Ibudilast (MN-166) in amyotrophic lateral sclerosis- an open label, safety and pharmacodynamic trial

Suma Babu, Baileigh G Hightower, James Chan, Nicole R Zürcher, Pia Kivisäkk, Chieh-En J Tseng, Danica L Sanders, Ashley Robichaud, Haruhiko Banno, Armineuza Evora, Akshata Ashokkumar, Lindsay Pothier, Sabrina Paganoni, Sheena Chew, Joanna Dojillo, Kazuko Matsuda, Mark Gudesblatt, James D Berry, Merit E Cudkowicz, Jacob M Hooker, Nazem Atassi, Suma Babu, Baileigh G Hightower, James Chan, Nicole R Zürcher, Pia Kivisäkk, Chieh-En J Tseng, Danica L Sanders, Ashley Robichaud, Haruhiko Banno, Armineuza Evora, Akshata Ashokkumar, Lindsay Pothier, Sabrina Paganoni, Sheena Chew, Joanna Dojillo, Kazuko Matsuda, Mark Gudesblatt, James D Berry, Merit E Cudkowicz, Jacob M Hooker, Nazem Atassi

Abstract

Ibudilast (MN-166) is an inhibitor of macrophage migration inhibitory factor (MIF) and phosphodiesterases 3,4,10 and 11 (Gibson et al., 2006; Cho et al., 2010). Ibudilast attenuates CNS microglial activation and secretion of pro-inflammatory cytokines (Fujimoto et al., 1999; Cho et al., 2010). In vitro evidence suggests that ibudilast is neuroprotective by suppressing neuronal cell death induced by microglial activation. People with ALS have increased microglial activation measured by [11C]PBR28-PET in the motor cortices. The primary objective is to determine the impact of ibudilast on reducing glial activation and neuroaxonal loss in ALS, measured by PBR28-PET and serum Neurofilament light (NfL). The secondary objectives included determining safety and tolerability of ibudilast high dosage (up to 100 mg/day) over 36 weeks. In this open label trial, 35 eligible ALS participants underwent ibudilast treatment up to 100 mg/day for 36 weeks. Of these, 30 participants were enrolled in the main study cohort and were included in biomarker, safety and tolerability analyses. Five additional participants were enrolled in the expanded access arm, who did not meet imaging eligibility criteria and were included in the safety and tolerability analyses. The primary endpoints were median change from baseline in (a) PBR28-PET uptake in primary motor cortices, measured by standard uptake value ratio (SUVR) over 12-24 weeks and (b) serum NfL over 36-40 weeks. The secondary safety and tolerability endpoints were collected through Week 40. The baseline median (range) of PBR28-PET SUVR was 1.033 (0.847, 1.170) and NfL was 60.3 (33.1, 219.3) pg/ml. Participants who completed both pre and post-treatment scans had PBR28-PET SUVR median(range) change from baseline of 0.002 (-0.184, 0.156) , P = 0.5 (n = 22). The median(range) NfL change from baseline was 0.4 pg/ml (-1.8, 17.5), P = 0.2 (n = 10 participants). 30(86%) participants experienced at least one, possibly study drug related adverse event. 13(37%) participants could not tolerate 100 mg/day and underwent dose reduction to 60-80 mg/day and 11(31%) participants discontinued study drug early due to drug related adverse events. The study concludes that following treatment with ibudilast up to 100 mg/day in ALS participants, there were no significant reductions in (a) motor cortical glial activation measured by PBR28-PET SUVR over 12-24 weeks or (b) CNS neuroaxonal loss, measured by serum NfL over 36-40 weeks. Dose reductions and discontinuations due to treatment emergent adverse events were common at this dosage in ALS participants. Future pharmacokinetic and dose-finding studies of ibudilast would help better understand tolerability and target engagement in ALS.

Trial registration: ClinicalTrials.gov NCT02714036.

Keywords: Biomarker endpoint; Clinical trial; Ibudilast; MIF; Neurofilament; PBR28; Phase 1b; TNF-alpha.

Conflict of interest statement

Nazem Atassi is an employee of Sanofi.

Baileigh Hightower, Pia Kivisäkk, Nicole R Zürcher, Chieh-En J Tseng, Danica L Sanders, Ashley Robichaud, Armineuza Evora, Akshata Ashokkumar and Lindsay Pothier report no COI related to this trial.

