical Neurosciences, NHS Lothian, Edinburgh, EH16 4SA, UK
| | - Pablo Garcia Reitboeck
- Atkinson Morley Regional Neurosciences Centre, St. George's University Hospitals NHS Foundation Trust, London SW17 0QT, UK
| | | | | | - Jenny Preston
- Department of Neurology, NHS Ayrshire & Arran, KA12 8SS, UK
| | - Arpan R Mehta
- Centre for Clinical Brain Sciences, Chancellor's Building, 49 Little France Crescent, The University of Edinburgh, Edinburgh, EH16 4SB, UK
- Anne Rowling Regenerative Neurology Clinic, Chancellor's Building, 49 Little France Crescent, The University of Edinburgh, Edinburgh, EH16 4SB, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, FU303F, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
- UK Dementia Research Institute, Chancellor’s Building, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Danielle Leighton
- Centre for Clinical Brain Sciences, Chancellor's Building, 49 Little France Crescent, The University of Edinburgh, Edinburgh, EH16 4SB, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, FU303F, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Stella Glasmacher
- Centre for Clinical Brain Sciences, Chancellor's Building, 49 Little France Crescent, The University of Edinburgh, Edinburgh, EH16 4SB, UK
- Anne Rowling Regenerative Neurology Clinic, Chancellor's Building, 49 Little France Crescent, The University of Edinburgh, Edinburgh, EH16 4SB, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, FU303F, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Emily Beswick
- Centre for Clinical Brain Sciences, Chancellor's Building, 49 Little France Crescent, The University of Edinburgh, Edinburgh, EH16 4SB, UK
- Anne Rowling Regenerative Neurology Clinic, Chancellor's Building, 49 Little France Crescent, The University of Edinburgh, Edinburgh, EH16 4SB, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, FU303F, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Jill Williamson
- Centre for Clinical Brain Sciences, Chancellor's Building, 49 Little France Crescent, The University of Edinburgh, Edinburgh, EH16 4SB, UK
- Anne Rowling Regenerative Neurology Clinic, Chancellor's Building, 49 Little France Crescent, The University of Edinburgh, Edinburgh, EH16 4SB, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, FU303F, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Amy Stenson
- Centre for Clinical Brain Sciences, Chancellor's Building, 49 Little France Crescent, The University of Edinburgh, Edinburgh, EH16 4SB, UK
- Anne Rowling Regenerative Neurology Clinic, Chancellor's Building, 49 Little France Crescent, The University of Edinburgh, Edinburgh, EH16 4SB, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, FU303F, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Christine Weaver
- Centre for Clinical Brain Sciences, Chancellor's Building, 49 Little France Crescent, The University of Edinburgh, Edinburgh, EH16 4SB, UK
- Anne Rowling Regenerative Neurology Clinic, Chancellor's Building, 49 Little France Crescent, The University of Edinburgh, Edinburgh, EH16 4SB, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, FU303F, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Judith Newton
- Centre for Clinical Brain Sciences, Chancellor's Building, 49 Little France Crescent, The University of Edinburgh, Edinburgh, EH16 4SB, UK
- Anne Rowling Regenerative Neurology Clinic, Chancellor's Building, 49 Little France Crescent, The University of Edinburgh, Edinburgh, EH16 4SB, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, FU303F, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Dawn Lyle
- Centre for Clinical Brain Sciences, Chancellor's Building, 49 Little France Crescent, The University of Edinburgh, Edinburgh, EH16 4SB, UK
- Anne Rowling Regenerative Neurology Clinic, Chancellor's Building, 49 Little France Crescent, The University of Edinburgh, Edinburgh, EH16 4SB, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, FU303F, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Rachel Dakin
- Centre for Clinical Brain Sciences, Chancellor's Building, 49 Little France Crescent, The University of Edinburgh, Edinburgh, EH16 4SB, UK
- Anne Rowling Regenerative Neurology Clinic, Chancellor's Building, 49 Little France Crescent, The University of Edinburgh, Edinburgh, EH16 4SB, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, FU303F, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Malcolm Macleod
- Centre for Clinical Brain Sciences, Chancellor's Building, 49 Little France Crescent, The University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Suvankar Pal
- Centre for Clinical Brain Sciences, Chancellor's Building, 49 Little France Crescent, The University of Edinburgh, Edinburgh, EH16 4SB, UK
- Anne Rowling Regenerative Neurology Clinic, Chancellor's Building, 49 Little France Crescent, The University of Edinburgh, Edinburgh, EH16 4SB, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, FU303F, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Siddharthan Chandran
- Centre for Clinical Brain Sciences, Chancellor's Building, 49 Little France Crescent, The University of Edinburgh, Edinburgh, EH16 4SB, UK
- Anne Rowling Regenerative Neurology Clinic, Chancellor's Building, 49 Little France Crescent, The University of Edinburgh, Edinburgh, EH16 4SB, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, FU303F, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
- UK Dementia Research Institute, Chancellor’s Building, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| |
Collapse
|
20
|
van de Locht M, Borsboom TC, Winter JM, Ottenheijm CAC. Troponin Variants in Congenital Myopathies: How They Affect Skeletal Muscle Mechanics. Int J Mol Sci 2021; 22:ijms22179187. [PMID: 34502093 PMCID: PMC8430961 DOI: 10.3390/ijms22179187] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/19/2021] [Accepted: 08/23/2021] [Indexed: 02/05/2023] Open
Abstract
The troponin complex is a key regulator of muscle contraction. Multiple variants in skeletal troponin encoding genes result in congenital myopathies. TNNC2 has been implicated in a novel congenital myopathy, TNNI2 and TNNT3 in distal arthrogryposis (DA), and TNNT1 and TNNT3 in nemaline myopathy (NEM). Variants in skeletal troponin encoding genes compromise sarcomere function, e.g., by altering the Ca2+ sensitivity of force or by inducing atrophy. Several potential therapeutic strategies are available to counter the effects of variants, such as troponin activators, introduction of wild-type protein through AAV gene therapy, and myosin modulation to improve muscle contraction. The mechanisms underlying the pathophysiological effects of the variants in skeletal troponin encoding genes are incompletely understood. Furthermore, limited knowledge is available on the structure of skeletal troponin. This review focusses on the physiology of slow and fast skeletal troponin and the pathophysiology of reported variants in skeletal troponin encoding genes. A better understanding of the pathophysiological effects of these variants, together with enhanced knowledge regarding the structure of slow and fast skeletal troponin, will direct the development of treatment strategies.
