Safety and Efficacy of Omaveloxolone in Friedreich Ataxia (MOXIe Study)

David R Lynch, Melanie P Chin, Martin B Delatycki, S H Subramony, Manuela Corti, J Chad Hoyle, Sylvia Boesch, Wolfgang Nachbauer, Caterina Mariotti, Katherine D Mathews, Paola Giunti, George Wilmot, Theresa Zesiewicz, Susan Perlman, Angie Goldsberry, Megan O'Grady, Colin J Meyer, David R Lynch, Melanie P Chin, Martin B Delatycki, S H Subramony, Manuela Corti, J Chad Hoyle, Sylvia Boesch, Wolfgang Nachbauer, Caterina Mariotti, Katherine D Mathews, Paola Giunti, George Wilmot, Theresa Zesiewicz, Susan Perlman, Angie Goldsberry, Megan O'Grady, Colin J Meyer

Abstract

Objective: Friedreich ataxia (FA) is a progressive genetic neurodegenerative disorder with no approved treatment. Omaveloxolone, an Nrf2 activator, improves mitochondrial function, restores redox balance, and reduces inflammation in models of FA. We investigated the safety and efficacy of omaveloxolone in patients with FA.

Methods: We conducted an international, double-blind, randomized, placebo-controlled, parallel-group, registrational phase 2 trial at 11 institutions in the United States, Europe, and Australia (NCT02255435, EudraCT2015-002762-23). Eligible patients, 16 to 40 years of age with genetically confirmed FA and baseline modified Friedreich's Ataxia Rating Scale (mFARS) scores between 20 and 80, were randomized 1:1 to placebo or 150mg per day of omaveloxolone. The primary outcome was change from baseline in the mFARS score in those treated with omaveloxolone compared with those on placebo at 48 weeks.

Results: One hundred fifty-five patients were screened, and 103 were randomly assigned to receive omaveloxolone (n = 51) or placebo (n = 52), with 40 omaveloxolone patients and 42 placebo patients analyzed in the full analysis set. Changes from baseline in mFARS scores in omaveloxolone (-1.55 ± 0.69) and placebo (0.85 ± 0.64) patients showed a difference between treatment groups of -2.40 ± 0.96 (p = 0.014). Transient reversible increases in aminotransferase levels were observed with omaveloxolone without increases in total bilirubin or other signs of liver injury. Headache, nausea, and fatigue were also more common among patients receiving omaveloxolone.

Interpretation: In the MOXIe trial, omaveloxolone significantly improved neurological function compared to placebo and was generally safe and well tolerated. It represents a potential therapeutic agent in FA. ANN NEUROL 2021;89:212-225.

Conflict of interest statement

This work was sponsored and funded by Reata Pharmaceuticals, which is developing omaveloxolone for clinical applications. M.P.C., C.J.M., M.O., and A.G. are employees of Reata Pharmaceuticals.

© 2020 The Authors. Annals of Neurology published by Wiley Periodicals LLC on behalf of American Neurological Association.

