Effect of masupirdine (SUVN-502) on cognition in patients with moderate Alzheimer's disease: A randomized, double-blind, phase 2, proof-of-concept study

Ramakrishna Nirogi, John Ieni, Vinod Kumar Goyal, Jyothsna Ravula, Satish Jetta, Anil Shinde, Pradeep Jayarajan, Vijay Benade, Veera Raghava Chowdary Palacharla, Dhanunjay Kumar Dogiparti, Venkat Jasti, Alireza Atri, Jeffrey Cummings, Ramakrishna Nirogi, John Ieni, Vinod Kumar Goyal, Jyothsna Ravula, Satish Jetta, Anil Shinde, Pradeep Jayarajan, Vijay Benade, Veera Raghava Chowdary Palacharla, Dhanunjay Kumar Dogiparti, Venkat Jasti, Alireza Atri, Jeffrey Cummings

Abstract

Introduction: This study explored the efficacy and safety of a serotonin-6 receptor antagonist, masupirdine, as adjunct treatment in patients with moderate Alzheimer's disease (AD) concomitantly treated with donepezil and memantine.

Methods: The effects of masupirdine were evaluated in patients with moderate AD dementia on background treatment with donepezil and memantine. Five hundred thirty-seven patients were expected to be randomized in a 1:1:1 ratio, using permuted blocked randomization. After a 2- to 4-week screening period, the study consisted of a 26-week double-blind treatment period, and a 4-week washout period. The primary efficacy measure was the 11-item cognitive subscale of the Alzheimer's Disease Assessment Scale (ADAS-Cog 11). Secondary efficacy measures were Clinical Dementia Rating Scale-Sum of Boxes, Mini-Mental State Examination, 23-item Alzheimer's Disease Co-operative Study Activities of Daily Living, and 12-item Neuropsychiatric Inventory. Changes from baseline were analyzed using a mixed effects model for repeated measures (MMRM). A total of 564 patients were randomized to receive either daily masupirdine 50 mg (190 patients), masupirdine 100 mg (185 patients), or placebo (189 patients). The study is registered at ClinicalTrials.gov (NCT02580305).

Results: The MMRM results showed statistically non-significant treatment differences in change from baseline in ADAS-Cog 11 scores at week 26, comparing each masupirdine dose arm to the placebo arm. No significant treatment effects were observed in the secondary evaluations.

Discussion: Masupirdine was generally safe and well tolerated. Possible reasons for the observed trial results are discussed.

Highlights: Masupirdine was evaluated in moderate Alzheimer's disease patients.First trial in class with background treatment of donepezil and memantine.Masupirdine was generally safe and well tolerated.Possible reasons for the observed trial results are discussed.

Keywords: 5‐HT6 receptor; Alzheimer's Disease Assessment Scale–Cognitive subscale; Alzheimer's disease; Clinical Dementia Rating–Sum of Boxes; SUVN‐502; clinical trials; masupirdine.

Conflict of interest statement

Dr. Cummings has provided consultation to AB Science, Acadia, Alkahest, AlphaCognition, ALZPathFinder, Annovis, AriBio, Artery, Avanir, Biogen, Biosplice, Cassava, Cerevel, Clinilabs, Cortexyme, Diadem, EIP Pharma, Eisai, GatehouseBio, GemVax, Genentech, Green Valley, Grifols, Janssen, Karuna, Lexeo, Lilly, Lundbeck, LSP, Merck, NervGen, Novo Nordisk, Oligomerix, Ono, Otsuka, PharmacotrophiX, PRODEO, Prothena, ReMYND, Renew, Resverlogix, Roche, Signant Health, Suven, Unlearn AI, Vaxxinity, VigilNeuro pharmaceutical, assessment, and investment companies. Dr. Alireza Atri has received honoraria for consulting; participated in independent data safety monitoring boards; provided educational lectures, programs, and materials; or served on advisory boards for AbbVie, Acadia, Allergan, the Alzheimer's Association, Axovant, AZ Therapies, Biogen, Eisai, Grifols, Harvard Medical School Graduate Continuing Education, JOMDD, Lundbeck, Merck, Roche/Genentech, Novo Nordisk, Qynapse, Sunovion, Suven, and Synexus. He receives royalties from Oxford University Press for a medical book on dementia. He receives institutional grant/contract funding from NIA/NIH 1P30AG072980, AZ DHS CTR040636, Washington University St. Louis, and Gates Ventures; and his institution receives funding for multiple clinical trial grants, contracts and projects from consortia, foundations, and companies for which he serves as site‐PI. Drs. Atri and Cummings did not receive compensation for developing this manuscript. John Ieni has provided consultation services for clinical development to Suven Life Sciences and Oligomerix, Inc. Ramakrishna Nirogi, Vinod Kumar Goyal, Jyothsna Ravula, Satish Jetta, Anil Shinde, Pradeep Jayarajan, Vijay Benade, Veera Raghava Chowdary Palacharla, Dhanunjay Kumar Dogiparti, Venkat Jasti are full‐time employees of Suven Life Sciences Ltd.