Collapse
|
21
|
de Winter JM, Gineste C, Minardi E, Brocca L, Rossi M, Borsboom T, Beggs AH, Bernard M, Bendahan D, Hwee DT, Malik FI, Pellegrino MA, Bottinelli R, Gondin J, Ottenheijm CAC. Acute and chronic tirasemtiv treatment improves in vivo and in vitro muscle performance in actin-based nemaline myopathy mice. Hum Mol Genet 2021; 30:1305-1320. [PMID: 33909041 PMCID: PMC8255131 DOI: 10.1093/hmg/ddab112] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/09/2021] [Accepted: 04/07/2021] [Indexed: 12/13/2022] Open
Abstract
Nemaline myopathy, a disease of the actin-based thin filament, is one of the most frequent congenital myopathies. To date, no specific therapy is available to treat muscle weakness in nemaline myopathy. We tested the ability of tirasemtiv, a fast skeletal troponin activator that targets the thin filament, to augment muscle force-both in vivo and in vitro-in a nemaline myopathy mouse model with a mutation (H40Y) in Acta1. In Acta1H40Y mice, treatment with tirasemtiv increased the force response of muscles to submaximal stimulation frequencies. This resulted in a reduced energetic cost of force generation, which increases the force production during a fatigue protocol. The inotropic effects of tirasemtiv were present in locomotor muscles and, albeit to a lesser extent, in respiratory muscles, and they persisted during chronic treatment, an important finding as respiratory failure is the main cause of death in patients with congenital myopathy. Finally, translational studies on permeabilized muscle fibers isolated from a biopsy of a patient with the ACTA1H40Y mutation revealed that at physiological Ca2+ concentrations, tirasemtiv increased force generation to values that were close to those generated in muscle fibers of healthy subjects. These findings indicate the therapeutic potential of fast skeletal muscle troponin activators to improve muscle function in nemaline myopathy due to the ACTA1H40Y mutation, and future studies should assess their merit for other forms of nemaline myopathy and for other congenital myopathies.
Collapse
Affiliation(s)
- Josine M de Winter
- Department of Physiology, Amsterdam UMC (location VUmc), Amsterdam 1081 HV, The Netherlands
| | | | - Elisa Minardi
- Department of Molecular Medicine, University of Pavia, Pavia 27100, Italy
| | - Lorenza Brocca
- Department of Molecular Medicine, University of Pavia, Pavia 27100, Italy
| | - Maira Rossi
- Department of Molecular Medicine, University of Pavia, Pavia 27100, Italy
| | - Tamara Borsboom
- Department of Physiology, Amsterdam UMC (location VUmc), Amsterdam 1081 HV, The Netherlands
| | - Alan H Beggs
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Monique Bernard
- Aix-Marseille Univ, CNRS, CRMBM, UMR 7339, 13005 Marseille, France
| | - David Bendahan
- Aix-Marseille Univ, CNRS, CRMBM, UMR 7339, 13005 Marseille, France
| | - Darren T Hwee
- Research and Early Development, Cytokinetics Inc., South San Francisco, CA 94080, USA
| | - Fady I Malik
- Research and Early Development, Cytokinetics Inc., South San Francisco, CA 94080, USA
| | - Maria Antonietta Pellegrino
- Department of Molecular Medicine, University of Pavia, Pavia 27100, Italy
- Interdipartimental Centre for Biology and Sport Medicine, University of Pavia, Pavia 27100, Italy
| | - Roberto Bottinelli
- Department of Molecular Medicine, University of Pavia, Pavia 27100, Italy
- Istituti Clinici Maugeri (IRCCS), Scientific Institute of Pavia, Pavia 27100, Italy
| | - Julien Gondin
- Aix-Marseille Univ, CNRS, CRMBM, UMR 7339, 13005 Marseille, France
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS 5310, INSERM U1217, 69008, Lyon, France
| | - Coen A C Ottenheijm
- Department of Physiology, Amsterdam UMC (location VUmc), Amsterdam 1081 HV, The Netherlands
| |
Collapse
|
22
|
van de Locht M, Donkervoort S, de Winter JM, Conijn S, Begthel L, Kusters B, Mohassel P, Hu Y, Medne L, Quinn C, Moore SA, Foley AR, Seo G, Hwee DT, Malik FI, Irving T, Ma W, Granzier HL, Kamsteeg EJ, Immadisetty K, Kekenes-Huskey P, Pinto JR, Voermans N, Bönnemann CG, Ottenheijm CA. Pathogenic variants in TNNC2 cause congenital myopathy due to an impaired force response to calcium. J Clin Invest 2021; 131:145700. [PMID: 33755597 PMCID: PMC8087209 DOI: 10.1172/jci145700] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 03/18/2021] [Indexed: 12/11/2022] Open
Abstract
