Early phase 2 trial of TAS-205 in patients with Duchenne muscular dystrophy

Hirofumi Komaki, Yoshihiro Maegaki, Tsuyoshi Matsumura, Kazuhiro Shiraishi, Hiroyuki Awano, Akinori Nakamura, Satoru Kinoshita, Katsuhisa Ogata, Keiko Ishigaki, Shinji Saitoh, Michinori Funato, Satoshi Kuru, Takahiro Nakayama, Yasuyuki Iwata, Hiroyuki Yajima, Shin'ichi Takeda, Hirofumi Komaki, Yoshihiro Maegaki, Tsuyoshi Matsumura, Kazuhiro Shiraishi, Hiroyuki Awano, Akinori Nakamura, Satoru Kinoshita, Katsuhisa Ogata, Keiko Ishigaki, Shinji Saitoh, Michinori Funato, Satoshi Kuru, Takahiro Nakayama, Yasuyuki Iwata, Hiroyuki Yajima, Shin'ichi Takeda

Abstract

Objective: Duchenne muscular dystrophy (DMD) is a progressive muscular disease characterized by chronic cycles of inflammatory and necrotic processes. Prostaglandin D2 (PGD2 ) is produced by hematopoietic PGD synthase (HPGDS), which is pathologically implicated in muscle necrosis. This randomized, double-blind, placebo-controlled early phase 2 study (NCT02752048) aimed to assess the efficacy and safety of the novel selective HPGDS inhibitor, TAS-205, with exploratory measures in male DMD patients aged ≥5 years.

Methods: Patients were randomized 1:1:1 to receive low-dose TAS-205 (6.67-13.33 mg/kg/dose), high-dose TAS-205 (13.33-26.67 mg/kg/dose), or placebo. The primary endpoint was the change from baseline in a 6-minute walk distance (6MWD) at Week 24.

Results: Thirty-six patients were enrolled, of whom 35 patients were analysed for safety. The mean (standard error) changes from baseline to Week 24 in 6MWD were -17.0 (17.6) m in the placebo group (n = 10), -3.5 (20.3) m in the TAS-205 low-dose group (n = 11), and -7.5 (11.2) m in the TAS-205 high-dose group (n = 11). The mean (95% confidence interval) difference from the placebo group was 13.5 (-43.3 to 70.2) m in the TAS-205 low-dose group and 9.5 (-33.3 to 52.4) m in the TAS-205 high-dose group. No obvious differences were observed in the incidences of adverse events between treatment groups. No adverse drug reactions specific to TAS-205 treatment were observed.

Interpretation: The HPGDS inhibitor TAS-205 showed a favorable safety profile in DMD patients. Further research is required to examine the effectiveness of TAS-205 in a larger trial.

Conflict of interest statement

HK reports grants from Taiho Pharmaceutical Co., Ltd. during the conduct of the study. TN reports grants from National Centre of Neurology and Psychiatry (NCNP) and Japan Agency for Medical Research and Development (AMED); and personal fees from Taiho Pharmaceutical Co., Ltd. during the conduct of the study. YM, HA, YI, HY, ST, TM, KS, AN, SK, KO, KI, SS, MF, and SK have nothing to disclose in relation to this manuscript.

© 2020 The Authors. Annals of Clinical and Translational Neurology published by Wiley Periodicals, Inc on behalf of American Neurological Association.

Figures

Figure 1
Figure 1
Patient disposition.
Figure 2
Figure 2
Change in 6‐minute walk distance from baseline to Week 24. Bars indicate standard error.
Figure 3
Figure 3
Percent change in pharmacodynamic endpoints from baseline to Week 24; (A) urinary tPGDM/Cre concentration ratio, (B) total urinary tPGDM excretion, (C) urinary tPGEM/Cre concentration ratio and (D) total urinary tPGEM excretion. Bars indicate standard error. Abbreviations: tPGDM, tetranor prostaglandin D2 metabolite; Cre, creatinine; tPGEM, tetranor prostaglandin E2 metabolite.

