A randomized, double-blind, phase 2b proof-of-concept clinical trial in early Alzheimer's disease with lecanemab, an anti-Aβ protofibril antibody

Chad J Swanson, Yong Zhang, Shobha Dhadda, Jinping Wang, June Kaplow, Robert Y K Lai, Lars Lannfelt, Heather Bradley, Martin Rabe, Akihiko Koyama, Larisa Reyderman, Donald A Berry, Scott Berry, Robert Gordon, Lynn D Kramer, Jeffrey L Cummings, Chad J Swanson, Yong Zhang, Shobha Dhadda, Jinping Wang, June Kaplow, Robert Y K Lai, Lars Lannfelt, Heather Bradley, Martin Rabe, Akihiko Koyama, Larisa Reyderman, Donald A Berry, Scott Berry, Robert Gordon, Lynn D Kramer, Jeffrey L Cummings

Abstract

Background: Lecanemab (BAN2401), an IgG1 monoclonal antibody, preferentially targets soluble aggregated amyloid beta (Aβ), with activity across oligomers, protofibrils, and insoluble fibrils. BAN2401-G000-201, a randomized double-blind clinical trial, utilized a Bayesian design with response-adaptive randomization to assess 3 doses across 2 regimens of lecanemab versus placebo in early Alzheimer's disease, mild cognitive impairment due to Alzheimer's disease (AD) and mild AD dementia.

Methods: BAN2401-G000-201 aimed to establish the effective dose 90% (ED90), defined as the simplest dose that achieves ≥90% of the maximum treatment effect. The primary endpoint was Bayesian analysis of 12-month clinical change on the Alzheimer's Disease Composite Score (ADCOMS) for the ED90 dose, which required an 80% probability of ≥25% clinical reduction in decline versus placebo. Key secondary endpoints included 18-month Bayesian and frequentist analyses of brain amyloid reduction using positron emission tomography; clinical decline on ADCOMS, Clinical Dementia Rating-Sum-of-Boxes (CDR-SB), and Alzheimer's Disease Assessment Scale-Cognitive Subscale (ADAS-Cog14); changes in CSF core biomarkers; and total hippocampal volume (HV) using volumetric magnetic resonance imaging.

Results: A total of 854 randomized subjects were treated (lecanemab, 609; placebo, 245). At 12 months, the 10-mg/kg biweekly ED90 dose showed a 64% probability to be better than placebo by 25% on ADCOMS, which missed the 80% threshold for the primary outcome. At 18 months, 10-mg/kg biweekly lecanemab reduced brain amyloid (-0.306 SUVr units) while showing a drug-placebo difference in favor of active treatment by 27% and 30% on ADCOMS, 56% and 47% on ADAS-Cog14, and 33% and 26% on CDR-SB versus placebo according to Bayesian and frequentist analyses, respectively. CSF biomarkers were supportive of a treatment effect. Lecanemab was well-tolerated with 9.9% incidence of amyloid-related imaging abnormalities-edema/effusion at 10 mg/kg biweekly.

Conclusions: BAN2401-G000-201 did not meet the 12-month primary endpoint. However, prespecified 18-month Bayesian and frequentist analyses demonstrated reduction in brain amyloid accompanied by a consistent reduction of clinical decline across several clinical and biomarker endpoints. A phase 3 study (Clarity AD) in early Alzheimer's disease is underway.

Trial registration: Clinical Trials.gov NCT01767311 .

Keywords: ADCOMS; Alzheimer’s disease; Amyloid; Amyloid PET; BAN2401; Biomarker; Clinical trial; Lecanemab; Neurofilament light; Neurogranin; p-tau.

Conflict of interest statement

CJS, YZ, SD, JW, JK, RYKL, HB, MR, AK, LR, RG, and LDK are employees of Eisai. LL is an employee of BioArctic. DAB and SB are employees of Berry Consultants. JLC provided consultation to the following pharmaceutical companies: Acadia, Accera, Actinogen, ADAMAS, Alkahest, Allergan, Alzheon, Avanir, Axovant, Axsome, BiOasis Technologies, Biogen, Eisai, Genentech, Grifols, Kyowa, Lilly, Lundbeck, Merck, Nutricia, Otsuka, QR Pharma, Resverlogix, Roche, Samus, Servier, Suven, Takeda, Toyoma, and United Neuroscience companies. JLC is supported by NIGMS grant P20GM109025; NINDS grant U01NS093334; NIA grant R01AG053798; and NIA grant P20AG068053.