Suma Babu reports research support from American Academy of Neurology, AANEM Foundation, The ALS Association., Muscular Dystrophy Association, Biogen Inc, Orion Corporation, Voyager Therapeutics and Novartis pharmaceuticals.

James D. Berry has attended advisory boards at Alexion, Biogen and Clene Nanomedicine, has received research support from Alexion, Amylyx Therapeutics, Biogen, MT Pharma of America, Anelixis Therapeutics, Brainstorm Cell Therapeutics, Genentech, nQ Medical, ALS Finding a Cure, NINDS, Muscular Dystrophy Association, and ALS One.

Sabrina Paganoni reports research grants from Amylyx Therapeutics, Revalesio Corporation, Ra Pharma/UCB, Biohaven Pharmaceuticals, Clene Nanomedicine, Prilenia Therapeutics, The ALS Association, the American Academy of Neurology, ALS Finding a Cure, the Salah Foundation, the Spastic Paraplegia Foundation and reports personal consulting fees for advisory panels from Orion Corporation.

Sheena Chew is a full-time employee of Biogen and reports research support from the American Academy of Neurology and Biogen Inc.

Haruhiko Banno reports research grant from Time Therapeutics and scientific advising fee from Sumitomo Dainippon Pharma.

Jacob Hooker holds equity or stock options and/or has received income from Eikonizo Therapetics, Psy Therapeutics, Fuzionaire Diagnostics, and Delix Therapeutics.

Merit Cudkowicz reports consulting fees from Biogen, Takeda, Sunovion, Cytokinetics, Immunitypharm, and Wave.

Kazuko Matsuda is an employee of MediciNova.

Joanna Dojillo is an employee of MediciNova.

Copyright © 2021 The Author(s). Published by Elsevier Inc. All rights reserved.

Figures

Fig. 1
Fig. 1
Consort diagram Some participants had more than one reason for screen failure.
Fig. 2
Fig. 2
[11C]PBR28-PET uptake in bilateral motor cortices for Ibudilast pre-treatment and post-treatment groups (n = 22 participants). [A] Group median [11C]PBR28 SUVRocc for pre-treatment (top) and 12–24 weeks from baseline (bottom) groups. The median pre-treatment SUVRocc in the motor cortices was 1.033 (range 0.847, 1.170). All images are projected onto standard MNI space at coordinates (x = −8, y =  − 20, z = +64). The color bar represents the group median [11C]PBR28 SUVRocc values in the motor cortices. [B] The box plots show that there were no significant changes from baseline observed in median [11C]PBR28 SUVRocc in the motor cortices at 12–24 weeks of ibudilast treatment [n = 22, median 0.002 (range −0.184, 0.156), Wilcoxon-Signed-Rank-Test V = 149, p = 0.5] [C] The spaghetti plot represents individual changes from baseline in median [11C]PBR28 SUVRocc in the motor cortices following 12–24 weeks of ibudilast treatment.
Fig. 3
Fig. 3
[Fig. 3A]: Baseline serum NfL was 60.6 (range 33.1, 219.3) pg/ml. The box plots show median serum NfL levels at (a) baseline (b) 24 weeks of ibudilast treatment [n = 10 participants, median(range) change 1.7 pg/ml (−2.7, 27.3), Wilcoxon-Signed-Rank-Test V = 41, p = 0.19] and (c) 36–40 weeks of ibudilast treatment [n = 10 participants, median (range) change 0.4 pg/ml (−1.8, 17.5), Wilcoxon-Signed-Rank-Test V = 40, p = 0.23] and. [Fig. 3B] The spaghetti plot represents individual changes from baseline in median serum NfL following 36–40 weeks of ibudilast treatment.