© 2022 The Authors. Alzheimer's & Dementia: Translational Research & Clinical Interventions published by Wiley Periodicals, LLC on behalf of Alzheimer's Association.

Figures

FIGURE 1
FIGURE 1
Patient disposition. mITT, modified intent to treat population
FIGURE 2
FIGURE 2
Adjusted mean change in ADAS‐Cog 11 (mITT); error bars represent standard error of mean; ADAS‐Cog 11, 11‐item Alzheimer's Disease Assessment Scale, Cognitive Subscale; mITT, modified intent to treat population

References

    1. 2020 Alzheimer's disease facts and figures. Alzheimers Dement. 2020;16(3):391‐460.
    1. Atri A. The Alzheimer's disease clinical spectrum: diagnosis and management. Med Clin North Am. 2019;103(2):263‐293.
    1. Alva G, Cummings JL. Relative tolerability of Alzheimer's disease treatments. Psychiatry (Edgmont). 2008;5(11):27‐36.
    1. Farlow MR, Cummings JL. Effective pharmacologic management of Alzheimer's disease. Am J Med. 2007;120(5):388‐397.
    1. Atri A. Current and future treatments in Alzheimer's disease. Semin Neurol. 2019;39(2):227‐240.
    1. Cummings J, Aisen P, Lemere C, Atri A, Sabbagh M, Salloway S. Aducanumab produced a clinically meaningful benefit in association with amyloid lowering. Alzheimers Res Ther. 2021;13(1):98.
    1. Cummings J, Aisen P, Apostolova LG, Atri A, Salloway S, Weiner M. Aducanumab: appropriate use recommendations. J Prev Alzheimers Dis. 2021;8(4):398‐410.
    1. Monsma FJ Jr, Shen Y, Ward RP, Hamblin MW, Sibley DR. Cloning and expression of a novel serotonin receptor with high affinity for tricyclic psychotropic drugs. Mol Pharmacol. 1993;43(3):320‐327.
    1. Roth BL, Craigo SC, Choudhary MS, et al. Binding of typical and atypical antipsychotic agents to 5‐hydroxytryptamine‐6 and 5‐hydroxytryptamine‐7 receptors. J Pharmacol Exp Ther. 1994;268(3):1403‐1410.
    1. Helboe L, Egebjerg J, de Jong IE. Distribution of serotonin receptor 5‐HT6 mRNA in rat neuronal subpopulations: a double in situ hybridization study. Neuroscience. 2015;310:442‐454.
    1. Dawson LA, Nguyen HQ, Li P. The 5‐HT(6) receptor antagonist SB‐271046 selectively enhances excitatory neurotransmission in the rat frontal cortex and hippocampus. Neuropsychopharmacology. 2001;25(5):662‐668.
    1. Dawson LA. The central role of 5‐HT6 receptors in modulating brain neurochemistry. Int Rev Neurobiol. 2011;96:1‐26.
    1. Mitchell ES, Neumaier JF. 5‐HT6 receptors: a novel target for cognitive enhancement. Pharmacol Ther. 2005;108(3):320‐333.
    1. Marazziti D, Baroni S, Borsini F, et al. Serotonin receptors of type 6 (5‐HT6): from neuroscience to clinical pharmacology. Curr Med Chem. 2013;20(3):371‐377.
    1. de Jong IEM, Mørk A. Antagonism of the 5‐HT6 receptor ‐ Preclinical rationale for the treatment of Alzheimer's disease. Neuropharmacology. 2017;125:50‐63.
    1. Ferrero H, Solas M, Francis PT, Ramirez MJ. Serotonin 5‐HT6 receptor antagonists in Alzheimer's disease: therapeutic rationale and current development status. CNS Drugs. 2017;31(1):19‐32.
    1. Lalut J, Karila D, Dallemagne P, Rochais C. Modulating 5‐HT4 and 5‐HT6 receptors in Alzheimer's disease treatment. Future Med Chem. 2017;9(8):781‐795.