Troponin C (TnC) is a critical regulator of skeletal muscle contraction; it binds Ca2+ to activate muscle contraction. Surprisingly, the gene encoding fast skeletal TnC (TNNC2) has not yet been implicated in muscle disease. Here, we report 2 families with pathogenic variants in TNNC2. Patients present with a distinct, dominantly inherited congenital muscle disease. Molecular dynamics simulations suggested that the pathomechanisms by which the variants cause muscle disease include disruption of the binding sites for Ca2+ and for troponin I. In line with these findings, physiological studies in myofibers isolated from patients' biopsies revealed a markedly reduced force response of the sarcomeres to [Ca2+]. This pathomechanism was further confirmed in experiments in which contractile dysfunction was evoked by replacing TnC in myofibers from healthy control subjects with recombinant, mutant TnC. Conversely, the contractile dysfunction of myofibers from patients was repaired by replacing endogenous, mutant TnC with recombinant, wild-type TnC. Finally, we tested the therapeutic potential of the fast skeletal muscle troponin activator tirasemtiv in patients' myofibers and showed that the contractile dysfunction was repaired. Thus, our data reveal that pathogenic variants in TNNC2 cause congenital muscle disease, and they provide therapeutic angles to repair muscle contractility.
Collapse
Affiliation(s)
- Martijn van de Locht
- Deptartment of Physiology, Amsterdam UMC (location VUmc), Amsterdam, Netherlands
| | - Sandra Donkervoort
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland, USA
| | - Josine M. de Winter
- Deptartment of Physiology, Amsterdam UMC (location VUmc), Amsterdam, Netherlands
| | - Stefan Conijn
- Deptartment of Physiology, Amsterdam UMC (location VUmc), Amsterdam, Netherlands
| | - Leon Begthel
- Deptartment of Physiology, Amsterdam UMC (location VUmc), Amsterdam, Netherlands
| | - Benno Kusters
- Department of Neurology and Human Genetics, Radboud University Medical Center, Nijmegen, Netherlands
| | - Payam Mohassel
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland, USA
| | - Ying Hu
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland, USA
| | - Livija Medne
- Division of Human Genetics, Department of Pediatrics, Individualized Medical Genetics Center, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Colin Quinn
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Steven A. Moore
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - A. Reghan Foley
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland, USA
| | - Gwimoon Seo
- Protein Expression Facility, Institute of Molecular Biophysics, The Florida State University, Tallahassee, Florida, USA
| | - Darren T. Hwee
- Research and Early Development, Cytokinetics Inc., South San Francisco, California, USA
| | - Fady I. Malik
- Research and Early Development, Cytokinetics Inc., South San Francisco, California, USA
| | - Thomas Irving
- BioCAT, Illinois Institute of Technology, Chicago, Illinois, USA
| | - Weikang Ma
- BioCAT, Illinois Institute of Technology, Chicago, Illinois, USA
| | - Henk L. Granzier
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| | - Erik-Jan Kamsteeg
- Department of Neurology and Human Genetics, Radboud University Medical Center, Nijmegen, Netherlands
| | - Kalyan Immadisetty
- Department of Cell and Molecular Physiology, Loyola University, Chicago, Illinois, USA
| | - Peter Kekenes-Huskey
- Department of Cell and Molecular Physiology, Loyola University, Chicago, Illinois, USA
| | - José R. Pinto
- Department of Biomedical Sciences, The Florida State University College of Medicine, Tallahassee, Florida, USA
| | - Nicol Voermans
- Department of Neurology and Human Genetics, Radboud University Medical Center, Nijmegen, Netherlands
| | - Carsten G. Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland, USA
| | - Coen A.C. Ottenheijm
- Deptartment of Physiology, Amsterdam UMC (location VUmc), Amsterdam, Netherlands
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
23
|
Karpicheva OE. Hallmark Features of the Tropomyosin
Regulatory Function in Several Variants of Congenital Myopathy. J EVOL BIOCHEM PHYS+ 2021. [DOI: 10.1134/s0022093021030133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
24
|
Li MX, Mercier P, Hartman JJ, Sykes BD. Structural Basis of Tirasemtiv Activation of Fast Skeletal Muscle. J Med Chem 2021; 64:3026-3034. [PMID: 33703886 DOI: 10.1021/acs.jmedchem.0c01412] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Troponin regulates the calcium-mediated activation of skeletal muscle. Muscle weakness in diseases such as amyotrophic lateral sclerosis and spinal muscular atrophy occurs from diminished neuromuscular output. The first direct fast skeletal troponin activator, tirasemtiv, amplifies the response of muscle to neuromuscular input. Tirasemtiv binds selectively and strongly to fast skeletal troponin, slowing the rate of calcium release and sensitizing muscle to calcium. We report the solution NMR structure of tirasemtiv bound to a fast skeletal troponin C-troponin I chimera. The structure reveals that tirasemtiv binds in a hydrophobic pocket between the regulatory domain of troponin C and the switch region of troponin I, which overlaps with that of Anapoe in the X-ray structure of skeletal troponin. Multiple interactions stabilize the troponin C-troponin I interface, increase the affinity of troponin C for the switch region of fast skeletal troponin I, and drive the equilibrium toward the active state.
