Skip to main content

Table 1 Completed trials of gantenerumab

From: Gantenerumab: an anti-amyloid monoclonal antibody with potential disease-modifying effects in early Alzheimer’s disease

Study name, design, and NCT

Study population

Gantenerumab dose

Primary endpoint

Other key findings

SCarlet RoAD

Phase 2/3, global double-blind placebo-controlled study (study start date: 2010)

(NCT01224106)

N = 797

Prodromal ADa, 50–85 years,

CSF Aβ1–42 confirmed pathology < 600 pg/mL

Gantenerumab 105 mg SC Q4W

Gantenerumab 225 mg SC Q4W

Placebo

Change from baseline in CDR-SB at 2 years in the gantenerumab group:

105 mg, 1.69 (= .67)

225 mg, 1.73 (= .45)

Dosing stopped due to preplanned interim futility in 2014

Study converted to OLE in 2015

CSF biomarkers in gantenerumab group (n = 209); change from baseline at 104 weeks:

Aβ(1–42): gantenerumab 105 mg, -1.06% (= .98); gantenerumab 225 mg, 7.55% (= .09)

t-tau: gantenerumab 105 mg, – 1.08% (= .05); gantenerumab 225 mg, – 2.91% (= .02)

p-tau: gantenerumab 105 mg, – 5.61% (≤ .001); gantenerumab 225 mg, – 7.15% (≤ 0.001)

neurogranin: gantenerumab 105 mg, – 4.58% (= .79); gantenerumab 225 mg, – 11.76 (= .18)

PET substudy [34] (n = 115):

No changes in CSF Aβ1–42 were found for either group

Gantenerumab 105 mg: – 4.85% change from baseline in CSF p-tau (< .01) at week 104; – 1.45% change from baseline in CSF t-tau (< .01) at week 104; 0.19% change from baseline in composite standardized uptake value on amyloid PET at week 100

Gantenerumab 225 mg: – 7.52% change in CSF p-tau (< .01) at week 104; – 2.94% change from baseline in CSF t-tau (< .01) at week 104; – 5.37% change from baseline in composite standardized uptake value on amyloid PET at week 100

Safety One adverse event, injection site erythema, had > 5% occurrence and 2X greater than placebo: gantenerumab 105 mg, 10.7%; gantenerumab 225 mg, 13.5%; placebo, 1.1%

Marguerite RoAD Phase 3 double-blind, placebo-controlled parallel-group study (study start date: 2014)

(NCT02051608)

N = 389

50–90 years

Probable mild dementiaa

CSF Aβ1–42 confirmed pathology < 700 pg/mL

Gantenerumab SC Q4W uptitrated to 225 mg at week 28 if no confirmed ARIA (Fig. 3)

Placebo

Change from baseline in ADAS-Cog13 scores at week 104 vs placebo

Change from baseline in ADCS-ADL scores at week 104 vs placebo

Terminated early following SCarlet RoAD futility analysis; study converted to OLE in 2015

Mean time on treatment = 66 weeks

CSF biomarkers in gantenerumab group (n = 12) median % from baseline at 104 weeks:

Aβ(1–40): – 10.03 [– 18.08; – 2.21], = .584

Aβ(1–42): 7.15 [– 8.52; 15.97], = .123

t-tau: – 6.17 [– 12.72; 1.25], = .184

p-tau: – 16.33 [– 22.84; – 4.49], = .053

SCarlet RoAD

Open-label extension study (study start date: 2015)

(NCT01224106)

N = 154

Patients enrolled in the SCarlet RoAD study

(NCT01224106)

Gantenerumab SC Q4W, uptitrated to 1200 mg based on APOE ε4 status (Fig. 3)

Safety

AEs were mostly mild to moderate. 31.2% of patients experienced ISR; one patient discontinued due to ISR, which was reported as mild to moderate.

 

Marguerite RoAD

Open-label extension study (study start date: 2015)

(NCT02051608)

N = 225

Patients enrolled in the Marguerite RoAD study

(NCT02051608)

Gantenerumab SC Q4W uptitrated to 1200 mg based on APOE ε4 carrier status (Fig. 3)

Safety

AEs were mostly mild to moderate. 29.8% of patients experienced ISR; 10.7% of patients discontinued due to AE.