References

    1. Albrecht D.S., Normandin M.D., Shcherbinin S., Wooten D.W., Schwarz A.J., Zurcher N.R. Pseudoreference regions for glial imaging with (11)C-PBR28: investigation in 2 clinical cohorts. J. Nucl. Med. 2018;59(1):107–114.
    1. Alshikho M.J., Zurcher N.R., Loggia M.L., Cernasov P., Chonde D.B., Izquierdo Garcia D. Glial activation colocalizes with structural abnormalities in amyotrophic lateral sclerosis. Neurology. 2016;87(24):2554–2561.
    1. Alshikho M.J., Zurcher N.R., Loggia M.L., Cernasov P., Reynolds B., Pijanowski O. Integrated magnetic resonance imaging and [(11) C]-PBR28 positron emission tomographic imaging in amyotrophic lateral sclerosis. Ann. Neurol. 2018;83(6):1186–1197.
    1. Babu S., Macklin E.A., Jackson K.E., Simpson E., Mahoney K., Yu H. Selection design phase II trial of high dosages of tamoxifen and creatine in amyotrophic lateral sclerosis. Amyotroph Lateral Scler. Frontotemporal Degener. 2020;21(1–2):15–23.
    1. Bakkar N., Boehringer A., Bowser R. Use of biomarkers in ALS drug development and clinical trials. Brain Res. 2015;14(1607):94–107.
    1. Barkhof F., Hulst H.E., Drulovic J., Uitdehaag B.M., Matsuda K., Landin R. Ibudilast in relapsing-remitting multiple sclerosis: a neuroprotectant? Neurology. 2010;74(13):1033–1040.
    1. Brettschneider J., Toledo J.B., Van Deerlin V.M., Elman L., McCluskey L., Lee V.M. Microglial activation correlates with disease progression and upper motor neuron clinical symptoms in amyotrophic lateral sclerosis. PLoS ONE. 2012;7(6)
    1. Bridel C, van Wieringen WN, Zetterberg H, Tijms BM, Teunissen CE, and the NFL Group, et al. Diagnostic Value of Cerebrospinal Fluid Neurofilament Light Protein in Neurology: A Systematic Review and Meta-analysis. JAMA Neurol 2019 Jun 17.
    1. Brooks B.R., Miller R.G., Swash M., Munsat T.L. World federation of neurology research group on motor neuron diseases. El Escorial revisited: revised criteria for the diagnosis of amyotrophic lateral sclerosis. Amyotroph. Lateral Scler. Other Motor Neuron Disord. 2000;1(5):293–299.
    1. Brooks B.R., Bravver E.K., Sanjak M.S., Bockenek W.L., Lindblom S.S., Lary C.L., Ranzinger L.H., Sturdivant A.N., Langford V.L., Holsten S.E., Ward A.L., Hillberry R., Wright A., Williamson T.A., Linville A.N., Lucas N.M., Brandon N.B., Dojillo J., Matsuda K., Iwaki Y., Graves D.C. Novel composite endpoint extended analysis during extension of Ibudilast Phase 1a/2b clinical trial better predicts post-wash-out survival. Platform Commun. Amyotroph. Lateral Scler. Frontotemporal Degener. 2018;19(sup1):1–84. doi: 10.1080/21678421.2018.1510202.
    1. Canto E, Barro C, Zhao C, Caillier SJ, Michalak Z, Bove R, et al. Association Between Serum Neurofilament Light Chain Levels and Long-term Disease Course Among Patients With Multiple Sclerosis Followed up for 12 Years. JAMA Neurol 2019 Aug 12.
    1. Cedarbaum J.M., Stambler N., Malta E., Fuller C., Hilt D., Thurmond B. The ALSFRS-R: a revised ALS functional rating scale that incorporates assessments of respiratory function. BDNF ALS Study Group (Phase III) J. Neurol. Sci. 1999;169(1–2):13–21.
    1. Chen Y., Wang H., Ying Z., Gao Q. Ibudilast enhances the clearance of SOD1 and TDP-43 aggregates through TFEB-mediated autophagy and lysosomal biogenesis: The new molecular mechanism of ibudilast and its implication for neuroprotective therapy. Biochem. Biophys. Res. Commun. 2020;526(1):231–238.
    1. Cho Y., Crichlow G.V., Vermeire J.J., Leng L., Du X., Hodsdon M.E. Allosteric inhibition of macrophage migration inhibitory factor revealed by ibudilast. Proc. Natl. Acad. Sci. USA. 2010;107(25):11313–11318.
