Safety, pharmacodynamics, and potential benefit of omaveloxolone in Friedreich ataxia

David R Lynch, Jennifer Farmer, Lauren Hauser, Ian A Blair, Qing Qing Wang, Clementina Mesaros, Nathaniel Snyder, Sylvia Boesch, Melanie Chin, Martin B Delatycki, Paola Giunti, Angela Goldsberry, Chad Hoyle, Michael G McBride, Wolfgang Nachbauer, Megan O'Grady, Susan Perlman, S H Subramony, George R Wilmot, Theresa Zesiewicz, Colin Meyer, David R Lynch, Jennifer Farmer, Lauren Hauser, Ian A Blair, Qing Qing Wang, Clementina Mesaros, Nathaniel Snyder, Sylvia Boesch, Melanie Chin, Martin B Delatycki, Paola Giunti, Angela Goldsberry, Chad Hoyle, Michael G McBride, Wolfgang Nachbauer, Megan O'Grady, Susan Perlman, S H Subramony, George R Wilmot, Theresa Zesiewicz, Colin Meyer

Abstract

Objective: Previous studies have demonstrated that suppression of Nrf2 in Friedreich ataxia tissues contributes to excess oxidative stress, mitochondrial dysfunction, and reduced ATP production. Omaveloxolone, an Nrf2 activator and NF-kB suppressor, targets dysfunctional inflammatory, metabolic, and bioenergetic pathways. The dose-ranging portion of this Phase 2 study assessed the safety, pharmacodynamics, and potential benefit of omaveloxolone in Friedreich ataxia patients (NCT02255435).

Methods: Sixty-nine Friedreich ataxia patients were randomized 3:1 to either omaveloxolone or placebo administered once daily for 12 weeks. Patients were randomized in cohorts of eight patients, at dose levels of 2.5-300 mg/day.

Results: Omaveloxolone was well tolerated, and adverse events were generally mild. Optimal pharmacodynamic changes (noted by changes in ferritin and GGT) were observed at doses of 80 and 160 mg/day. No significant changes were observed in the primary outcome, peak work load in maximal exercise testing (0.9 ± 2.9 W, placebo corrected). At the 160 mg/day dose, omaveloxolone improved the secondary outcome of the mFARS by 3.8 points versus baseline (P =0.0001) and by 2.3 points versus placebo (P =0.06). Omaveloxolone produced greater improvements in mFARS in patients that did not have musculoskeletal foot deformity (pes cavus). In patients without this foot deformity, omaveloxolone improved mFARS by 6.0 points from baseline (P <0.0001) and by 4.4 points versus placebo (P =0.01) at the 160 mg/day.

Interpretation: Treatment of Friedreich ataxia patients with omaveloxolone at the optimal dose level of 160 mg/day appears to improve neurological function. Therefore, omaveloxolone treatment is being examined in greater detail at 150 mg/day for Friedreich ataxia.

Figures

Figure 1
Figure 1
Consort diagram of MOXie, part 1.
Figure 2
Figure 2
Pharmacokinetics of Omav. Maximal concentration of Omaveloxolone Cmax levels are shown at different doses. Data are presented as a Box and whisker plot. Plasma concentrations increased exponentially over the dose range of the study.
Figure 3
Figure 3
Pharmacodynamic effects of Omav. Omav had dose dependent effects on Ferritin (A), GGT (B), AST (C) and creatine kinase (D). In general, effects of Omav increased through doses of 180 mg, then were blunted at the highest dose (300 mg).
Figure 4
Figure 4
Platelet isotopologue analysis. Isotopic incorporation from [13C6] glucose (A) and [13C16] palmitate (B) to HMG‐CoA (%) was determined in subjects at different dose of Omav. Cohorts were pooled into placebo (= 3), cohorts 1 and 2 (n = 8) and cohorts 3‐8 (n = 13) for analysis due to the small number of participants in each individual cohort.
Figure 5
Figure 5
Effect of Omav on mFARS exam results. Omav produced a dose dependent improvement in mFARS score. The difference was more apparent at the higher doses in the study, with less benefit at 300 mg, consistent with AST, ferritin, GGT and CK changes at 300 mg.
Figure 6
Figure 6
Pes cavus is associated with less response to Omav. The magnitude of improvement from Omav was higher on the mFARS exam in subjects without pes cavus compared with those with pes cavus. Similarly, a benefit of Omav on cardiac exercise stress testing was noted in the subgroup without pes cavus, whereas there was minimal effect in the overall cohort. Without pes cavus: n = 30 Omav, n = 7 placebo. With pes cavus: n = 22 Omav, n = 10 placebo