 

SCarlet RoAD OLE

(NCT01224106) and Marguerite RoAD OLE (NCT02051608)

PET substudy: exploratory analyses (study start date: 2015)

(n = 67)

Prodromal ADa, 50–85 years,

CSF Aβ1–42 confirmed pathology

Three cohorts:

1) Pooled SR gantenerumab 105 mg or 225 mg or placebo Q4W (n = 19)

2) MR double-blind placebo (n = 27)

3) MR double-blind active gantenerumab 105 mg or 225 mg (n = 21)

Gantenerumab 1200 mg SC Q4W (Fig. 3)

Change from baseline in mean Aβ PET centiloid values

52 weeks: pooled SR, – 21; MR double-blind placebo, – 42; MR double-blind active, – 48

104 weeks: pooled SR, – 34; MR double-blind placebo, – 71; MR double-blind active, – 62

36 months: pooled SR, – 57.0; MR double-blind placebo, – 90.3; MR double-blind active, – 74.9

At 104 weeks, 51% of patients were below the amyloid positivity threshold.

At the 36-month follow-up (140 weeks total), 80% of participants were below the amyloid positivity threshold.

DIAN-TU-001

Public-private collaboration, randomized, placebo-controlled, multi-arm phase 2/3 trial (study start date: 2012)

(NCT01760005)

N = 142

Asymptomatic or mildly symptomatic participants with DIAD

Gantenerumab SC uptitrated to 1200 mg Q4W (see Fig. 3)

Solanezumab uptitrated to 1600 mg

Placebo

DIAN-MCE evaluated via Bayesian multivariate CPR compared with pooled placebo:

Gantenerumab probability CPR < 1 = 0.144 (no treatment benefit)

No benefit with gantenerumab in the following:

MMRM change from baseline in each component of DIAN-MCE

MMRM change from baseline in CDR-SB MMRM change from baseline in Functional Assessment Scale

Changes in biological endpoints from baseline to year 4: between-group difference (gantenerumab vs placebo)

PiB-PET Aβ: 24.3% (12.7 + 11.6) decrease with gantenerumab(< .001)

CSF total Aβ42: 42.6% (19.3 + 23.3) increase with gantenerumab (< .001)

CSF total tau: 20.6% (15.3 + 5.3) decrease with gantenerumab (< .001)

Phospho-tau181: 32.8% (23.4 + 9.4) decrease with gantenerumab (< .001)

CSF NfL: 2.2% (3.9 – 1.7) slowed increase with gantenerumab (< .05)

Cortical metabolism measured with 18F-FDG-PET (no difference)

Precuneus thickness and hippocampal volume (no difference)

Safety

The most common adverse event with gantenerumab was injection-site reactions (90%; < .0001 vs placebo). No other adverse event occurred statistically significantly more with gantenerumab than placebo.

  1. Abbreviations: 18F-FDG-PET 18F-fluorodexyglucose positron emission tomography, amyloid-beta, Aβ(1–40) amyloid beta protein fragment 1-40, Aβ(1–42) amyloid beta protein fragment 1-42, AD Alzheimer’s disease, ADAS-Cog13 Alzheimer’s Disease Assessment Scale–Cognitive Subscale 13, ADCS-ADL Alzheimer’s Disease Cooperative Study–Activities of Daily Living instrumental subscale, AE adverse event, APOE ε4 apolipoprotein E ε4, ARIA amyloid-related imaging abnormalities, CDR Clinical Dementia Rating, CDR-SB Clinical Dementia Rating Scale–Sum of Boxes, CPR cognitive progression ratio, CSF cerebrospinal fluid, DIAD dominantly inherited Alzheimer’s disease, DIAN-MCE Dominantly Inherited Alzheimer Network–Multivariate Cognitive Endpoint, DIAN-TU Dominantly Inherited Alzheimer Network–Trials Unit, ISR injection site reaction, mg milligram, mL milliliter, MMRM mixed models for repeated measures, MR Marguerite RoAD, NCT National Clinical Trial number, NfL neurofilament light chain, OLE open-label extension, PET positron emission tomography, pg picogram, PiB-PET Pittsburgh compound-B positron emission tomography, p-tau phosphorylated tau, Q4W every 4 weeks, SC subcutaneous, SR SCarlet RoAD, t-tau total tau.
  2. aInternational Working Group Criteria for Prodromal AD [30]