Efficacy and safety of GV1001 in patients with moderate-to-severe Alzheimer's disease already receiving donepezil: a phase 2 randomized, double-blind, placebo-controlled, multicenter clinical trial

Seong-Ho Koh, Hyuk Sung Kwon, Seong Hye Choi, Jee Hyang Jeong, Hae Ri Na, Chan Nyoung Lee, YoungSoon Yang, Ae Young Lee, Jae-Hong Lee, Kyung Won Park, Hyun Jeong Han, Byeong C Kim, Jin Se Park, Jee-Young Lee, Sangjae Kim, Kyu-Yong Lee, Seong-Ho Koh, Hyuk Sung Kwon, Seong Hye Choi, Jee Hyang Jeong, Hae Ri Na, Chan Nyoung Lee, YoungSoon Yang, Ae Young Lee, Jae-Hong Lee, Kyung Won Park, Hyun Jeong Han, Byeong C Kim, Jin Se Park, Jee-Young Lee, Sangjae Kim, Kyu-Yong Lee

Abstract

Background: Our previous studies showed that GV1001 has various protective effects against β-amyloid and other stressors. Based on these findings, we hypothesized that GV1001 might have beneficial effects in patients with Alzheimer's disease (AD).

Methods: A phase 2, double-blind, parallel-group, placebo-controlled, 6-month randomized clinical trial was performed to evaluate the safety and efficacy of subcutaneously administered GV1001. Between September 2017 and September 2019, 13 centers in South Korea recruited participants. A total of 106 patients were screened, and 96 patients with moderate-to-severe AD were randomized 1:1:1 to the placebo (group 1, n = 31), GV1001 0.56 mg (group 2, n = 33), and 1.12 mg (group 3, n = 32) groups. GV1001 was administered every week for 4 weeks (4 times), followed by every 2 weeks until week 24 (10 times). The primary endpoint was the change in the Severe Impairment Battery (SIB) score from baseline to week 24. The key secondary efficacy endpoints were the change in the Clinical Dementia Rating Sum of Box (CDR-SOB), Alzheimer's Disease Cooperative Study-Activities of Daily Living (ADCS-ADL), Neuropsychiatric Inventory (NPI), Mini-Mental State Examination, and Global Deterioration Scale scores. The safety endpoints were also assessed based on adverse events, laboratory test results, vital signs, and other observations related to safety.

Results: Group 3 showed less decrease in the SIB score at 12 and 24 weeks compared with group 1 (P < 0.05). These were not significantly observed in group 2. Among the secondary endpoints, only the NPI score showed significantly better improvement in group 2 than in group 3 at week 12; however, there were no other significant differences between the groups. Although the ADCS-ADL and CDR-SOB scores showed a pattern similar to SIB scores, a statistically significant result was not found. Adverse events were similar across all three groups.

Conclusions: The results indicate that GV1001 1.12 mg met the primary endpoint of a statistically significant difference. GV1001 was well tolerated without safety concerns. This study warrants a larger clinical trial.

Trial registration: ClinicalTrials.gov NCT03184467 . Registered on June 12, 2017.

Keywords: Alzheimer’s disease; Clinical trial; Efficacy; GV1001; Safety.

Conflict of interest statement

KSJ is an employer of GemVax & Kael Co., Ltd. and holds equity in the company. Other authors declare no competing interests.

Figures

Fig. 1
Fig. 1
Study design. Overall study design of the present clinical trial
Fig. 2
Fig. 2
Patient disposition. Participant flow throughout the trial. aOther medical conditions include renal dysfunction (n = 1), hepatic dysfunction (n = 2), and condition that in the opinion of the investigator can interfere with the interpretation of the study result (n = 1). bOne participant was excluded according to multiple reasons. cParticipants from a single center were excluded after the administration of the first study treatment, as mandatory clinical data, including baseline clinical data and the results of neurological examination performed at corresponding time points, were not uploaded to the central web-based system and the investigator of this center could not verify the source of these data. Abbreviations: MMSE Mini-Mental State Examination, I/E inclusion/exclusion, IP investigational product, FAS full analysis set, PPS per-protocol set
Fig. 3
Fig. 3
Effects of GV1001 on SIB, ADCS-ADL, and CDR-SOB scores in patients with AD with moderate-to-severe dementia. The change in the SIB score from baseline to 24 weeks was considered the primary endpoint. In the FAS and PPS, the patients assigned 1.12 mg of GV1001 had significantly better mean change from the baseline scores than the placebo group at weeks 12 and 24 (a and b, respectively). Among the secondary endpoints, ADCS-ADL (c, d) and CDR-SOB (e, f) scores showed a similar pattern to the SIB scores; however, statistical significance was not achieved in the FAS and PPS. LS indicates least squares. Error bars indicate standard error. *P < 0.05 (group 2 vs. group 3). Abbreviations: SIB Severe Impairment Battery, ADCS-ADL Alzheimer’s Disease Cooperative Study-Activities of Daily Living, CDR-SOB Clinical Dementia Rating Scale-Sum of Boxes, AD Alzheimer’s disease, FAS full analysis set, PPS per-protocol set, LS least square