Collapse
Affiliation(s)
- Monica X Li
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Pascal Mercier
- National High Field NMR Centre, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - James J Hartman
- Cytokinetics, Inc., South San Francisco, California 94080, United States
| | - Brian D Sykes
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| |
Collapse
|
25
|
Scaricamazza S, Salvatori I, Ferri A, Valle C. Skeletal Muscle in ALS: An Unappreciated Therapeutic Opportunity? Cells 2021; 10:525. [PMID: 33801336 PMCID: PMC8000428 DOI: 10.3390/cells10030525] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder characterized by the selective degeneration of upper and lower motor neurons and by the progressive weakness and paralysis of voluntary muscles. Despite intense research efforts and numerous clinical trials, it is still an incurable disease. ALS had long been considered a pure motor neuron disease; however, recent studies have shown that motor neuron protection is not sufficient to prevent the course of the disease since the dismantlement of neuromuscular junctions occurs before motor neuron degeneration. Skeletal muscle alterations have been described in the early stages of the disease, and they seem to be mainly involved in the "dying back" phenomenon of motor neurons and metabolic dysfunctions. In recent years, skeletal muscles have been considered crucial not only for the etiology of ALS but also for its treatment. Here, we review clinical and preclinical studies that targeted skeletal muscles and discuss the different approaches, including pharmacological interventions, supplements or diets, genetic modifications, and training programs.
Collapse
Affiliation(s)
- Silvia Scaricamazza
- Fondazione Santa Lucia IRCCS, c/o CERC, 00143 Rome, Italy; (S.S.); (I.S.)
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Illari Salvatori
- Fondazione Santa Lucia IRCCS, c/o CERC, 00143 Rome, Italy; (S.S.); (I.S.)
- Department of Experimental Medicine, University of Rome “La Sapienza”, 00161 Rome, Italy
| | - Alberto Ferri
- Fondazione Santa Lucia IRCCS, c/o CERC, 00143 Rome, Italy; (S.S.); (I.S.)
- Institute of Translational Pharmacology, National Research Council, 00133 Rome, Italy
| | - Cristiana Valle
- Fondazione Santa Lucia IRCCS, c/o CERC, 00143 Rome, Italy; (S.S.); (I.S.)
- Institute of Translational Pharmacology, National Research Council, 00133 Rome, Italy
| |
Collapse
|
26
|
Beaulieu D, Berry JD, Paganoni S, Glass JD, Fournier C, Cuerdo J, Schactman M, Ennist DL. Development and validation of a machine-learning ALS survival model lacking vital capacity (VC-Free) for use in clinical trials during the COVID-19 pandemic. Amyotroph Lateral Scler Frontotemporal Degener 2021; 22:22-32. [PMID: 34348539 DOI: 10.1080/21678421.2021.1924207] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 04/27/2021] [Accepted: 04/27/2021] [Indexed: 10/20/2022]
Abstract
Introduction: Vital capacity (VC) is routinely used for ALS clinical trial eligibility determinations, often to exclude patients unlikely to survive trial duration. However, spirometry has been limited by the COVID-19 pandemic. We developed a machine-learning survival model without the use of baseline VC and asked whether it could stratify clinical trial participants and a wider ALS clinic population. Methods. A gradient boosting machine survival model lacking baseline VC (VC-Free) was trained using the PRO-ACT ALS database and compared to a multivariable model that included VC (VCI) and a univariable baseline %VC model (UNI). Discrimination, calibration-in-the-large and calibration slope were quantified. Models were validated using 10-fold internal cross validation, the VITALITY-ALS clinical trial placebo arm and data from the Emory University tertiary care clinic. Simulations were performed using each model to estimate survival of patients predicted to have a > 50% one year survival probability. Results. The VC-Free model suffered a minor performance decline compared to the VCI model yet retained strong discrimination for stratifying ALS patients. Both models outperformed the UNI model. The proportion of excluded vs. included patients who died through one year was on average 27% vs. 6% (VCI), 31% vs. 7% (VC-Free), and 13% vs. 10% (UNI). Conclusions. The VC-Free model offers an alternative to the use of VC for eligibility determinations during the COVID-19 pandemic. The observation that the VC-Free model outperforms the use of VC in a broad ALS patient population suggests the use of prognostic strata in future, post-pandemic ALS clinical trial eligibility screening determinations.