Figures

Fig. 1
Fig. 1
Lecanemab Study 201 study design. Study 201 (NCT01767311) was an 18-month, multicenter, double-blind, placebo-controlled Bayesian design clinical trial employing response adaptive randomization across placebo and five lecanemab arms (2.5 mg/kg biweekly, 5 mg/kg monthly, 5 mg/kg biweekly, 10 mg/kg monthly, 10 mg/kg biweekly) to assess safety and efficacy in subjects with early Alzheimer's disease. At the first three interim analyses, if there is a .5% posterior probability that the most likely ED90 is superior to placebo by the (clinically significant difference; 25%), the trial will stop early for futility. Beginning at the 350-subject IA, and continuing to completion of the trial, the futility criterion is increased to 7.5%. Interim monitoring for early success occurs at each IA beginning when 350 subjects have been enrolled. At this point, if enrollment were to stop for early success, enough subjects would be available to complete the trial so that the full dose response could be modeled. If there is a .95% posterior probability that the most likely ED90 is better than placebo by the CSD, then early success is declared. Enrollment is stopped, but all randomized subjects continue for the full 18-month duration of the study. If the trial is not stopped early for futility or success, then trial success is evaluated at the completion of the trial when both accrual and follow-up for the primary endpoint are complete. At that time, if there is a .80% probability that the most likely ED90 is better than placebo by the CSD, the trial will be considered a success. R, randomization
Fig. 2
Fig. 2
Randomization allocations by treatment group per protocol-defined interim analyses. The response adaptive randomization correctly allocated subjects into the dose groups likely to be ED90 doses (10 mg/kg monthly and biweekly) as early as the first interim analysis at 197 subjects, with both emerging by the 300th subject randomized, and these doses remained the most likely doses to demonstrate efficacy throughout the remainder of the study. However, before the interim analysis of 350 subjects, Health Authorities restricted randomization around ApoE4 carrier status, whereby ApoE4 carriers (hetero- or homozygous) were not to be randomized to the 10 mg/kg biweekly dose going forward. As a consequence, the response adaptive randomization algorithm was revised. After each subsequent interim analysis (starting with 350 subjects randomized), the randomization probability vector was split between ApoE4 carrier and non-carrier strata to ensure no ApoE4 carriers were enrolled on the 10 mg/kg biweekly dose (more details in Appendix C). At the same time, the revised response adaptive randomization preserved the overall randomization probabilities
Fig. 3
Fig. 3
Change from baseline in brain amyloid pathophysiology. Results as measured by amyloid PET SUVr are shown in a. Outcomes from the qualitative (binary) visual read of the PET scans for conversion of brain amyloid pathology from positive to negative are depicted in b. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001 (all nominal). For PET analysis, N = 306 at 12 months and N = 288 at 18 months. The PET substudy was optional, so only a portion of the total enrolled subject population opted to participate
Fig. 4
Fig. 4
Efficacy assessments. a Change from baseline for 10 mg/kg biweekly and monthly doses in the Alzheimer’s Disease Composite Score (ADCOMS). The MMRM used treatment group, visit, clinical subgroup (MCI due to AD, Mild AD), the presence or absence of ongoing AD treatment at baseline, ApoE4 status (positive, negative), region, treatment group-by-visit interaction as factors, and baseline value as covariate. *P < 0.05 (nominal). The primary analysis conducted at month 12 of treatment for all subjects indicated that the 10 mg/kg biweekly dose had a 64% probability to be better than placebo by 25% on ADCOMS at 12 months, missing the pre-specified 80% probability threshold for success. Bayesian analysis at 18 months determined that the lecanemab 10 mg/kg biweekly dose had a 76% probability of being better than placebo by 25% on ADCOMS. In addition, Bayesian analyses indicated a 98% probability of being superior to placebo by any magnitude at both 12 and 18 months, respectively, which is consistent with subsequent conventional analysis results. b Results for 10 mg/kg biweekly and monthly doses on CDR-SB. The number of subjects that were assessed at each time point are indicated in the table. The MMRM used treatment group, visit, clinical subgroup (MCI due to AD, Mild AD), the presence or absence of ongoing AD treatment at baseline, ApoE4 status (positive, negative), region, treatment group-by-visit interaction as factors, and baseline value as covariate. *P < 0.05 (nominal). c Results for 10 mg/kg biweekly and monthly doses on ADAS-Cog14. The number of subjects that were assessed at each time point are indicated in the table. The MMRM used treatment group, visit, clinical subgroup (MCI due to AD, Mild AD), the presence or absence of ongoing AD treatment at baseline, ApoE4 status (positive, negative), region, treatment group-by-visit interaction as factors, and baseline value as covariate. *P < 0.05 (nominal)
Fig. 5
Fig. 5
Change from baseline in CSF biomarker measures. a Change from baseline in CSF Aβ1–42 measures. The combined 10 mg/kg monthly and 10 mg/kg biweekly group is compared versus placebo. The number of subjects that were assessed at each time point are indicated in the table. The MMRM used treatment group, visit, clinical subgroup (MCI due to AD, Mild AD), the presence or absence of ongoing AD treatment at baseline, ApoE4 status (positive, negative), region, treatment group-by-visit interaction as factors, and baseline value as covariate. *P < 0.001. b Change from baseline in p-tau measures. The combined 10 mg/kg monthly and 10 mg/kg biweekly group is compared versus placebo. The number of subjects that were assessed at each time point are indicated in the table. The MMRM used treatment group, visit, clinical subgroup (MCI due to AD, Mild AD), the presence or absence of ongoing AD treatment at baseline, ApoE4 status (positive, negative), region, treatment group-by-visit interaction as factors, and baseline value as covariate. *P < 0.001, **P = 0.005. c Change from baseline in t-tau measures. The combined 10 mg/kg monthly and 10 mg/kg biweekly group is compared versus placebo. The number of subjects that were assessed at each time point are indicated in the table. The MMRM used treatment group, visit, clinical subgroup (MCI due to AD, Mild AD), the presence or absence of ongoing AD treatment at baseline, ApoE4 status (positive, negative), region, treatment group-by-visit interaction as factors, and baseline value as covariate. *P = 0.029