References

    1. Prince M, Ali GC, Guerchet M, Prina AM, Albanese E, Wu YT. Recent global trends in the prevalence and incidence of dementia and survival with dementia. Alzheimers Res Ther. 2016;8(1):23. doi: 10.1186/s13195-016-0188-8.
    1. Cahill S. WHO’s global action plan on the public health response to dementia: some challenges and opportunities. Aging Ment Health. 2020;24(2):197–199. doi: 10.1080/13607863.2018.1544213.
    1. Braak H, Braak E. Neuropathological staging of Alzheimer-related changes. Acta Neuropathol. 1991;82(4):239–259. doi: 10.1007/BF00308809.
    1. Baufeld C, O'Loughlin E, Calcagno N, Madore C, Butovsky O. Differential contribution of microglia and monocytes in neurodegenerative diseases. J Neural Transm (Vienna) 2018;125(5):809–826. doi: 10.1007/s00702-017-1795-7.
    1. Cummings J, Lee G, Ritter A, Sabbagh M, Zhong K. Alzheimer’s disease drug development pipeline: 2020. Alzheimers Dement (Y) 2020;6:e12050.
    1. Tolar M, Abushakra S, Hey JA, Porsteinsson A, Sabbagh M. Aducanumab, gantenerumab, BAN2401, and ALZ-801-the first wave of amyloid-targeting drugs for Alzheimer’s disease with potential for near term approval. Alzheimers Res Ther. 2020;12(1):95. doi: 10.1186/s13195-020-00663-w.
    1. Schneider L. A resurrection of aducanumab for Alzheimer’s disease. Lancet Neurol. 2020;19(2):111–112. doi: 10.1016/S1474-4422(19)30480-6.
    1. Cummings J, Lee G, Ritter A, Sabbagh M, Zhong K. Alzheimer’s disease drug development pipeline: 2019. Alzheimers Dement ( Y) 2019;5(1):272–293. doi: 10.1016/j.trci.2019.05.008.
    1. Frozza RL, Lourenco MV, De Felice FG. Challenges for Alzheimer’s disease therapy: insights from novel mechanisms beyond memory defects. Front Neurosci. 2018;12:37. doi: 10.3389/fnins.2018.00037.
    1. Cummings J, Feldman HH, Scheltens P. The “rights” of precision drug development for Alzheimer’s disease. Alzheimers Res Ther. 2019;11(1):76. doi: 10.1186/s13195-019-0529-5.
    1. Schott JM, Aisen PS, Cummings JL, Howard RJ, Fox NC. Unsuccessful trials of therapies for Alzheimer’s disease. Lancet. 2019;393:29. doi: 10.1016/S0140-6736(18)31896-8.
    1. Anderson RM, Hadjichrysanthou C, Evans S, Wong MM. Why do so many clinical trials of therapies for AD fail? Lancet. 2017;390(10110):2327–2329. doi: 10.1016/S0140-6736(17)32399-1.
    1. Cao J, Hou J, Ping J, Cai D. Advances in developing novel therapeutic strategies for Alzheimer’s disease. Mol Neurodegener. 2018;13(1):64. doi: 10.1186/s13024-018-0299-8.
    1. Park HH, Lee KY, Kim S, Lee JW, Choi NY, Lee EH, Lee YJ, Lee SH, Koh SH. The novel vaccine peptide GV1001 effectively blocks beta-amyloid toxicity by mimicking the extra-telomeric functions of human telomerase reverse transcriptase. Neurobiol Aging. 2014;35(6):1255–1274. doi: 10.1016/j.neurobiolaging.2013.12.015.
    1. Park HH, Yu HJ, Kim S, Kim G, Choi NY, Lee EH, Lee YJ, Yoon MY, Lee KY, Koh SH. Neural stem cells injured by oxidative stress can be rejuvenated by GV1001, a novel peptide, by scavenging free radicals and enhancing survival signals. Neurotoxicology. 2016;55:131–141. doi: 10.1016/j.neuro.2016.05.022.
    1. Park HH, Lee KY, Park DW, Choi NY, Lee YJ, Son JW, Kim S, Moon C, Kim HW, Rhyu IJ, Koh SH. Tracking and protection of transplanted stem cells using a ferrocenecarboxylic acid-conjugated peptide that mimics hTERT. Biomaterials. 2018;155:80–91. doi: 10.1016/j.biomaterials.2017.11.009.
    1. McKhann GM, Knopman DS, Chertkow H, Hyman BT, Jack CR, Jr, Kawas CH, et al. The diagnosis of dementia due to Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 2011;7(3):263–269. doi: 10.1016/j.jalz.2011.03.005.
    1. Cooper J. Diagnostic and Statistical Manual of Mental Disorders (4th edn, text revision) (DSM–IV–TR) Washington, DC: American Psychiatric Association 2000. Br J Psychiatry. 2001;179:85. doi: 10.1192/bjp.179.1.85-a.