Collapse
Affiliation(s)
| | - James D Berry
- Sean M. Healey & AMG Center for ALS and Neurological Clinical Research Institute, Massachusetts General Hospital, Boston, MA, USA
| | - Sabrina Paganoni
- Sean M. Healey & AMG Center for ALS and Neurological Clinical Research Institute, Massachusetts General Hospital, Boston, MA, USA
| | - Jonathan D Glass
- Department of Neurology, Emory University School of Medicine Atlanta, Atlanta, GA USA
| | - Christina Fournier
- Department of Neurology, Emory University School of Medicine Atlanta, Atlanta, GA USA
| | | | | | | |
Collapse
|
27
|
Alsulami K, Marston S. Small Molecules acting on Myofilaments as Treatments for Heart and Skeletal Muscle Diseases. Int J Mol Sci 2020; 21:E9599. [PMID: 33339418 PMCID: PMC7767104 DOI: 10.3390/ijms21249599] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/11/2020] [Accepted: 12/11/2020] [Indexed: 01/10/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM) are the most prevalent forms of the chronic and progressive pathological condition known as cardiomyopathy. These diseases have different aetiologies; however, they share the feature of haemodynamic abnormalities, which is mainly due to dysfunction in the contractile proteins that make up the contractile unit known as the sarcomere. To date, pharmacological treatment options are not disease-specific and rather focus on managing the symptoms, without addressing the disease mechanism. Earliest attempts at improving cardiac contractility by modulating the sarcomere indirectly (inotropes) resulted in unwanted effects. In contrast, targeting the sarcomere directly, aided by high-throughput screening systems, could identify small molecules with a superior therapeutic value in cardiac muscle disorders. Herein, an extensive literature review of 21 small molecules directed to five different targets was conducted. A simple scoring system was created to assess the suitability of small molecules for therapy by evaluating them in eight different criteria. Most of the compounds failed due to lack of target specificity or poor physicochemical properties. Six compounds stood out, showing a potential therapeutic value in HCM, DCM or heart failure (HF). Omecamtiv Mecarbil and Danicamtiv (myosin activators), Mavacamten, CK-274 and MYK-581 (myosin inhibitors) and AMG 594 (Ca2+-sensitiser) are all small molecules that allosterically modulate troponin or myosin. Omecamtiv Mecarbil showed limited efficacy in phase III GALACTIC-HF trial, while, results from phase III EXPLORER-HCM trial were recently published, indicating that Mavacamten reduced left ventricular outflow tract (LVOT) obstruction and diastolic dysfunction and improved the health status of patients with HCM. A novel category of small molecules known as "recouplers" was reported to target a phenomenon termed uncoupling commonly found in familial cardiomyopathies but has not progressed beyond preclinical work. In conclusion, the contractile apparatus is a promising target for new drug development.
Collapse
Affiliation(s)
- Khulud Alsulami
- Imperial Centre for Translational and Experimental Medicine, Cardiovascular Division, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK;
- National Centre for Pharmaceutical Technology, King Abdulaziz City for Science and Technology, Riyadh 11461, Saudi Arabia
| | - Steven Marston
- Imperial Centre for Translational and Experimental Medicine, Cardiovascular Division, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK;
| |
Collapse
|
28
|
Shefner JM, Andrews JA, Genge A, Jackson C, Lechtzin N, Miller TM, Cockroft BM, Meng L, Wei J, Wolff AA, Malik FI, Bodkin C, Brooks BR, Caress J, Dionne A, Fee D, Goutman SA, Goyal NA, Hardiman O, Hayat G, Heiman-Patterson T, Heitzman D, Henderson RD, Johnston W, Karam C, Kiernan MC, Kolb SJ, Korngut L, Ladha S, Matte G, Mora JS, Needham M, Oskarsson B, Pattee GL, Pioro EP, Pulley M, Quan D, Rezania K, Schellenberg KL, Schultz D, Shoesmith C, Simmons Z, Statland J, Sultan S, Swenson A, Berg LHVD, Vu T, Vucic S, Weiss M, Whyte-Rayson A, Wymer J, Zinman L, Rudnicki SA. A Phase 2, Double-Blind, Randomized, Dose-Ranging Trial Of Reldesemtiv In Patients With ALS. Amyotroph Lateral Scler Frontotemporal Degener 2020; 22:287-299. [PMID: 32969758 PMCID: PMC8117790 DOI: 10.1080/21678421.2020.1822410] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Objective: To evaluate safety, dose response, and preliminary efficacy of reldesemtiv over 12 weeks in patients with amyotrophic lateral sclerosis (ALS). Methods: Patients (≤2 years since diagnosis) with slow upright vital capacity (SVC) of ≥60% were randomized 1:1:1:1 to reldesemtiv 150, 300, or 450 mg twice daily (bid) or placebo; active treatment was 12 weeks with 4-week follow-up. Primary endpoint was change in percent predicted SVC at 12 weeks; secondary measures included ALS Functional Rating Scale-Revised (ALSFRS-R) and muscle strength mega-score. Results: Patients (N = 458) were enrolled; 85% completed 12-week treatment. The primary analysis failed to reach statistical significance (p = 0.11); secondary endpoints showed no