A phase 2, randomized, double-blind, placebo-controlled trial of AMG 301, a pituitary adenylate cyclase-activating polypeptide PAC1 receptor monoclonal antibody for migraine prevention

Messoud Ashina, David Doležil, Jo H Bonner, Lifen Zhou, Jan Klatt, Hernan Picard, Daniel D Mikol, Messoud Ashina, David Doležil, Jo H Bonner, Lifen Zhou, Jan Klatt, Hernan Picard, Daniel D Mikol

Abstract

Objective: To assess the safety and efficacy of AMG 301, an inhibitor of the pituitary adenylate cyclase-activating polypeptide (PACAP)-1 (PAC1) receptor, for prevention of migraine.

Methods: In a double-blind trial, patients were randomized 4:3:3 to placebo, AMG 301 210 mg every 4 weeks, or AMG 301 420 mg every 2 weeks for 12 weeks. Effect on monthly migraine days and other secondary measures were assessed over weeks 9-12. Safety and tolerability were assessed.

Results: Of 343 randomized patients (mean age, 41.8-42.5 years), the majority were women (85.4-90.4%), white (94.1-96.2%), and had episodic migraine (62.5-67.9%). A total of 305 patients completed treatment (placebo, n = 124; AMG 301 210 mg, n = 94; AMG 301 420 mg, n = 87). Least squares mean reduction at week 12 in monthly migraine days from baseline was -2.5 (0.4) days for placebo and -2.2 (0.5) days for both AMG 301 treatment groups. No difference between AMG 301 and placebo on any measure of efficacy was observed; mean (95% confidence interval) treatment difference versus placebo for monthly migraine days for AMG 301 210 mg, 0.3 (-0.9 to 1.4); AMG 301 420 mg, 0.3 (-0.9 to 1.4). The incidence of adverse events was similar across groups.

Conclusion: AMG 301 offered no benefit over placebo for migraine prevention; further studies may be necessary to fully understand the role of PACAP isoforms and its receptors in migraine pathophysiology.

Study registration: ClinicalTrials.gov: NCT03238781.

Keywords: AMG 301; chronic migraine; episodic migraine; pituitary adenylate cyclase-activating polypeptide; preventive treatment.

Conflict of interest statement

Declaration of conflicting interests: The authors declared the following potential conflicts of interest with respect to the research, authorship, and/or publication of this article: MA is a consultant, speaker or scientific advisor for Alder, Allergan, Amgen, Eli Lilly, Lundbeck, Novartis and Teva; and a primary investigator for Alder, Allergan, Amgen, Eli Lilly, Novartis and Teva. He has no ownership interest and does not own stocks in any pharmaceutical company. He serves as associate editor of Cephalalgia, associate editor of Headache, and associate editor of the Journal of Headache and Pain; and is the President of the International Headache Society.

DD is a speaker, consultant or scientific advisor for Allergan, Amgen, Eli Lilly, Novartis, and Teva, as well as a primary investigator for Alder, Allergan, Amgen, Eli Lilly, Novartis, and Teva.

JHB reports no conflicts. JK is an employee of Novartis and owns Novartis stock. LZ, HP and DDM are employees of Amgen Inc., and own Amgen stock/stock options.

Figures

Figure 1.
Figure 1.
Trial design. Q2W: every 2 weeks; Q4W: every 4 weeks; SC: subcutaneous. *18 weeks after last dose of investigational product.
Figure 2.
Figure 2.
Patient disposition. Q2W: every 2 weeks; Q4W: every 4 weeks; TEAE: treatment-emergent adverse event.
Figure 3.
Figure 3.
Effect of AMG 301 and placebo on (a) change from baseline of MMD, (b) proportion of patients achieving a ≥ 50% reduction in MMD, and (c) change from baseline of monthly acute migraine-specific medication use as assessed in the last 4 weeks of a 12-week double-blind treatment period. LS: least squares; MMD: monthly migraine days; MSMD: migraine-specific medication days; N: number of patients in the primary analysis dataset (i.e. received ≥ 1 dose of investigational product and completed ≥ 1 post-baseline monthly electronic diary measurement); N1: number of patients with observed data; OR: odds ratio; Q2W: every 2 weeks; Q4W: every 4 weeks. *Odds ratio versus placebo.
Figure 4.
Figure 4.
Effect of AMG 301 and placebo on change from baseline for (a) MPFID impact on everyday activities sub-score, and (b) MPFID on physical impairment sub-score as assessed in the last 4 weeks of a 12-week double-blind treatment period. LS: least squares; MPFID: Migraine Physical Function Impact Diary; N: number of patients in the primary analysis dataset (i.e. received ≥ 1 dose of investigational product and completed ≥ 1 post-baseline monthly electronic diary measurement); Q2W: every 2 weeks; Q4W: every 4 weeks.

