Safety, Tolerability, and Pharmacokinetics of Crenezumab in Patients with Mild-to-Moderate Alzheimer's Disease Treated with Escalating Doses for up to 133 Weeks

Heather Guthrie, Lawrence S Honig, Helen Lin, Kaycee M Sink, Kathleen Blondeau, Angelica Quartino, Michael Dolton, Montserrat Carrasco-Triguero, Qinshu Lian, Tobias Bittner, David Clayton, Jillian Smith, Susanne Ostrowitzki, Heather Guthrie, Lawrence S Honig, Helen Lin, Kaycee M Sink, Kathleen Blondeau, Angelica Quartino, Michael Dolton, Montserrat Carrasco-Triguero, Qinshu Lian, Tobias Bittner, David Clayton, Jillian Smith, Susanne Ostrowitzki

Abstract

Background: Crenezumab is a fully humanized, monoclonal anti-amyloid-β immunoglobulin G4 antibody.

Objective: This Phase Ib study (NCT02353598) evaluated the safety, tolerability, and pharmacokinetics of crenezumabat doses of ≤120 mg/kg administered intravenously every 4 weeks (q4w). Immunogenicity and exploratory biomarkers were also evaluated.

Methods: In this multicenter, double-blind study, participants (aged 50-90 years) with mild-to-moderate Alzheimer's disease (AD) and amyloid-positive positron emission tomography (PET) scan were randomized to receive crenezumab 30 or 45 mg/kg (Cohort 1, n = 21), 60 mg/kg (Cohort 2, n = 21), or 120 mg/kg (Cohort 3, n = 19) or corresponding placebo (n = 14) intravenously q4w for 13 weeks. Seventy-one participants were subsequently enrolled in an optional open-label extension (OLE) and received crenezumab at the originally assigned dose level, except for Cohort 3 (crenezumab 60 mg/kg during OLE). Participants received regular brain MRIs to assess amyloid-related imaging abnormalities (ARIA). Results up to Week 133 are reported.

Results: Approximately 94% of participants experienced ≥1 adverse event (AE). Most AEs were mild or moderate; 15.5% experienced a Grade ≥3 AE. No ARIA-edema/effusion (ARIA-E) events were observed. New ARIA-micro hemorrhages and hemosiderosis (ARIA-H) were reported in 4.9% (double-blind treatment period) and 9.9% (combined double-blind treatment and OLE periods) of participants. Steady-state trough concentrations of crenezumab were dose-proportional and maintained for each dose level.

Conclusion: Crenezumab doses of ≤120 mg/kg intravenously q4w were well tolerated. The observed safety profile for ≤133 weeks of treatment in a mild-to-moderate AD population was similar to that seen in previous trials.

Keywords: Alzheimer’s disease; amyloid positron emission tomography; amyloid-β peptide; crenezumab; humanized monoclonal antibody; infusion site adverse event; magnetic resonance imaging; safety.

Conflict of interest statement

Authors’ disclosures available online (https://www.j-alz.com/manuscript-disclosures/20-0134r1).

Figures

Fig. 1
Fig. 1
Study design. Participants were enrolled sequentially. The next ascending dose cohort (60 or 120 mg/kg) was initiated if all previous dosing cohorts were supported by adequate safety and tolerability and provided that no previous dosing cohort was closed to enrollment due to safety concerns by the time the last participant enrolled in the study had completed the Week 5 MRI scan and the second study drug administration. Participation in the OLE phase was optional. The OLE dose was equivalent to the dose assigned during the double-blind randomization phase for all participants, except for those in the 120 mg/kg cohort who received 60 mg/kg IV every 4 weeks upon starting the OLE period. Additionally, at or after Week 133, participants in the 30 or 45 mg/kg dosing cohort were given the option to increase the dose of active drug to 60 mg/kg IV every 4 weeks. D, Day; IV, intravenously; MRI, magnetic resonance imaging; OLE, open-label extension; SMC, safety monitoring committee; W, week.
Fig. 2
Fig. 2
Mean (SD) serum crenezumab concentrations. Data shown are from the Phase II ABBY study (SRI cohort) for the 15 mg/kg dose and from the Phase Ib (GN29632) study reported here for the 30–120 mg/kg doses during the 13-week, randomized, placebo-controlled period. SD, standard deviation; SRI, safety run-in.
Fig. 3
Fig. 3
Plasma Aβ1–40(A) and Aβ1–42 (B) concentrations following administration of crenezumab during the 13-week, double-blind, randomized treatment phase. Aβ, amyloid-β.