References

    1. Burns A, Iliffe S. Alzheimer's disease. BMJ. 2009;338(feb05 1):b158. doi: 10.1136/bmj.b158.
    1. Dubois B, Feldman HH, Jacova C, Cummings JL, DeKosky ST, Barberger-Gateau P, Delacourte A, Frisoni G, Fox NC, Galasko D, Gauthier S, Hampel H, Jicha GA, Meguro K, O'Brien J, Pasquier F, Robert P, Rossor M, Salloway S, Sarazin M, de Souza LC, Stern Y, Visser PJ, Scheltens P. Revising the definition of Alzheimer’s disease: a new lexicon. Lancet Neurol. 2010;9(11):1118–1127. doi: 10.1016/S1474-4422(10)70223-4.
    1. Masters C, Bateman R, Blennow K, et al. Alzheimer’s disease. Nat Rev. 2015;1:15056.
    1. O’Nuallain B, Freir DB, Nicoll AJ, et al. Amyloid β-protein dimers rapidly form stable synaptotoxic protofibrils. J Neurosci. 2010;30(43):14411–14419. doi: 10.1523/JNEUROSCI.3537-10.2010.
    1. Paravastua AK, Leapmanb RD, Yaua WM, Tycko R. Molecular structural basis for polymorphism in Alzheimer’s amyloid fibrils. Proc Natl Acad Sci U S A. 2008;105(47):18349–18354. doi: 10.1073/pnas.0806270105.
    1. Yu X, Zheng J. Polymorphic structures of Alzheimer’s β-amyloid globulomers. PLoS One. 2011;6(6):e20575. doi: 10.1371/journal.pone.0020575.
    1. Lublin AL, Gandy S. Amyloid-β oligomers: possible roles as key neurotoxins in Alzheimer’s disease. Mt Sinai J Med. 2010;77(1):43–49. doi: 10.1002/msj.20160.
    1. Walsh DM, Selkoe DJ. Deciphering the molecular basis of memory failure in Alzheimer’s disease. Neuron. 2004;44(1):181–193. doi: 10.1016/j.neuron.2004.09.010.
    1. Knobloch M, Farinelli M, Konietzko U, Nitsch RM, Mansuy IM. Abeta oligomer-mediated long-term potentiation impairment involves protein phosphatase 1-dependent mechanisms. J Neurosci. 2007;27(29):7648–7653. doi: 10.1523/JNEUROSCI.0395-07.2007.
    1. Nilsberth C, Westlind-Danielsson A, Eckman CB, Condron MM, Axelman K, Forsell C, Stenh C, Luthman J, Teplow DB, Younkin SG, Näslund J, Lannfelt L. The ‘Arctic’ APP mutation (E693G) causes Alzheimer’s disease by enhanced Abeta protofibril formation. Nat Neurosci. 2001;4(9):887–893. doi: 10.1038/nn0901-887.
    1. Jicha GA. Is passive immunization for Alzheimer’s disease ‘alive and well’ or ‘dead and buried’? Expert Opin Biol Ther. 2009;9(4):481–491. doi: 10.1517/14712590902828285.
    1. Lacor PN, Buniel MC, Chang L, Fernandez SJ, Gong Y, Viola KL, Lambert MP, Velasco PT, Bigio EH, Finch CE, Krafft GA, Klein WL. Synaptic targeting by Alzheimer’s-related amyloid oligomers. J Neurosci. 2004;24(45):10191–10200. doi: 10.1523/JNEUROSCI.3432-04.2004.