    1. Coalition Adaptive Platform Trials. Adaptive platform trials: definition, design, conduct and reporting considerations. Nat Rev Drug Discov. 2019;18(10):797–807.
    1. Cox G.M., Kithcart A.P., Pitt D., Guan Z., Alexander J., Williams J.L. Macrophage migration inhibitory factor potentiates autoimmune-mediated neuroinflammation. J. Immunol. 2013;191(3):1043–1054.
    1. DeYoung D.Z., Heinzerling K.G., Swanson A.N., Tsuang J., Furst B.A., Yi Y. Safety of intravenous methamphetamine administration during ibudilast treatment. J. Clin. Psychopharmacol. 2016;36(4):347–354.
    1. Downer OM, Marcus REG, Zurcher NR, Hooker JM. Tracing the History of the Human Translocator Protein to Recent Neurodegenerative and Psychiatric Imaging. ACS Chem Neurosci 2020 Jul 23.
    1. Fox R.J., Coffey C.S., Conwit R., Cudkowicz M.E., Gleason T., Goodman A. Phase 2 Trial of Ibudilast in Progressive Multiple Sclerosis. N. Engl. J. Med. 2018;379(9):846–855.
    1. Fujimoto T., Sakoda S., Fujimura H., Yanagihara T. Ibudilast, a phosphodiesterase inhibitor, ameliorates experimental autoimmune encephalomyelitis in Dark August rats. J. Neuroimmunol. 1999;95(1–2):35–42.
    1. Fujita M., Imaizumi M., Zoghbi S.S., Fujimura Y., Farris A.G., Suhara T. Kinetic analysis in healthy humans of a novel positron emission tomography radioligand to image the peripheral benzodiazepine receptor, a potential biomarker for inflammation. Neuroimage. 2008;40(1):43–52.
    1. Gibson L.C., Hastings S.F., McPhee I., Clayton R.A., Darroch C.E., Mackenzie A. The inhibitory profile of Ibudilast against the human phosphodiesterase enzyme family. Eur. J. Pharmacol. 2006;538(1–3):39–42.
    1. Ha W, Sevim-Nalkiran H, Zaman AM, Matsuda K, Khasraw M, Nowak AK, et al. Ibudilast sensitizes glioblastoma to temozolomide by targeting Macrophage Migration Inhibitory Factor (MIF). Sci Rep 2019 Feb 27;9(1):2905-019-39427-4.
    1. Huang F, Zhu Y, Hsiao-Nakamoto J, Tang X, Dugas JC, Moscovitch-Lopatin M, et al. Longitudinal biomarkers in amyotrophic lateral sclerosis. Ann Clin Transl Neurol 2020 Jun 9.
    1. Jucaite A., Svenningsson P., Rinne J.O., Cselenyi Z., Varnas K., Johnstrom P. Effect of the myeloperoxidase inhibitor AZD3241 on microglia: a PET study in Parkinson's disease. Brain. 2015;138(Pt 9):2687–2700.
    1. Kaufmann P., Thompson J.L., Levy G., Buchsbaum R., Shefner J., Krivickas L.S. Phase II trial of CoQ10 for ALS finds insufficient evidence to justify phase III. Ann. Neurol. 2009;66(2):235–244.
    1. Keshavan A., Heslegrave A., Zetterberg H., Schott J.M. Stability of blood-based biomarkers of Alzheimer's disease over multiple freeze-thaw cycles. Alzheimers Dement (Amst) 2018;2(10):448–451.
    1. Knudson R.J., Lebowitz M.D., Holberg C.J., Burrows B. Changes in the normal maximal expiratory flow-volume curve with growth and aging. Am. Rev. Respir. Dis. 1983;127(6):725–734.
    1. Kuhle J., Barro C., Andreasson U., Derfuss T., Lindberg R., Sandelius A. Comparison of three analytical platforms for quantification of the neurofilament light chain in blood samples: ELISA, electrochemiluminescence immunoassay and Simoa. Clin. Chem. Lab. Med. 2016;54(10):1655–1661.
    1. Labra J., Menon P., Byth K., Morrison S., Vucic S. Rate of disease progression: a prognostic biomarker in ALS. J. Neurol. Neurosurg. Psychiatry. 2016;87(6):628–632.
    1. Lu C.H., Macdonald-Wallis C., Gray E., Pearce N., Petzold A., Norgren N. Neurofilament light chain: A prognostic biomarker in amyotrophic lateral sclerosis. Neurology. 2015;84(22):2247–2257.