statistically significant effects (ALSFRS-R, p = 0.09; muscle strength megascore, p = 0.31). Post hoc analyses pooling all active reldesemtiv-treated patients compared against placebo showed trends toward benefit in all endpoints (progression rate for SVC, ALSFRS-R, and muscle strength mega-score (nominal p values of 0.10, 0.01 and 0.20 respectively)). Reldesemtiv was well tolerated, with nausea and fatigue being the most common side effects. A dose-dependent decrease in estimated glomerular filtration rate was noted, and transaminase elevations were seen in approximately 5% of patients. Both hepatic and renal abnormalities trended toward resolution after study drug discontinuation. Conclusions: Although the primary efficacy analysis did not demonstrate statistical significance, there were trends favoring reldesemtiv for all three endpoints, with effect sizes generally regarded as clinically important. Tolerability was good; modest hepatic and renal abnormalities were reversible. The impact of reldesemtiv on patients with ALS should be assessed in a pivotal Phase 3 trial. (ClinicalTrials.gov Identifier: NCT03160898)
Collapse
Affiliation(s)
| | - Jinsy A Andrews
- The Eleanor and Lou Gehrig ALS Center, The Neurological Institute, New York, NY, USA
| | - Angela Genge
- Montreal Neurological Institute, Montreal, QC, Canada
| | | | - Noah Lechtzin
- Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | | | - Lisa Meng
- Cytokinetics, Inc, South San Francisco, CA, USA
| | - Jenny Wei
- Cytokinetics, Inc, South San Francisco, CA, USA
| | | | | | - Cynthia Bodkin
- Indiana University School of Medicine, Indianapolis, IN, USA
| | - Benjamin R Brooks
- Atrium Health Neurosciences Institute-Carolinas Neuromuscular/ALS MDA Care Center, Charlotte, NC, USA
| | - James Caress
- Wake Forest Health Sciences, Winston-Salem, NC, USA
| | | | - Dominic Fee
- Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Namita A Goyal
- The ALS & Neuromuscular Center, UCI Health, Orange, CA, USA
| | | | | | | | | | | | | | - Chafic Karam
- Oregon Health & Science University, Portland, OR, USA
| | - Matthew C Kiernan
- Royal Prince Alfred Hospital, University of Sydney, Sydney, Australia
| | - Stephen J Kolb
- Wexner Medical Center, Ohio State University, Columbus, OH, USA
| | | | - Shafeeq Ladha
- St. Joseph's Hospital and Medical Center, Neurological Institute, AZ, USA Barrow Phoenix
| | - Genevieve Matte
- Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
| | | | - Merrilee Needham
- Perron Institute, Department of Neurology, Fiona Stanley Hospital, The University of Notre Dame Australia, Murdoch University, Perth, Australia
| | | | | | | | | | - Dianna Quan
- University of Colorado Denver, Aurora, CO, USA
| | | | | | | | | | | | | | | | | | - Leonard H Van Den Berg
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Tuan Vu
- University of South Florida, Tampa, FL, USA
| | | | | | | | | | - Lorne Zinman
- ALS/Neuromuscular Clinic Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | | |
Collapse
|
29
|
Rutkove SB, Narayanaswami P, Berisha V, Liss J, Hahn S, Shelton K, Qi K, Pandeya S, Shefner JM. Improved ALS clinical trials through frequent at-home self-assessment: a proof of concept study. Ann Clin Transl Neurol 2020; 7:1148-1157. [PMID: 32515889 PMCID: PMC7359124 DOI: 10.1002/acn3.51096] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE To determine the potential for improving amyotrophic lateral sclerosis (ALS) clinical trials by having patients or caregivers perform frequent self-assessments at home. METHODS AND PARTICIPANTS We enrolled ALS patients into a nonblinded, longitudinal 9-month study in which patients and caregivers obtained daily data using several different instruments, including a slow-vital capacity device, a hand grip dynamometer, an electrical impedance myography-based fitness device, an activity tracker, a speech app, and the ALS functional rating scale-revised. Questions as to acceptability were asked at two time points. RESULTS A total of 113 individuals enrolled, with 61 (43 men, 18 women, mean age 60.1 ± 9.9 years) collecting a minimum of 7 days data and being included in the analysis. Daily measurements resulted in more accurate assessments of the slope of progression of the disease, resulting in smaller sample size estimates for a hypothetical clinical trial. For example, by performing daily slow-vital capacity measurements, calculated sample size was reduced to 182 subjects/study arm from 882/arm for monthly measurements. Similarly, performing the ALS functional rating scale weekly rather than monthly led to a calculated sample size of 73/arm as compared to 274/arm. Participants generally found the procedures acceptable and, for many, improved their sense of control of their disease. INTERPRETATION Frequent at-home measurements using standard tools holds the prospect of tracking progression and reducing sample size requirements for clinical trials in ALS while also being acceptable to the patients. Future studies in this and other neurological disorders should consider adopting this approach to data collection.