    1. Tucker S, Möller C, Tegerstedt K, Lord A, Laudon H, Sjödahl J, Söderberg L, Spens E, Sahlin C, Waara ER, Satlin A, Gellerfors P, Osswald G, Lannfelt L. The murine version of BAN2401 (mAb158) selectively reduces amyloid-β protofibrils in brain and cerebrospinal fluid of tg-ArcSwe mice. J Alzheimers Dis. 2015;43(2):575–588. doi: 10.3233/JAD-140741.
    1. Sehlin D, Hedlund M, Lord A, Englund H, Gellerfors P, Paulie S, Lannfelt L, Pettersson FE. Heavy-chain complementarity-determining regions determine conformation selectivity of anti-Aβ antibodies. Neurodegener Dis. 2011;8(3):117–123. doi: 10.1159/000316530.
    1. Sehlin D, Englund H, Simu B, Karlsson M, Ingelsson M, Nikolajeff F, Lannfelt L, Pettersson FE. Large aggregates are the major soluble Aβ species in AD brain fractionated with density gradient ultracentrifugation. PLoS One. 2012;7(2):e32014. doi: 10.1371/journal.pone.0032014.
    1. Magnusson K, Sehlin D, Syvänen S, Svedberg MM, Philipson O, Söderberg L, Tegerstedt K, Holmquist M, Gellerfors P, Tolmachev V, Antoni G, Lannfelt L, Hall H, Nilsson LNG. Specific uptake of an amyloid-β-Protofibril-binding antibody-tracer in AβPP transgenic mouse brain. J Alzheimers Dis. 2013;37(1):29–40. doi: 10.3233/JAD-130029.
    1. Englund H, Sehlin D, Johansson AS, et al. Sensitive ELISA detection of amyloid-beta protofibrils in biological samples. J Neurochem. 2007;103:334–345.
    1. Lord A, Gumucio A, Englund H, Sehlin D, Sundquist VS, Söderberg L, Möller C, Gellerfors P, Lannfelt L, Pettersson FE, Nilsson LNG. An amyloid-beta protofibril-selective antibody prevents amyloid formation in a mouse model of Alzheimer’s disease. Neurobiol Dis. 2009;36(3):425–434. doi: 10.1016/j.nbd.2009.08.007.
    1. Logovinsky V, Satlin A, Lai R, Swanson C, Kaplow J, Osswald G, Basun H, Lannfelt L. Safety and tolerability of BAN2401 - a clinical study in Alzheimer’s disease with a protofibril selective Aβ antibody. Alzheimers Res Ther. 2016;8(1):14. doi: 10.1186/s13195-016-0181-2.
    1. Satlin A, Wang J, Logovinsky V, et al. Design of a Bayesian adaptive phase 2 proof-of-concept trial for BAN2401, a putative disease-modifying monoclonal antibody for the treatment of Alzheimer's disease. Alzheimers Dement (N Y) 2016;1:1–12.
    1. Wang J, Logovinsky V, Hendrix SB, Stanworth SH, Perdomo C, Xu L, Dhadda S, Do I, Rabe M, Luthman J, Cummings J, Satlin A. ADCOMS: a composite clinical outcome for prodromal Alzheimer’s disease trials. J Neurol Neurosurg Psychiatry. 2016;87(9):993–999. doi: 10.1136/jnnp-2015-312383.
    1. Novak G, Fox N, Clegg S, Nielsen C, Einstein S, Lu Y, Tudor IC, Gregg K, di J, Collins P, Wyman BT, Yuen E, Grundman M, Brashear HR, Liu E. Changes in brain volume with Bapineuzumab in mild to moderate Alzheimer’s disease. J Alzheimers Dis. 2016;49(4):1123–1134. doi: 10.3233/JAD-150448.