    1. Miller T., Cudkowicz M., Shaw P.J., Andersen P.M., Atassi N., Bucelli R.C. Phase 1–2 Trial of Antisense Oligonucleotide Tofersen for SOD1 ALS. N. Engl. J. Med. 2020;383(2):109–119.
    1. Mizrahi R., Rusjan P.M., Kennedy J., Pollock B., Mulsant B., Suridjan I. Translocator protein (18 kDa) polymorphism (rs6971) explains in-vivo brain binding affinity of the PET radioligand [(18)F]-FEPPA. J. Cereb. Blood Flow Metab. 2012;32(6):968–972.
    1. Mizuno T., Kurotani T., Komatsu Y., Kawanokuchi J., Kato H., Mitsuma N. Neuroprotective role of phosphodiesterase inhibitor ibudilast on neuronal cell death induced by activated microglia. Neuropharmacology. 2004;46(3):404–411.
    1. Novakova L., Zetterberg H., Sundstrom P., Axelsson M., Khademi M., Gunnarsson M. Monitoring disease activity in multiple sclerosis using serum neurofilament light protein. Neurology. 2017;89(22):2230–2237.
    1. Owen D.R., Yeo A.J., Gunn R.N., Song K., Wadsworth G., Lewis A. An 18-kDa translocator protein (TSPO) polymorphism explains differences in binding affinity of the PET radioligand PBR28. J. Cereb. Blood Flow Metab. 2012;32(1):1–5.
    1. Paganoni S., Alshikho M.J., Luppino S., Chan J., Pothier L., Schoenfeld D. A pilot trial of RNS60 in amyotrophic lateral sclerosis. Muscle Nerve. 2019;59(3):303–308.
    1. Park J.J.H., Harari O., Dron L., Lester R.T., Thorlund K., Mills E.J. An overview of platform trials with a checklist for clinical readers. J. Clin. Epidemiol. 2020;13(125):1–8.
    1. Parmar A., Zurcher N.R., Hooker J.M. Time Will Tell the Utility of Biomarkers. ACS Chem. Neurosci. 2020;11(12):1692–1695.
    1. Pascuzzi R.M., Shefner J., Chappell A.S., Bjerke J.S., Tamura R., Chaudhry V. A phase II trial of talampanel in subjects with amyotrophic lateral sclerosis. Amyotroph Lateral Scler. 2010;11(3):266–271.
    1. Poesen K., Van Damme P. Diagnostic and prognostic performance of neurofilaments in ALS. Front. Neurol. 2019;18(9):1167.
    1. Proudfoot M, Jones A, Talbot K, Al-Chalabi A, Turner MR. The ALSFRS as an outcome measure in therapeutic trials and its relationship to symptom onset. Amyotroph Lateral Scler Frontotemporal Degener 2016 Jul-Aug;17(5-6):414-425.
    1. Ray L.A., Bujarski S., Shoptaw S., Roche D.J., Heinzerling K., Miotto K. Development of the neuroimmune modulator ibudilast for the treatment of alcoholism: a randomized, placebo-controlled, human laboratory trial. Neuropsychopharmacology. 2017;42(9):1776–1788.
    1. Sanftner L.M., Gibbons J.A., Gross M.I., Suzuki B.M., Gaeta F.C., Johnson K.W. Cross-species comparisons of the pharmacokinetics of ibudilast. Xenobiotica. 2009;39(12):964–977.
    1. Sejbaek T., Nielsen H.H., Penner N., Plavina T., Mendoza J.P., Martin N.A. Dimethyl fumarate decreases neurofilament light chain in CSF and blood of treatment naive relapsing MS patients. J. Neurol. Neurosurg. Psychiatry. 2019;90(12):1324–1330.
    1. Shefner J.M., Liu D., Leitner M.L., Schoenfeld D., Johns D.R., Ferguson T. Quantitative strength testing in ALS clinical trials. Neurology. 2016;87(6):617–624.
    1. Suzumura A., Ito A., Yoshikawa M., Sawada M. Ibudilast suppresses TNFalpha production by glial cells functioning mainly as type III phosphodiesterase inhibitor in the CNS. Brain Res. 1999;837(1–2):203–212.
    1. Zach N., Ennist D.L., Taylor A.A., Alon H., Sherman A., Kueffner R. Being PRO-ACTive: what can a clinical trial database reveal about ALS? Neurotherapeutics. 2015;12(2):417–423.
    1. Zurcher N.R., Loggia M.L., Lawson R., Chonde D.B., Izquierdo-Garcia D., Yasek J.E. Increased in vivo glial activation in patients with amyotrophic lateral sclerosis: assessed with [(11)C]-PBR28. Neuroimage Clin. 2015;19(7):409–414.

Source: PubMed

3
Abonner