    1. Ahn IS, Kim JH, Ku HM, Saxton J, Kim DK. Reliability and validity of severe impairment battery (SIB) in Korean patients with dementia. J Korean Med Sci. 2006;21(3):506–517. doi: 10.3346/jkms.2006.21.3.506.
    1. Morris JC. The Clinical Dementia Rating (CDR): current version and scoring rules. Neurology. 1993;43(11):2412–2414. doi: 10.1212/WNL.43.11.2412-a.
    1. Galasko D, Bennett D, Sano M, Ernesto C, Thomas R, Grundman 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. doi: 10.1097/00002093-199700112-00005.
    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. doi: 10.1212/WNL.44.12.2308.
    1. Kang YW, Na DL, Hahn SH. A validity study on the Korean mini-mental state examination (K-MMSE) in dementia patients. J Korean Neurol Assoc. 1997;15:300–308.
    1. Reisberg B, Ferris SH, de Leon MJ, Crook T. The Global Deterioration Scale for the assessment of primary degenerative dementia. Am J Psychiatry. 1982;139(9):1136–1139. doi: 10.1176/ajp.139.9.1136.
    1. Siddiqui O, Hung HM, O'Neill R. MMRM vs. LOCF: a comprehensive comparison based on simulation study and 25 NDA datasets. J Biopharm Stat. 2009;19(2):227–246. doi: 10.1080/10543400802609797.
    1. Chauhan V, Chauhan A. Oxidative stress in Alzheimer’s disease. Pathophysiology. 2006;13(3):195–208. doi: 10.1016/j.pathophys.2006.05.004.
    1. Kwon HS, Koh SH. Neuroinflammation in neurodegenerative disorders: the roles of microglia and astrocytes. Transl Neurodegener. 2020;9(1):42. doi: 10.1186/s40035-020-00221-2.
    1. Jimenez S, Baglietto-Vargas D, Caballero C, Moreno-Gonzalez I, Torres M, Sanchez-Varo R, Ruano D, Vizuete M, Gutierrez A, Vitorica J. Inflammatory response in the hippocampus of PS1M146L/APP751SL mouse model of Alzheimer’s disease: age-dependent switch in the microglial phenotype from alternative to classic. J Neurosci. 2008;28(45):11650–11661. doi: 10.1523/JNEUROSCI.3024-08.2008.
    1. Saxton J, Swihart AA. Neuropsychological assessment of severely impaired elderly patients. Clin Geriatr Med. 1989;5(3):531–543. doi: 10.1016/S0749-0690(18)30669-4.
    1. Reisberg B, Doody R, Stoffler A, Schmitt F, Ferris S, Mobius HJ, et al. Memantine in moderate-to-severe Alzheimer’s disease. N Engl J Med. 2003;348(14):1333–1341. doi: 10.1056/NEJMoa013128.
    1. Farlow MR, Grossberg GT, Sadowsky CH, Meng X, Somogyi M. A 24-week, randomized, controlled trial of rivastigmine patch 13.3 mg/24 h versus 4.6 mg/24 h in severe Alzheimer’s dementia. CNS Neurosci Ther. 2013;19(10):745–752. doi: 10.1111/cns.12158.
    1. Feldman H, Gauthier S, Hecker J, Vellas B, Subbiah P, Whalen E, Donepezil MSAD Study Investigators Group A 24-week, randomized, double-blind study of donepezil in moderate to severe Alzheimer’s disease. Neurology. 2001;57(4):613–620. doi: 10.1212/WNL.57.4.613.
    1. Tariot PN, Farlow MR, Grossberg GT, Graham SM, McDonald S, Gergel I, Memantine Study Group Memantine treatment in patients with moderate to severe Alzheimer disease already receiving donepezil: a randomized controlled trial. JAMA. 2004;291(3):317–324. doi: 10.1001/jama.291.3.317.
    1. van Dyck CH, Tariot PN, Meyers B, Malca RE. A 24-week randomized, controlled trial of memantine in patients with moderate-to-severe Alzheimer’s disease. Alzheimer Dis Assoc Disord. 2007;21(2):136–143. doi: 10.1097/WAD.0b013e318065c495.
    1. Kyte JA. Cancer vaccination with the telomerase peptide GV1001. Expert Opin Investig Drugs. 2009;18(5):687–694. doi: 10.1517/13543780902897631.
    1. Jack CR, Jr, Bennett DA, Blennow K, Carrillo MC, Dunn B, Haeberlein SB, Holtzman DM, Jagust W, Jessen F, Karlawish J, Liu E, Molinuevo JL, Montine T, Phelps C, Rankin KP, Rowe CC, Scheltens P, Siemers E, Snyder HM, Sperling R, Elliott C, Masliah E, Ryan L, Silverberg N. NIA-AA research framework: toward a biological definition of AD. Alzheimers Dement. 2018;14(4):535–562. doi: 10.1016/j.jalz.2018.02.018.

Source: PubMed

3
Abonnere