Safety, Tolerability, and Pharmacokinetics of SMT C1100, a 2-Arylbenzoxazole Utrophin Modulator, following Single- and Multiple-Dose Administration to Pediatric Patients with Duchenne Muscular Dystrophy

Valeria Ricotti, Stefan Spinty, Helen Roper, Imelda Hughes, Bina Tejura, Neil Robinson, Gary Layton, Kay Davies, Francesco Muntoni, Jonathon Tinsley, Valeria Ricotti, Stefan Spinty, Helen Roper, Imelda Hughes, Bina Tejura, Neil Robinson, Gary Layton, Kay Davies, Francesco Muntoni, Jonathon Tinsley

Abstract

Purpose: SMT C1100 is a utrophin modulator being evaluated as a treatment for Duchenne muscular dystrophy (DMD). This study, the first in pediatric DMD patients, reports the safety, tolerability and PK parameters of single and multiple doses of SMT C1100, as well as analyze potential biomarkers of muscle damage.

Methods: This multicenter, Phase 1 study enrolled 12 patients, divided equally into three groups (A-C). Group A were given 50 mg/kg on Days 1 and 11, and 50 mg/kg bid on Days 2 to 10. Group B and C received 100 mg/kg on Days 1 and 11; Group B and Group C were given 100 mg/kg bid and 100 mg/kg tid, respectively, on Days 2 to 10. A safety review was performed on all patients following the single dose and there was at least 2 weeks between each dose escalation, for safety and PK review. Adverse events (AEs) were monitored throughout the study.

Results: Most patients experienced mild AEs and there were no serious AEs. Two patients required analgesia for pain (headache, ear pain and toothache). One patient experienced moderate psychiatric AEs (abnormal behaviour and mood swings). Plasma concentrations of SMT C1100 at Days 1 and 11 indicated a high degree of patient variability regardless of dose. Unexpectedly the SMT C1100 levels were significantly lower than similar doses administered to healthy volunteers in an earlier clinical study. In general, individual baseline changes of creatine phosphokinase, alanine aminotransferase, aspartate aminotransferase levels fell with SMT C1100 dosing.

Conclusions: SMT C1100 was well tolerated in pediatric DMD patients.

Trial registration: ClinicalTrials.gov NCT02383511.

Conflict of interest statement

Competing Interests: This study was funded by Summit Therapeutics. JT and BT are employees of Summit Therapeutics. NR is employed by SHB Enterprises Limited. GL is employed by ParamStat Limited. NR and GL are paid consultants to Summit Therapeutics. KED is a shareholder in Summit Therapeutics. FM is involved as a Chief Investigator in a Summit Clinical trial of SMT C1100. He is also Chief Investigator of a Sarepta sponsored clinical trial; and Principal investigator of ISIS Pharmaceutics / Biogen; Roche; PTC and Prosensa/ Biomarin sponsored clinical trials. FM is a current member of the Pfizer Rare Disease Scientific Advisory Board and has provided ad hoc consultancies for Nicox; Italfarmaco; Ashaki, Summit Therapeutics, Roche and PTC Therapeutics. This does not alter the authors' adherence to all the PLOS ONE policies on sharing data and materials.

Figures

Fig 1. Study flow chart showing a…
Fig 1. Study flow chart showing a multiple ascending dose design.
Fig 2
Fig 2
Individual plasma concentrations of SMT C1100 for each of four patients in Groups A (black line), B (red line) and C (blue line). Group A patients received a single 50 mg/kg dose on Day 1 and on the morning of Day 11, with 50 mg/kg bid on Days 2 to 10. Group B and Group C patients received a single 100 mg/kg dose on Day 1 and on the morning of Day 11, with 100 mg/kg bid (Group B) or 100 mg/kg tid (Group C) on Days 2 to 10.
Fig 3. Plot of the average reduction…
Fig 3. Plot of the average reduction from baseline (estimate, square) of the following plasma markers: creatine phosphokinase (CPK), alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALK), gamma-glutamyl transferase (GGT), and albumen (ALB).
Upper and lower 95% confidence intervals for the following time points after dosing shown: Day 7, Day 12, and follow-up (3 days after completion of dosing).
Fig 4. Individual patient waterfall plots of…
Fig 4. Individual patient waterfall plots of gamma-glutamyl transferase (GGT), aspartate aminotransferase (AST), creatine phosphokinase (CPK), and alanine aminotransferase (ALT) showing changes from baseline after 10 days of dosing with SMT C1100.
Fig 5. Comparison of the average plasma…
Fig 5. Comparison of the average plasma levels of SMT C1100 from the 10 Duchenne muscular dystrophy pediatric patients (DMD fed) who had unexpectedly low exposure to SMT C1100 compared with the average plasma level of five healthy volunteers (HV) fasted prior to a single 200 mg/kg dose of SMT C1100.