References

    1. Prince M, Comas-Herrera A, Knapp M, Guerchet M, Karagiannidou M (2016) World Alzheimer Report 2016. Improving healthcare for peopleliving with dementia coverage: Quality and costs now and in the future. Alzheimer’s Disease International, London, UK.
    1. Alzheimer’s Association, What is Alzheimer’s? Alzheimer’s and Dementia basics,
    1. Masters CL, Bateman R, Blennow K, Rowe CC, Sperling RA, Cummings JL (2015) Alzheimer’s disease. Nat Rev Dis Primers 1, 15056.
    1. Cummings J, Gould H, Zhong K (2012) Advances in designs for Alzheimer’s disease clinical trials. Am J Neurodegener Dis 1, 205–216.
    1. Hardy J, Selkoe DJ (2002) The amyloid hypothesis of Alzheimer’s disease: Progress and problems on the road to therapeutics. Science 297, 353–356.
    1. Hyman B, West H, Rebeck G, Buldyrev S, Mantegna R, Ukleja M, Havlin S, Stanley H (1995) Quantitative analysis of senile plaques in Alzheimer’s disease: Observation of log-normal size distribution and molecular epidemiology of differences associated with apolipoprotein E genotype and trisomy 21 (Down syndrome). Proc Natl Acad Sci U S A 92, 3586–3590.
    1. Spires-Jones T, Meyer-Luehmann M, Osetek J, Jones P, Stern E, Bacskai B, Hyman B (2007) Impaired spine stability underlies plaque-related spine loss in an Alzheimer’s disease mouse model. Am J Pathol 171, 1304–1311.
    1. Koffie R, Meyer-Luehmann M, Hashimoto T, Adams K, Mielke M, Garcia-Alloza M, Micheva K, Smith S, Kim M, Lee V, Hyman B, Spires-Jones T (2009) Oligomeric amyloid β associates with postsynaptic densities and correlates with excitatory synapse loss near senile plaques. Proc Natl Acad Sci U S A 106, 4012–4017.
    1. Serrano-Pozo A, William C, Ferrer I, Uro-Coste E, Delisle M, Maurage C, Hock C, Nitsch R, Masliah E, Growdon J, Frosch M, Hyman B (2010) Benefical effect of human anti-amyloid-β active immunization on neurite morphology and tau pathology. Brain 133, 1312–1327.
    1. Palop J, Mucke L (2010) Amyloid-β-induced neuronal dysfunction in Alzheimer’s disease: From synapses toward neural networks. Nat Neurosci 13, 812–818.
    1. Mucke L, Selkoe D (2012) Neurotoxicity of amyloid β-protein: Synaptic and network dysfunction. Cold Spring Harb Perspect Med 2, a006338.
    1. Schneider LS, Mangialasche F, Andreasen N, Feldman H, Giacobini E, Jones R, Manua V, Mecocci P, Winblad B, Kivipelto M (2014) Clinical trials and late-stage drug development for Alzheimer’s disease: An appraisal from 1984 to 2014. J Intern Med 275, 251–283.
    1. Adolfsson O, Pihlgren M, Toni N, Varisco Y, Buccarello AL, Antoniello K, Lohmann S, Piorkowska K, Gafner V, Atwal JK, Maloney J, Chen M, Gogineni A, Weimer RM, Mortensen DL, Friesenhahn M, Ho C, Paul R, Pfeifer A, Muhs A, Watts RJ (2012) An effector-reduced anti-β-amyloid (Aβ) antibody with unique aβ binding properties promotes neuroprotection and glial engulfment of Aβ. J Neurosci 32, 9677–9689.
    1. Ultsch M, Li B, Maurer T, Mathieu M, Adolfsson O, Muhs A, Pfeifer A, Pihlgren M, Bainbridge TW, Reichelt M, Ernst JA, Eigenbrot C, Fuh G, Atwal JK, Watts RJ, Wang W (2016) Structure of crenezumab complex with Aβ shows loss of β-hairpin. Sci Rep 6, 39374.
    1. Yang T, Dang Y, Ostaszewski B, Mengel D, Steffen V, Rabe C, Bittner T, Walsh DM, Selkoe DJ (2018) Target engagement in an AD trial: Crenezumab lowers Aβ oligomer levels in CSF. Alzheimers Dement 14 (Suppl), P1669–P1670.