    1. Wilkinson D, Windfeld K, Colding‐Jørgensen E. Safety and efficacy of idalopirdine, a 5‐HT6 receptor antagonist, in patients with moderate Alzheimer's disease (LADDER): a randomised, double‐blind, placebo‐controlled phase 2 trial. Lancet Neurol. 2014;13(11):1092‐1099.
    1. Atri A, Frölich L, Ballard C, et al. Effect of idalopirdine as adjunct to cholinesterase inhibitors on change in cognition in patients with Alzheimer disease: three randomized clinical trials. JAMA. 2018;319(2):130‐142.
    1. Maher‐Edwards G, Watson C, Ascher J, et al. Two randomized controlled trials of SB742457 in mild‐to‐moderate Alzheimer's disease. Alzheimers Dement (N Y). 2015;1(1):23‐36.
    1. Lang FM, Mo Y, Sabbagh M, et al. Intepirdine as adjunctive therapy to donepezil for mild‐to‐moderate Alzheimer's disease: a randomized, placebo‐controlled, phase 3 clinical trial (MINDSET). Alzheimers Dement (N Y). 2021;7(1):e12136.
    1. Fullerton T, Binneman B, David W, et al. A Phase 2 clinical trial of PF‐05212377 (SAM‐760) in subjects with mild to moderate Alzheimer's disease with existing neuropsychiatric symptoms on a stable daily dose of donepezil. Alzheimers Res Ther. 2018;10(1):38.
    1. Weintraub D. Impact of SYN120 (a Dual 5‐HT6/5‐HT2A antagonist) on psychiatric symptoms in patients with Parkinson's disease dementia: results from a Phase 2a Study. Movement disorders, 2018, 33, S105
    1. Lang FM, Kwon DY, Aarsland D, et al. An international, randomized, placebo‐controlled, phase 2b clinical trial of intepirdine for dementia with Lewy bodies (HEADWAY‐DLB). Alzheimers Dement (N Y). 2021;7(1):e12171.
    1. Nirogi R, Shinde A, Kambhampati RS, et al. Discovery and development of 1‐[(2‐bromophenyl)sulfonyl]‐5‐methoxy‐3‐[(4‐methyl‐1‐piperazinyl)methyl]‐1H‐indole dimesylate monohydrate (SUVN‐502): A novel, potent, selective and orally active serotonin 6 (5‐HT6) receptor antagonist for potential treatment of Alzheimer's disease. J Med Chem. 2017;60(5):1843‐1859.
    1. Nirogi R, Abraham R, Benade V, et al. SUVN‐502, a novel, potent, pure, and orally active 5‐HT6 receptor antagonist: pharmacological, behavioral, and neurochemical characterization. Behav Pharmacol. 2019;30(1):16‐35.
    1. Folstein MF, Folstein SE, McHugh PR. “Mini‐mental state”. A practical method for grading the cognitive state of patients for the clinician. J Psychiatr Res. 1975;12(3):189‐198.
    1. McKhann G, Drachman D, Folstein M, Katzman R, Price D, Stadlan EM. Clinical diagnosis of Alzheimer's disease: report of the NINCDS‐ADRDA Work Group under the auspices of Department of Health and Human Services Task Force on Alzheimer's Disease. Neurology. 1984;34(7):939‐944.
    1. Rosen WG, Mohs RC, Davis KL. A new rating scale for Alzheimer's disease. Am J Psychiatry. 1984;141(11):1356‐1364.
    1. Hughes CP, Berg L, Danziger WL, Coben LA, Martin RL. A new clinical scale for the staging of dementia. Br J Psychiatry. 1982;140:566‐572.
    1. Galasko D, Bennett D, Sano M, et al. An inventory to assess activities of daily living for clinical trials in Alzheimer's disease. The Alzheimer's disease cooperative study. Alzheimer Dis Assoc Disord. 1997;11 Suppl 2:S33‐S39.