References

    1. Lynch DR, Farmer JM, Balcer LJ, Wilson RB. Friedreich ataxia: effects of genetic understanding on clinical evaluation and therapy. Arch Neurol 2002;59:743–747.
    1. Pandolfo M. Friedreich ataxia. Arch Neurol 2008;65:1296–1303.
    1. Schulz JB, Boesch S, Bürk K, et al. Diagnosis and treatment of Friedreich ataxia: a European perspective. Nat Rev Neurol 2009;5:222–234.
    1. Delatycki MB, Corben LA. Clinical features of Friedreich ataxia. J Child Neurol 2012;27:1133–1137.
    1. Patel M, Isaacs CJ, Seyer L, et al. Progression of Friedreich ataxia: quantitative characterization over 5 years. Ann Clin Transl Neurol 2016;3:684–694.
    1. Reetz K, Dogan I, Hilgers RD, et al.; EFACTS Study Group . Progression characteristics of the European Friedreich's ataxia Consortium for Translational Studies (EFACTS): a 2 year cohort study. Lancet Neurol 2016; 15:1346–1354.
    1. Sahdeo S, Scott BD, McMackin MZ, et al. Dyclonine rescues frataxin deficiency in animal models and buccal cells of patients with Friedreich's ataxia. Hum Mol Genet 2014;23:6848–6862.
    1. Shan Y, Schoenfeld RA, Hayashi G, et al. Frataxin deficiency leads to defects inexpression of antioxidants and Nrf2 expression in dorsal root ganglia of the Friedreich's ataxia YG8R mouse model. Antioxid Redox Signal 2013;19:1481–1493.
    1. Paupe V, Dassa EP, Goncalves S, et al. Impaired nuclear Nrf2 translocation undermines the oxidative stress response in Friedreich ataxia. PLoS ONE 2009;4:e4253.
    1. D'Oria V, Petrini S, Travaglini L, et al. Frataxin deficiency leads to reduced expression and impaired translocation of NF‐E2‐related factor (Nrf2) in cultured motor neurons. Int J Mol Sci 2013;14:7853–7865.
    1. Reisman SA, Lee CY, Meyer CJ, et al. Topical application of the synthetic triterpenoid RTA 408 activates Nrf2 and induces cytoprotective genes in rat skin. Arch Dermatol Res 2014;306:447–454.
    1. Abeti R, Uzun E, Renganathan I, et al. Targeting lipid peroxidation and mitochondrial imbalance in Friedreich's ataxia. Pharmacol Res 2015;99:344–350.
    1. Hayashi G, Cortopassi G. Lymphoblast oxidative stress genes as potential biomarkers of disease severity and drug effect in Friedreich's ataxia. PLoS ONE 2016;11:e0153574.
    1. Abeti R, Baccaro A, Esteras N, Giunti P. Novel Nrf2‐inducer prevents mitochondrial defects and oxidative stress in Friedreich's ataxia models. Front Cell Neurosci 2018;12:188.
    1. Regner SR, Wilcox NS, Friedman LS, et al. Friedreich ataxia clinical outcome measures: natural history evaluation in 410 participants. J Child Neurol 2012;27:1152–1158.
    1. Worth AJ, Basu SS, Deutsch EC, et al. Stable isotopes and LC‐MS for monitoring metabolic disturbances in Friedreich's ataxia platelets. Bioanalysis 2015;7:1843–1855.
    1. Friedman LS, Farmer JM, Perlman S, et al. Measuring the rate of progression in Friedreich ataxia: implications for clinical trial design. Mov Disord 2010;25:426–432.
    1. Lynch DR, Farmer JM, Tsou AY, et al. Measuring Friedreich ataxia: complementary features of examination and performance measures. Neurology 2006;66:1711–1716.