References

    1. Estemalik E, Tepper S. Preventive treatment in migraine and the new US guidelines. Neuropsychiatr Dis Treat 2013; 9: 709–720.
    1. Hepp Z, Dodick DW, Varon SF, et al. Persistence and switching patterns of oral migraine prophylactic medications among patients with chronic migraine: A retrospective claims analysis. Cephalalgia 2017; 37: 470–485.
    1. Hargreaves R, Olesen J. Calcitonin gene-related peptide modulators – the history and renaissance of a new migraine drug class. Headache 2019; 59: 951–970.
    1. Charles A, Pozo-Rosich P. Targeting calcitonin gene-related peptide: A new era in migraine therapy. Lancet 2019; 394: 1765–1774.
    1. Dodick DW. CGRP ligand and receptor monoclonal antibodies for migraine prevention: Evidence review and clinical implications. Cephalalgia 2019; 39: 445–458.
    1. Schytz HW, Olesen J, Ashina M. The PACAP receptor: A novel target for migraine treatment. Neurotherapeutics 2010; 7: 191–196.
    1. Schytz HW, Birk S, Wienecke T, et al. PACAP38 induces migraine-like attacks in patients with migraine without aura. Brain 2009; 132: 16–25.
    1. Tajti J, Uddman R, Edvinsson L. Neuropeptide localization in the “migraine generator” region of the human brainstem. Cephalalgia 2001; 21: 96–101.
    1. Uddman R, Tajti J, Hou M, et al. Neuropeptide expression in the human trigeminal nucleus caudalis and in the cervical spinal cord C1 and C2. Cephalalgia 2002; 22: 112–116.
    1. Guo S, Vollesen AL, Hansen YB, et al. Part II: Biochemical changes after pituitary adenylate cyclase-activating polypeptide-38 infusion in migraine patients. Cephalalgia 2017; 37: 136–147.
    1. Vaudry D, Falluel-Morel A, Bourgault S, et al. Pituitary adenylate cyclase-activating polypeptide and its receptors: 20 years after the discovery. Pharmacol Rev 2009; 61: 283–357.
    1. Ghanizada H, Al-Karagholi MA, Arngrim N, et al. PACAP27 induces migraine-like attacks in migraine patients. Cephalalgia 2020; 40: 57–67.
    1. Rahmann A, Wienecke T, Hansen JM, et al. Vasoactive intestinal peptide causes marked cephalic vasodilation, but does not induce migraine. Cephalalgia 2008; 28: 226–236.
    1. Amin FM, Hougaard A, Schytz HW, et al. Investigation of the pathophysiological mechanisms of migraine attacks induced by pituitary adenylate cyclase-activating polypeptide-38. Brain 2014; 137: 779–794.
    1. Han X, Ran Y, Su M, et al. Chronic changes in pituitary adenylate cyclase-activating polypeptide and related receptors in response to repeated chemical dural stimulation in rats. Mol Pain 2017; 13: 1–10.
    1. Rubio-Beltran E, Correnti E, Deen M, et al. PACAP38 and PAC1 receptor blockade: A new target for headache? J Headache Pain 2018; 19: 64.
    1. Headache Classification Committee of the International Headache Society. The International Classification of Headache Disorders, 3rd edition. Cephalalgia 2018; 38: 1–211.
    1. Riesco N, Perez-Alvarez AI, Verano L, et al. Prevalence of cranial autonomic parasympathetic symptoms in chronic migraine: Usefulness of a new scale. Cephalalgia 2016; 36: 346–350.
    1. Kawata AK, Hsieh R, Bender R, et al. Psychometric evaluation of a novel instrument assessing the impact of migraine on physical functioning: The Migraine Physical Function Impact Diary. Headache 2017; 57: 1385–1398.
    1. Vollesen ALH, Amin FM, Ashina M. Targeted pituitary adenylate cyclase-activating peptide therapies for migraine. Neurotherapeutics 2018; 15: 371–376.
    1. Amin FM, Asghar MS, Guo S, et al. Headache and prolonged dilatation of the middle meningeal artery by PACAP38 in healthy volunteers. Cephalalgia 2012; 32: 140–149.
    1. Guo S, Vollesen AL, Olesen J, et al. Premonitory and nonheadache symptoms induced by CGRP and PACAP38 in patients with migraine. Pain 2016; 157: 2773–2781.
    1. Hoffmann J, Miller S, Martins-Oliveira M, et al. PAC1 receptor blockade reduces central nociceptive activity: New approach for primary headache? Pain 2020; 161: 1670–1681.
    1. Hoffmann J, Supronsinchai W, Akerman S, et al. Evidence for orexinergic mechanisms in migraine. Neurobiol Dis 2015; 74: 137–143.
    1. Chabi A, Zhang Y, Jackson S, et al. Randomized controlled trial of the orexin receptor antagonist filorexant for migraine prophylaxis. Cephalalgia 2015; 35: 379–388.
    1. Ashina H, Guo S, Vollesen ALH, et al. PACAP38 in human models of primary headaches. J Headache Pain 2017; 18: 110.
    1. Holland PR, Barloese M, Fahrenkrug J. PACAP in hypothalamic regulation of sleep and circadian rhythm: Importance for headache. J Headache Pain 2018; 19: 20.
    1. Bertels Z, Pradhan AAA. Emerging treatment targets for migraine and other headaches. Headache 2019; 59: 50–65.
    1. Uluduz D, Ayta S, Ozge A, et al. Cranial autonomic features in migraine and migrainous features in cluster headache. Noro Psikiyatr Ars 2018; 55: 220–224.
    1. Snoer AH, Lund N, Jensen RH, et al. More precise phenotyping of cluster headache using prospective attack reports. Eur J Neurol 2019; 26: 1303–1309, e85.
    1. Fahrenkrug J, Hannibal J, Tams J, et al. Immunohistochemical localization of the VIP1 receptor (VPAC1R) in rat cerebral blood vessels: Relation to PACAP and VIP containing nerves. J Cereb Blood Flow Metab 2000; 20: 1205–1214.
    1. Grant S, Lutz EM, McPhaden AR, et al. Location and function of VPAC1, VPAC2 and NPR-C receptors in VIP-induced vasodilation of porcine basilar arteries. J Cereb Blood Flow Metab 2006; 26: 58–67.
    1. Akerman S, Goadsby PJ. Neuronal PAC1 receptors mediate delayed activation and sensitization of trigeminocervical neurons: Relevance to migraine. Sci Transl Med 2015; 7: 308ra157.

Source: PubMed

3
Iratkozz fel