Collapse
Affiliation(s)
- Seward B Rutkove
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | | | - Julie Liss
- Arizona State University, Phoenix, AZ, USA
| | - Shira Hahn
- Arizona State University, Phoenix, AZ, USA
| | | | - Kristin Qi
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sarbesh Pandeya
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
30
|
Liscic RM, Alberici A, Cairns NJ, Romano M, Buratti E. From basic research to the clinic: innovative therapies for ALS and FTD in the pipeline. Mol Neurodegener 2020; 15:31. [PMID: 32487123 PMCID: PMC7268618 DOI: 10.1186/s13024-020-00373-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 03/27/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and Frontotemporal Degeneration (FTD) are neurodegenerative disorders, related by deterioration of motor and cognitive functions and short survival. Aside from cases with an inherited pathogenic mutation, the causes of the disorders are still largely unknown and no effective treatment currently exists. It has been shown that FTD may coexist with ALS and this overlap occurs at clinical, genetic, and molecular levels. In this work, we review the main pathological aspects of these complex diseases and discuss how the integration of the novel pathogenic molecular insights and the analysis of molecular interaction networks among all the genetic players represents a critical step to shed light on discovering novel therapeutic strategies and possibly tailoring personalized medicine approaches to specific ALS and FTD patients.
Collapse
Affiliation(s)
- Rajka Maria Liscic
- Department of Neurology, Johannes Kepler University, Linz, Austria
- School of Medicine, University of Osijek, Osijek, Croatia
| | - Antonella Alberici
- Neurology Unit, Department of Neurological Sciences and Vision, ASST-Spedali Civili-University of Brescia, Brescia, Italy
| | - Nigel John Cairns
- College of Medicine and Health and Living Systems Institute, University of Exeter, Exeter, UK
| | - Maurizio Romano
- Department of Life Sciences, Via Valerio 28, University of Trieste, 34127, Trieste, Italy
| | - Emanuele Buratti
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano 99, 34149, Trieste, Italy.
| |
Collapse
|
31
|
Filipi T, Hermanova Z, Tureckova J, Vanatko O, Anderova M. Glial Cells-The Strategic Targets in Amyotrophic Lateral Sclerosis Treatment. J Clin Med 2020; 9:E261. [PMID: 31963681 PMCID: PMC7020059 DOI: 10.3390/jcm9010261] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 01/13/2020] [Accepted: 01/16/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurological disease, which is characterized by the degeneration of motor neurons in the motor cortex and the spinal cord and subsequently by muscle atrophy. To date, numerous gene mutations have been linked to both sporadic and familial ALS, but the effort of many experimental groups to develop a suitable therapy has not, as of yet, proven successful. The original focus was on the degenerating motor neurons, when researchers tried to understand the pathological mechanisms that cause their slow death. However, it was soon discovered that ALS is a complicated and diverse pathology, where not only neurons, but also other cell types, play a crucial role via the so-called non-cell autonomous effect, which strongly deteriorates neuronal conditions. Subsequently, variable glia-based in vitro and in vivo models of ALS were established and used for brand-new experimental and clinical approaches. Such a shift towards glia soon bore its fruit in the form of several clinical studies, which more or less successfully tried to ward the unfavourable prognosis of ALS progression off. In this review, we aimed to summarize current knowledge regarding the involvement of each glial cell type in the progression of ALS, currently available treatments, and to provide an overview of diverse clinical trials covering pharmacological approaches, gene, and cell therapies.
Collapse
Affiliation(s)
- Tereza Filipi
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, 14200 Prague, Czech Republic; (T.F.); (Z.H.); (J.T.); (O.V.)
- 2nd Faculty of Medicine, Charles University, 15006 Prague, Czech Republic
| | - Zuzana Hermanova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, 14200 Prague, Czech Republic; (T.F.); (Z.H.); (J.T.); (O.V.)