    1. Meilandt WJ, Maloney JA, Imperio J, Lalehzadeh G, Earr T, Crowell S, Bainbridge TW, Lu Y, Ernst JA, Fuji RN, Atwal JK (2019) Characterization of the selective in vitro and in vivo binding properties of crenezumab to oligomeric Aβ. Alzheimers Res Ther 11, 97.
    1. Cummings J, Cohen S, Van Dyck CH, Brody M, Curtis C, Cho W, Ward M, Friesenhahn M, Rabe C, Brunstein F, Quartino A, Honigberg LA, Fuji R, Clayton D, Mortensen D, Ho C, Paul R (2018) ABBY: A phase 2 randomized trial of crenezumab in mild to moderate Alzheimer disease. Neurology 90, e1889–e1897.
    1. Salloway S, Honigberg LA, Cho W, Ward M, Friesenhahn M, Brunstein F, Quartino A, Clayton D, Mortensen D, Bittner T, Ho C, Rabe C, Schauer SP, Wildsmith KR, Fuji RN, Suliman S, Reiman EM, Chen K, Paul R (2018) Amyloid positron emission tomography and cerebrospinal fluid results from a crenezumab anti-amyloid-beta antibody double-blind, placebo-controlled, randomized phase II study in mild-to-moderate Alzheimer’s disease (BLAZE). Alzheimers Res Ther 10, 96.
    1. , CREAD Study: A Study of Crenezumab Versus Placebo to Evaluate the Efficacy and Safety in Participants With Prodromal to Mild Alzheimer’s Disease (AD). Identifier: NCT02670083, .
    1. , A Study of Crenezumab Versus Placebo to Evaluate the Efficacy and Safety in Participants With Prodromal to Mild Alzheimer’s Disease (AD) (CREAD 2). Identifier: NCT03114657,
    1. Tariot PN, Lopera F, Langbaum JB, Thomas RG, Hendrix S, Schneider LS, Rios-Romenets S, Giraldo M, Acosta N, Tobon C, Ramos C, Espinosa A, Cho W, Ward M, Clayton D, Friesenhahn M, Mackey H, Honigberg L, Sanabria Bohorquez S, Chen K, Walsh T, Langlois C, Reiman EM (2018) The Alzheimer’s prevention initiative autosomal-dominant alzheimer’s disease trial: A study of crenezumab versus placebo in preclinical PSEN1 E280A mutation carriers to evaluate efficacy and safety in the treatment of autosomal-dominant Alzheimer’s disease, including a placebo-treated noncarrier cohort. Alzheimers Dement 4, 150–160.
    1. Barkhof F, Daams M, Scheltens P, Brashear HR, Arrighi HM, Bechten A, Morris K, McGovern M, Wattjes MP (2013) An MRI rating scale for amyloid-related imaging abnormalities with edema or effusion. AJNR Am J Neuroradiol 34, 1550–1555.
    1. Yoshida K, Moein A, Bittner T, Ostrowitzki S, Lin H, Honigberg L, Jin JY, Quartino A, (2020) Pharmacokinetics and pharmacodynamic effect of crenezumab on plasma and cerebrospinal fluid beta-amyloid in patients with mild-to-moderate Alzheimer’s disease. Alzheimers Res Ther 12, 16.
    1. Lin H, Schneider A, Quartino A, Bittner T, Hu N, Smith J, Cho W, Ostrowitzki S (2018) Safety, tolerability and pharmacokinetics of crenezumab in mild-to-moderate AD patients treated with escalating doses for up to 32 months. Poster presentation at the 70th Annual American Academy of Neurology (AAN) Annual Meeting, Los Angeles, CA, USA.
    1. Atwal JK, Ferl G, Meilandt WJ, Wetzel-Smith M, Li B, Bainbridge TW, Reichelt M, Ultsch M, Ernst JA, Wang W, Crowell S, Quartino AL, Fuji RN (2017) Crenezumab’s preferential binding of oligomeric amyloid-beta species underlies its unique mechanism of action. Neurology 88, S7.005.
    1. Kolditz M, Tesch F, Mocke L, Hoffken G, Ewig S, Schmitt J (2016) Burden and risk factors of ambulatory or hospitalized CAP: A population based cohort study. Respir Med 121, 32–38.

Source: PubMed

3
Abonnieren