    1. Cummings JL, Mega M, Gray K, Rosenberg‐Thompson S, Carusi DA, Gornbein J. The Neuropsychiatric Inventory: comprehensive assessment of psychopathology in dementia. Neurology. 1994;44(12):2308‐2314.
    1. Cummings JL. The Neuropsychiatric inventory: assessing psychopathology in dementia patients. Neurology. 1997;48(5 Suppl 6):S10‐S16.
    1. Schneider LS, Sano M. Current Alzheimer's disease clinical trials: methods and placebo outcomes. Alzheimers Dement. 2009;5(5):388‐397.
    1. Posner K, Brown GK, Stanley B, et al. The Columbia‐Suicide Severity Rating Scale: initial validity and internal consistency findings from three multisite studies with adolescents and adults. Am J Psychiatry. 2011;168(12):1266‐1277.
    1. Nirogi R, Mudigonda K, Bhyrapuneni G, et al. Safety, tolerability and pharmacokinetics of the serotonin 5‐HT6 receptor antagonist, SUVN‐502, in healthy young adults and elderly subjects. Clin Drug Investig. 2018;38(5):401‐415.
    1. Jelic V, Darreh‐Shori T. Donepezil: a review of pharmacological characteristics and role in the management of Alzheimer disease. Clinical Medical Insights: Therapeutics. 2010;2:771‐788.
    1. Johnson JW, Kotermanski SE. Mechanism of action of memantine. Curr Opin Pharmacol. 2006;6(1):61‐67.
    1. Atri A, Hendrix SB, Pejović V, et al. Cumulative, additive benefits of memantine‐donepezil combination over component monotherapies in moderate to severe Alzheimer's dementia: a pooled area under the curve analysis. Alzheimers Res Ther. 2015;7(1):28.
    1. Deardorff WJ, Grossberg GT. A fixed‐dose combination of memantine extended‐release and donepezil in the treatment of moderate‐to‐severe Alzheimer's disease. Drug Des Devel Ther. 2016;10:3267‐3279.
    1. Atri A, Shaughnessy LW, Locascio JJ, Growdon JH. Long‐term course and effectiveness of combination therapy in Alzheimer disease. Alzheimer Dis Assoc Disord. 2008;22(3):209‐221.
    1. Jack CR Jr, Bennett DA, Blennow K, et al. NIA‐AA research framework: toward a biological definition of Alzheimer's disease. Alzheimers Dement. 2018;14(4):535‐562.
    1. Nelson PT, Head E, Schmitt FA, et al. Alzheimer's disease is not “brain aging”: neuropathological, genetic, and epidemiological human studies. Acta Neuropathol. 2011;121(5):571‐587.
    1. Jack CR Jr, Lowe VJ, Senjem ML, et al. 11C PiB and structural MRI provide complementary information in imaging of Alzheimer's disease and amnestic mild cognitive impairment. Brain. 2008;131(Pt 3):665‐680.
    1. Johnson KA, Sperling RA, Gidicsin CM, et al. Florbetapir (F18‐AV‐45) PET to assess amyloid burden in Alzheimer's disease dementia, mild cognitive impairment, and normal aging. Alzheimers Dement. 2013;9(5 Suppl):S72‐S83.
    1. Rowe CC, Ng S, Ackermann U, et al. Imaging beta‐amyloid burden in aging and dementia. Neurology. 2007;68(20):1718‐1725.
    1. Chakraborty S, Lennon JC, Malkaram SA, Zeng Y, Fisher DW, Dong H. Serotonergic system, cognition, and BPSD in Alzheimer's disease. Neurosci Lett. 2019;704:36‐44.
    1. Cummings J, Nirogi R, Jayarajan P, et al. Potential benefits of Masupirdine (SUVN‐502) on behavioral and psychological symptoms in patients with moderate Alzheimer's disease. J Prev Alz Dis 2019; 6(S1):P038.

Source: PubMed

3
Iratkozz fel