    1. Basu SS, Deutsch EC, Schmaier AA, et al. Human platelets as a platform to monitor metabolic biomarkers using stable isotopes and LC‐MS. Bioanalysis 2013;5:3009–3021.
    1. Purkins L, Love ER, Eve MD, et al. The influence of diet upon liver function tests and serum lipids in healthy male volunteers resident in a Phase I unit. Br J Clin Pharmacol 2004;57:199–208.
    1. Miller GA, Bumeister R, Laidlaw J, et al. Bardoxolone methyl transcriptionally regulates transaminase levels and increases glutathione levels. Am Soc Nephrol 2011;2011:PO2086.
    1. Kerins MJ, Ooi A. The roles of NRF2 in modulating cellular iron homeostasis. Antioxid Redox Signal 2018; 10.1089/ars.2017.7176.
    1. MacKenzie EL, Ray PD, Tsuji Y. Role and regulation of ferritin H in rotenone‐mediated mitochondrial oxidative stress. Free Radic Biol Med 2008;44:1762–1771.
    1. Ravuri C, Svineng G, Huseby NE. Differential regulation of γ‐glutamyl transferase and glutamate cysteine ligase expression after mitochondrial uncoupling: γ‐glutamyltransferase is regulated in an Nrf2‐ and NFκB‐independent manner. Free Radic Res 2013;47:394–403.
    1. Zhang H, Liu H, Dickinson DA, et al. gamma‐Glutamyl transpeptidase is induced by 4‐hydroxynonenal via EpRE/Nrf2 signaling in rat epithelial type II cells. Free Radic Biol Med 2006;40:1281–1292.
    1. Wilson RB, Lynch DR, Fischbeck KH. Normal serum iron and ferritin concentrations in patients with Friedreich's ataxia. Ann Neurol 1998;44:132–134.
    1. Wilson RB, Lynch DR, Farmer JM, et al. Increased serum transferrin receptor concentrations in Friedreich ataxia. Ann Neurol 2000;47:659–661.
    1. Phillips JT, Fox RJ. BG‐12 in multiple sclerosis. Semin Neurol 2013;33:56–65.
    1. Yiu EM, Tai G, Peverill RE, et al. An open‐label trial in Friedreich ataxia suggests clinical benefit with high‐dose resveratrol, without effect on frataxin levels. J Neurol 2015;262:1344–1353.
    1. Seyer L, Greeley N, Foerster D, et al. Open‐label pilot study of interferon gamma‐1b in Friedreich ataxia. Acta Neurol Scand 2015;132:7–15.
    1. Pandolfo M, Arpa J, Delatycki MB, et al. Deferiprone in Friedreich ataxia: a 6‐month randomized controlled trial. Ann Neurol 2014;76:509–521.
    1. Lynch DR, Willi SM, Wilson RB, et al. A0001 in Friedreich ataxia: biochemical characterization and effects in a clinical trial. Mov Disord 2012;27:1026–1033.
    1. Mariotti C, Fancellu R, Caldarazzo S, et al. Erythropoietin in Friedreich ataxia: no effect on frataxin in a randomized controlled trial. Mov Disord 2012;27:446–449.
    1. Lynch DR, Perlman SL, Meier T. A phase 3, double‐blind, placebo‐controlled trial of idebenone in friedreich ataxia. Arch Neurol 2010;67:941–947.
    1. Schulz JB, Di Prospero NA, Fischbeck K. Clinical experience with high‐dose idebenone in Friedreich ataxia. J Neurol 2009;256:42–45.
    1. Boesch S, Sturm B, Hering S, et al. Neurological effects of recombinant human erythropoietin in Friedreich's ataxia: a clinical pilot trial. Mov Disord 2008;23:1940–1944.

Source: PubMed

3
Sottoscrivi