References

    1. Emery AE. Population frequencies of inherited neuromuscular diseases–a world survey. Neuromuscul Disord. 1991; 1: 19–29.
    1. Van Deutekom JC, van Ommen GJ. Advances in Duchenne muscular dystrophy gene therapy. Nat Rev Genet 2003; 4: 774–784.
    1. Squire S, Raymackers JM, Vandebrouck C, Potter A, Tinsley J, Fischer R, et al. Prevention of pathology in mdx mice by expression of utrophin: analysis using an inducible transgenic expression system. Hum Mol Genet 2002; 11: 3333–3344.
    1. Tinsley J, Deconinck N, Fisher R, Kahn D, Phelps S, Gillis JM, et al. Expression of full-length utrophin prevents muscular dystrophy in mdx mice. Nat Med 1998; 4: 1441–1444.
    1. Chakkalakal JV, Stocksley MA, Harrison MA, Angus LM, Deschenes-Furry J, St-Pierre S, et al. Expression of utrophin A mRNA correlates with the oxidative capacity of skeletal muscle fiber types and is regulated by calcineurin/NFAT signaling. Proc Natl Acad Sci USA 2003; 100: 7791–7796.
    1. Dennis CL, Tinsley JM, Deconinck AE, Davies KE. Molecular and functional analysis of the utrophin promoter. Nucleic Acids Res 1996; 24: 1646–1652.
    1. Chancellor DR, Davies KE, De Moor O, Dorgan CR, Johnson PD, Lambert AG, et al. Discovery of 2-arylbenzoxazoles as upregulators of utrophin production for the treatment of Duchenne muscular dystrophy. J Med Chem 2011; 54: 3241–3250. 10.1021/jm200135z
    1. Tinsley JM, Fairclough RJ, Storer R, Wilkes FJ, Potter AC, Squire SE, et al. Daily treatment with SMTC1100, a novel small molecule utrophin upregulator, dramatically reduces the dystrophic symptoms in the mdx mouse. PLoS One 2011; 6: e19189 10.1371/journal.pone.0019189
    1. Tinsley J, Robinson N, Davies KE. Safety, tolerability, and pharmacokinetics of SMT C1100, a 2-arylbenzoxazole utrophin modulator, following single- and multiple-dose administration to healthy male adult volunteers. J Clin Pharmacol 2015; 55: 698–707. 10.1002/jcph.468
    1. Milic Rasic V, Vojinovic D, Pesovic J, Mijalkovic G, Lukic V, Mladenovic J, et al. Intellectual ability in the Duchenne muscular dystrophy and dystrophin gene mutation location. Balkan J Med Genet 2014; 17: 25–36. 10.2478/bjmg-2014-0071
    1. Ricotti V, Mandy WP, Scoto M, Pane M, Deconinck N, Messina S, et al. Neurodevelopmental, emotional, and behavioural problems in Duchenne muscular dystrophy in relation to underlying dystrophin gene mutations. Dev Med Child Neurol 2016; 58: 77–84. 10.1111/dmcn.12922
    1. Tanaka K, Takeshita K, Suganuma I, Kasagi S. Low serum cholic acid concentration of duchenne muscular dystrophy. Brain Devel 1983; 5: 511–513.
    1. Finkel RS, Flanigan KM, Wong B, Bönnemann C, Sampson J, Sweeney HL, et al. Phase 2a study of ataluren-mediated dystrophin production in patients with nonsense mutation Duchenne muscular dystrophy. PLoS One 2013; 8: e81302 10.1371/journal.pone.0081302
    1. Voit T, Topaloglu H, Straub V, Muntoni F, Deconinck N, Campion G, 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. 10.1016/S1474-4422(14)70195-4

Source: PubMed

3
購読する