References

    1. Hoffman E, Brown R, Kinkel L. Dystrophin: the protein product of the Duchenne muscular dystrophy locus. Cell 1987;51:919–928.
    1. Evans N, Misyak S, Robertson J, et al. Dysregulated intracellular signaling and inflammatory gene expression during initial disease onset in Duchenne muscular dystrophy. Am J Phys Med Rehabil 2009;88:502–522.
    1. Birnkrant D, Bushby K, Bann C, et al. Diagnosis and management of Duchenne muscular dystrophy, part 1: diagnosis, and neuromuscular, rehabilitation, endocrine, and gastrointestinal and nutritional management. Lancet Neurol 2018;17:251–267.
    1. Birnkrant D, Bushby K, Bann C, et al. Diagnosis and management of Duchenne muscular dystrophy, part 2: respiratory, cardiac, bone health, and orthopaedic management. Lancet Neurol 2018;17:347–361.
    1. Birnkrant D, Bushby K, Bann C, et al. Diagnosis and management of Duchenne muscular dystrophy, part 3: primary care, emergency management, psychosocial care, and transitions of care across the lifespan. Lancet Neurol 2018;17:445–455.
    1. McDonald C, Campbell C, Torricelli R, et al. Ataluren in patients with nonsense mutation Duchenne muscular dystrophy (ACT DMD): a multicentre, randomised, double‐blind, placebo‐controlled, phase 3 trial. Lancet 2017;390:1489–1498.
    1. Bushby K, Finkel R, Wong B, et al. Ataluren treatment of patients with nonsense mutation dystrophinopathy. Muscle Nerve 2014;50:477–487.
    1. Goemans N, Mercuri E, Belousova E, et al. A randomized placebo‐controlled phase 3 trial of an antisense oligonucleotide, drisapersen, in Duchenne muscular dystrophy. Neuromuscul Disord 2018;28:4–15.
    1. Voit T, Topaloglu H, Straub V, et al. Safety and efficacy of drisapersen for the treatment of Duchenne muscular dystrophy (DEMAND II): an exploratory, randomised, placebo‐controlled phase 2 study. Lancet Neurol 2014;13:987–996.
    1. Mendell J, Rodino‐Klapac L, Sahenk Z, et al. Eteplirsen for the treatment of Duchenne muscular dystrophy. Ann Neurol 2013;74:637–647.
    1. Falzarano M, Scotton C, Passarelli C, Ferlini A. Duchenne muscular dystrophy: from diagnosis to therapy. Molecules 2015;20:18168–18184.
    1. Verhaart IEC, Aartsma‐Rus A. Therapeutic developments for Duchenne muscular dystrophy. Nat Rev Neurol 2019;15:373–386.
    1. Ricciotti E, FitzGerald G. Prostaglandins and inflammation. Arterioscler Thromb Vasc Biol 2012;31:986–1000.
    1. Urade Y, Hayaishi O. Prostaglandin D synthase: structure and function. Vitam Horm 2000;58:89–120.
    1. Matsuoka T, Hirata M, Tanaka H, et al. Prostaglandin D2 as a mediator of allergic asthma. Science 2000;287:2013–2017.
    1. Mohri I, Aritake K, Taniguchi H, et al. Inhibition of prostaglandin D synthase suppresses muscular necrosis. Am J Pathol 2009;174:1735–744.
    1. Nakagawa T, Takeuchi A, Kakiuchi R, et al. A prostaglandin D2 metabolite is elevated in the urine of Duchenne muscular dystrophy patients and increases further from 8 years old. Clinica Chemica Acta 2013;423:10–14.
    1. Song W, Wang M, Ricciotti E, et al. Tetranor PGDM, an abundant urinary metabolite reflects biosynthesis of prostaglandin d2 in mice and humans. J Biol Chem 2008;283:1179–1188.
    1. Takeshita E, Komaki H, Tachimori H, et al. Urinary prostaglandin metabolites as Duchenne muscular dystrophy progression markers. Brain Dev 2018;40:918–925.
    1. Tanaka K, Aritake K, Tayama M, et al. Novel inhibitor of hematopoietic prostaglandin D synthase improves the muscle disorder in an experimental model of Duchenne muscular dystrophy. Neuromuscul Disord 2014;24:821 (Abstract).
    1. Takeshita E, Komaki H, Shimizu‐Motohashi Y, et al. A phase I study of TAS‐205 in patients with Duchenne muscular dystrophy. Ann Clin Transl Neurol 2018;5:1338–1349.
    1. Kuru S, Sakai M, Tanaka N, et al. Natural course of muscular involvement assessed by a new computed tomography method in Duchenne muscular dystrophy. Neurol Clin Neurosci 2013;1:63–68.
    1. Nakayama T, Kuru S, Okura M, et al. Estimation of net muscle volume in patients with muscular dystrophy using muscle CT for prospective muscle volume analysis: an observational study. BMJ Open 2013;3:e003603.
    1. McDonald C, Henricson E, Abresch R, et al. The 6‐minute walk test and other endpoints in Duchenne muscular dystrophy: longitudinal natural history observations over 48 weeks from a multicenter study. Muscle Nerve 2013;48:343–356.
    1. McDonald C, Henricson E, Abresch R, et al. The 6‐minute walk test and other clinical endpoints in Duchenne muscular dystrophy: reliability, concurrent validity, and minimal clinically important differences from a multicentre study. Muscle Nerve 2013;48:357–368.
    1. Haas M, Vlcek V, Balabanov P, et al. European medicines agency review of ataluren for the treatment of ambulant patients aged 5 years and older with Duchenne muscular dystrophy resulting from a nonsense mutation in the dystrophin gene. Neuromuscul Disord 2015;25:5–13.
    1. Drugs FDA. Drug approval package: Exondys 51 injection (eteplirsen). . (accessed Feb 12, 2017).
    1. Godi C, Ambrosi A, Nicastro F, et al. Longitudinal MRI quantification of muscle degeneration in Duchenne muscular dystrophy. Ann Clin Transl Neurol 2016;16:607–622.

Source: PubMed

3
Se inscrever