    1. Fox NC, Black RS, Gilman S, Rossor MN, Griffith SG, Jenkins L, Koller M. Effects of Abeta immunization (AN1792) on MRI measures of cerebral volume in Alzheimer disease. Neurology. 2005;64(9):1563–1572. doi: 10.1212/01.WNL.0000159743.08996.99.
    1. Sur C, Kost J, Scott D, Adamczuk K, Fox NC, Cummings JL, Tariot PN, Aisen PS, Vellas B, Voss T, Mahoney E, Mukai Y, Kennedy ME, Lines C, Michelson D, Egan MF. BACE inhibition causes rapid, regional, and non-progressive volume reduction in Alzheimer’s disease brain. Brain. 2020;143(12):3816–3826. doi: 10.1093/brain/awaa332.
    1. Adam J, Schwarz AJ, Sundell KL, Charil A, Case MG, Jaeger RK, Scott D, Bracoud L, Oh J, Suhy J, Pontecorvo MJ, Dickerson BC, Siemers ER. Magnetic resonance imaging measures of brain atrophy from the EXPEDITION3 trial in mild Alzheimer’s disease. Alzheimers Dement. 2019;5:328–337.
    1. Khalil M, Teunissen CE, Otto M, Piehl F, Sormani MP, Gattringer T, Barro C, Kappos L, Comabella M, Fazekas F, Petzold A, Blennow K, Zetterberg H, Kuhle J. Neurofilaments as biomarkers in neurological disorders. Nat Rev Neurol. 2018;14(10):577–589. doi: 10.1038/s41582-018-0058-z.
    1. Boche D, Donald J, Love S, Harris S, Neal JW, Holmes C, Nicoll JA. Reduction of aggregated tau in neuronal processes but not in the cell bodies after Abeta42 immunisation in Alzheimer’s disease. Acta Neuropathol. 2010;120(1):13–20. doi: 10.1007/s00401-010-0705-y.
    1. Blennow K, Zetterberg H, Rinne JO, Salloway S, Wei J, Black R, Grundman M, Liu E, AAB-001 201/202 Investigators Effect of immunotherapy with bapineuzumab on cerebrospinal fluid biomarker levels in patients with mild to moderate Alzheimer disease. Arch Neurol. 2012;69(8):1002–1010. doi: 10.1001/archneurol.2012.90.
    1. Ostrowitzki S, Lasser RA, Dorflinger E, et al. A phase III randomized trial of gantenerumab in prodromal Alzheimer’s disease [published correction appears in Alzheimers Res Ther. 2018 Sep 27;10(1):99] Alzheimers Res Ther. 2017;9(1):95. doi: 10.1186/s13195-017-0318-y.
    1. Molinuevo JL, Ayton S, Batrla R, Bednar MM, Bittner T, Cummings J, Fagan AM, Hampel H, Mielke MM, Mikulskis A, O’Bryant S, Scheltens P, Sevigny J, Shaw LM, Soares HD, Tong G, Trojanowski JQ, Zetterberg H, Blennow K. Current state of Alzheimer’s fluid biomarkers. Acta Neuropathol. 2018;136(6):821–853. doi: 10.1007/s00401-018-1932-x.
    1. Cummings J, Lee G, Ritter A, Zhong K. Alzheimer’s disease drug development pipeline: 2018. Alzheimers Dement (N Y). 2018;4(1):195–214. doi: 10.1016/j.trci.2018.03.009.

Source: PubMed

3
Předplatit