- 2nd Faculty of Medicine, Charles University, 15006 Prague, Czech Republic
| | - Jana Tureckova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, 14200 Prague, Czech Republic; (T.F.); (Z.H.); (J.T.); (O.V.)
| | - Ondrej Vanatko
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, 14200 Prague, Czech Republic; (T.F.); (Z.H.); (J.T.); (O.V.)
| | - Miroslava Anderova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, 14200 Prague, Czech Republic; (T.F.); (Z.H.); (J.T.); (O.V.)
| |
Collapse
|
32
|
Lee EJ, Kolb J, Hwee DT, Malik FI, Granzier HL. Functional Characterization of the Intact Diaphragm in a Nebulin-Based Nemaline Myopathy (NM) Model-Effects of the Fast Skeletal Muscle Troponin Activator tirasemtiv. Int J Mol Sci 2019; 20:E5008. [PMID: 31658633 PMCID: PMC6829460 DOI: 10.3390/ijms20205008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/05/2019] [Accepted: 10/06/2019] [Indexed: 02/08/2023] Open
Abstract
Respiratory failure due to diaphragm dysfunction is considered a main cause of death in nemaline myopathy (NM) and we studied both isometric force and isotonic shortening of diaphragm muscle in a mouse model of nebulin-based NM (Neb cKO). A large contractile deficit was found in nebulin-deficient intact muscle that is frequency dependent, with the largest deficits at low-intermediate stimulation frequencies (e.g., a deficit of 72% at a stimulation frequency of 20 Hz). The effect of the fast skeletal muscle troponin activator (FSTA) tirasemtiv on force was examined. Tirasemtiv had a negligible effect at maximal stimulation frequencies, but greatly reduced the force deficit of the diaphragm at sub-maximal stimulation levels with an effect that was largest in Neb cKO diaphragm. As a result, the force deficit of Neb cKO diaphragm fell (from 72% to 29% at 20 Hz). Similar effects were found in in vivo experiments on the nerve-stimulated gastrocnemius muscle complex. Load-clamp experiments on diaphragm muscle showed that tirasemtiv increased the shortening velocity, and reduced the deficit in mechanical power by 33%. Thus, tirasemtiv significantly improves muscle function in a mouse model of nebulin-based nemaline myopathy.
Collapse
Affiliation(s)
- Eun-Jeong Lee
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, USA.
| | - Justin Kolb
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, USA.
| | - Darren T Hwee
- Research and Early Development, Cytokinetics, Inc., South San Francisco, CA 94080, USA.
| | - Fady I Malik
- Research and Early Development, Cytokinetics, Inc., South San Francisco, CA 94080, USA.
| | - Henk L Granzier
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, USA.
- Medical Research Building, RM 325, 1656 E Mabel St, Tucson, AZ 85721, USA.
| |
Collapse
|
33
|
Roesthuis L, van der Hoeven H, Sinderby C, Frenzel T, Ottenheijm C, Brochard L, Doorduin J, Heunks L. Effects of levosimendan on respiratory muscle function in patients weaning from mechanical ventilation. Intensive Care Med 2019; 45:1372-1381. [PMID: 31576436 PMCID: PMC6773912 DOI: 10.1007/s00134-019-05767-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 08/23/2019] [Indexed: 12/13/2022]
Abstract
PURPOSE Respiratory muscle weakness frequently develops in critically ill patients and is associated with adverse outcome, including difficult weaning from mechanical ventilation. Today, no drug is approved to improve respiratory muscle function in these patients. Previously, we have shown that the calcium sensitizer levosimendan improves calcium sensitivity of human diaphragm muscle fibers in vitro and contractile efficiency of the diaphragm in healthy subjects. The main purpose of this study is to investigate the effects of levosimendan on diaphragm contractile efficiency in mechanically ventilated patients. METHODS In a double-blind randomized placebo-controlled trial, mechanically ventilated patients performed two 30-min continuous positive airway pressure (CPAP) trials with 5-h interval. After the first CPAP trial, study medication (levosimendan 0.2 µg/kg/min continuous infusion or placebo) was administered. During the CPAP trials, electrical activity of the diaphragm (EAdi), transdiaphragmatic pressure (Pdi), and flow were measured. Neuromechanical efficiency (primary outcome parameter) was calculated. RESULTS Thirty-nine patients were included in the study. Neuromechanical efficiency was not different during the CPAP trial after levosimendan administration compared to the CPAP trial before study medication. Tidal volume and minute ventilation were higher after levosimendan administration (11 and 21%, respectively), whereas EAdi and Pdi were higher in both groups in the CPAP trial after study medication compared to the CPAP trial before study medication. CONCLUSIONS Levosimendan does not improve diaphragm contractile efficiency.
Collapse
Affiliation(s)
- Lisanne Roesthuis
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hans van der Hoeven
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Christer Sinderby
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada
- Institute for Biomedical Engineering and Science Technology (iBEST), Ryerson University and St. Michael's Hospital, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Tim Frenzel
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Coen Ottenheijm
- Department of Physiology, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Laurent Brochard
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, ON, Canada
| | - Jonne Doorduin
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Leo Heunks
- Department of Intensive Care Medicine, Amsterdam UMC, location VUmc, Postbox 7057, 1007 MB, Amsterdam, The Netherlands.
| |
Collapse
|
34
|
Allen DG. Calcium sensitivity and muscle disease. J Physiol 2019; 597:4435-4436. [DOI: 10.1113/jp278471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- David G. Allen
- School of Medical SciencesFaculty of MedicineUniversity of Sydney Australia
| |
Collapse
|