Figures

FIGURE 1
FIGURE 1
Study schema for the MOXIe Part 2 trial and CONSORT diagram. FARS = Friedreich's Ataxia Rating Scale; Scr=screening. [Color figure can be viewed at www.annalsofneurology.org]
FIGURE 2
FIGURE 2
(A) Mean changes from baseline in modified Friedrich's Ataxia Rating Scale (mFARS) score over time in the full analysis set (FAS) for patients randomized to omaveloxolone (n = 40) or placebo (n = 42). The change from baseline in mFARS and p value was estimated using mixed models repeated measures (MMRM) analysis. Significant differences in the change from baseline in mFARS in the omaveloxolone group, as compared with the placebo group, were observed at week 48 (p = 0.014). The error bars indicate standard errors. SEM = standard error of the mean. (B) Mean changes from baseline in the upright stability scores (Section E) of mFARS over time estimated using MMRM analysis. (C) Forest plot representing the difference between omaveloxolone and placebo treatment groups for the change from baseline in mFARS score at week 48 for the following prespecified analysis populations: FAS (n = 82), all randomized patients (ARP; n = 103), and prespecified subgroups. The change from baseline at week 48 was estimated using MMRM analysis, and each p value was estimated from a test comparing the difference in means between the omaveloxolone and placebo groups.
FIGURE 3
FIGURE 3
Post hoc analyses of change from baseline in modified Friedreich's Ataxia Rating Scale (mFARS) at week 48 with additional baseline covariates (full analysis set population). Data are presented as bar graphs comparing mean changes from baseline in mFARS at week 48 for patients randomized to omaveloxolone (Omav; n = 40) or placebo (n = 42) using the primary mixed model repeated measures (MMRM) methodology (A) or analysis of covariance (ANCOVA; B) with the inclusion of history of cardiomyopathy, GAA1 repeat length, or history of cardiomyopathy and GAA1 repeat length included as covariates. Note that the model with GAA1 repeat length as a covariate includes only those patients with baseline GAA1 repeat length data (n = 31 for omaveloxolone and n = 36 for placebo). [Color figure can be viewed at www.annalsofneurology.org]
FIGURE 4
FIGURE 4
(A–C) Mean (± standard error [SE]) alanine aminotransferase (ALT; A), aspartate aminotransferase (B), and total bilirubin values (C) for all randomized patients in the omaveloxolone (Omav; n = 51) or placebo (n = 52) groups through 48 weeks of treatment. Post‐treatment values collected at week 52, 4 weeks after the last dose of study drug was administered, are also shown. (D) eDISH (Evaluation of Drug‐Induced Serious Hepatotoxicity) plot. Vertical lines correspond to 3 × the upper limit of normal (ULN) for ALT. Horizontal lines correspond to 2 × ULN for total bilirubin. No patients met potential Hy's criteria in the upper‐right quadrant.
FIGURE 5
FIGURE 5
Data shown are mean (± standard error of the mean [SEM]) changes in serum ferritin (μg/l) and estimated glomerular filtration rate (eGFR; ml/min/1.73m2) over time for patients randomized to omaveloxolone or 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. Galea CA, Huq A, Lockhart PJ, et al. Compound heterozygous FXN mutations and clinical outcome in Friedreich ataxia. Ann Neurol 2016;79:485–495.
    1. Delatycki MB, Bidichandani SI. Friedreich ataxia—pathogenesis and implications for therapies. Neurobiol Dis 2019;132:104606.
    1. Martelli A, Puccio H. Dysregulation of cellular iron metabolism in Friedreich ataxia: from primary iron‐sulfur cluster deficit to mitochondrial iron accumulation. Front Pharmacol 2014;5:130.
    1. Tsou AY, Paulsen EK, Lagedrost SJ, et al. Mortality in Friedreich ataxia. J Neurol Sci 2011;307:46–49.
    1. Pousset F, Legrand L, Monin ML, et al. A 22‐year follow‐up study of long‐term cardiac outcome and predictors of survival in Friedreich ataxia. JAMA Neurol 2015;72:1334–1341.
    1. Strawser C, Schadt K, Hauser L, et al. Pharmacological therapeutics in Friedreich ataxia: the present state. Expert Rev Neurother 2017;17:895–907.
    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. 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. Shan Y, Schoenfeld RA, Hayashi G, et al. Frataxin deficiency leads to defects in expression 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. Anzovino A, Chiang S, Brown BE, et al. Molecular alterations in a mouse cardiac model of Friedreich ataxia: an impaired Nrf2 response mediated via upregulation of Keap1 and activation of the Gsk3beta axis. Am J Pathol 2017;187:2858–2875.
    1. Petrillo S, Piermarini E, Pastore A, et al. Nrf2‐inducers counteract neurodegeneration in frataxin‐silenced motor neurons: disclosing new therapeutic targets for Friedreich's ataxia. Int J Mol Sci 2017;18:2173.
    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. Dinkova‐Kostova AT, Liby KT, Stephenson KK, et al. Extremely potent triterpenoid inducers of the phase 2 response: correlations of protection against oxidant and inflammatory stress. Proc Natl Acad Sci U S A 2005;102:4584–4589.
    1. Abeti R, Baccaro A, Esteras N, Giunti P. Novel Nrf2‐inducer prevents mitochondrial defects and oxidative stress in friedreich's ataxia models. Frontiers in Cellular Neuroscience. 2018;12 10.3389/fncel.2018.00188.
    1. Lynch DR, Farmer J, Hauser L, et al. Safety, pharmacodynamics, and potential benefit of omaveloxolone in Friedreich ataxia. Ann Clin Transl Neurol 2019;6:15–26.
    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. 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. Rummey C, Farmer JM, Lynch DR. Predictors of loss of ambulation in Friedreich's ataxia. EClinicalMedicine. 2020;18:100213.
    1. Reetz K, Dogan I, Hohenfeld C, et al. Nonataxia symptoms in Friedreich ataxia: report from the registry of the European Friedreich's Ataxia Consortium for Translational Studies (EFACTS). Neurology 2018;91:e917–e930.
    1. Martinez AR, Moro A, Abrahao A, et al. Nonneurological involvement in late‐onset Friedreich ataxia (LOFA): exploring the phenotypes. Cerebellum 2017;16:253–256.
    1. Koutnikova H, Campuzano V, Foury F, Dollé P, Cazzalini O, Koenig M. Studies of human, mouse and yeast homologues indicate a mitochondrial function for frataxin. Nature Genetics. 1997;16:345–351. 10.1038/ng0897-345.
    1. Chandran V, Gao K, Swarup V, et al. Inducible and reversible phenotypes in a novel mouse model of Friedreich's ataxia. Elife 2017;6:e30054.
    1. Stuwe SH, Goetze O, Arning L, et al. Hepatic mitochondrial dysfunction in Friedreich ataxia. BMC Neurol 2011;11:145.
    1. Martelli A, Friedman LS, Reutenauer L, et al. Clinical data and characterization of the liver conditional mouse model exclude neoplasia as a non‐neurological manifestation associated with Friedreich's ataxia. Dis Model Mech 2012;5:860–869.
    1. Wang Q, Guo L, Strawser CJ, et al. Low apolipoprotein A‐I levels in Friedreich's ataxia and in frataxin‐deficient cells: implications for therapy. PLoS One 2018;13:e0192779.
    1. Miller GA, Bumeister R, Laidlaw J, et al. Bardoxolone methyl transcriptionally regulates transaminase levels and increases glutathione levels. Poster presented at: American Society of Nephrology 44th Annual Meeting; November 8–13, 2001; Philadelphia, PA.
    1. Lewis JH, Jadoul M, Block GA, et al. Effects of bardoxolone methyl on hepatic enzymes in patients with type 2 diabetes mellitus and stage 4 CKD. Clin Transl Sci (in press). 10.1111/cts.12868.
    1. Ellinger JJ, Lewis IA, Markley JL. Role of aminotransferases in glutamate metabolism of human erythrocytes. J Biomol NMR 2011;49:221–229.
    1. McGill MR. The past and present of serum aminotransferases and the future of liver injury biomarkers. EXCLI J 2016;15:817–828.
    1. Sookoian S, Pirola CJ. Liver enzymes, metabolomics and genome‐wide association studies: from systems biology to the personalized medicine. World J Gastroenterol 2015;21:711–725.

Source: PubMed

3
Iratkozz fel