SUVR in week 69 compared to SUVR at baseline using different processing methods and reference regions: MNI-CB (A and D), BAI-WM (B and E), and MNI-WM (C and F) in both the low-dose SC (ACC) and high-dose IV (D CF) cohorts

SUVR in week 69 compared to SUVR at baseline using different processing methods and reference regions: MNI-CB (A and D), BAI-WM (B and E), and MNI-WM (C and F) in both the low-dose SC (ACC) and high-dose IV (D CF) cohorts. high-dose IV (B) cohorts. (PDF 65 kb) 13195_2018_424_MOESM4_ESM.pdf (74K) GUID:?6B343B6A-DCDA-40F5-BFAB-20A7BE5EBDD3 Additional file 5: Table S1. Summary of ARIA events. Summary of ARIA events 4-Pyridoxic acid in the low-dose SC cohort, high-dose IV cohort, and all patients. (PDF 40 kb) 13195_2018_424_MOESM5_ESM.pdf (57K) GUID:?12A4CD6F-D9A4-4ED7-B94B-46B71F0BE629 Additional file 6: Figure S4. CSF A(1C42) crenezumab correlation analysis. Correlation analysis of change in CSF A(1C42) from baseline and crenezumab concentrations at week 69 in patients in the low-dose SC cohort (circles) and high-dose cohort (triangles). (PDF 98 kb) 13195_2018_424_MOESM6_ESM.pdf (108K) GUID:?7C53D2A7-1131-4BAA-BA87-C33CED4BB960 Additional file 7: Figure S5. CDR-SB. Change from baseline (BL) of CDR-SB score in patients with mild-to-moderate AD (A and C) or moderate AD (B and D) in the low-dose SC (A and B) and high-dose IV (C and D) cohorts. (PDF 112 kb) 13195_2018_424_MOESM7_ESM.pdf (122K) GUID:?29FC4C9F-2169-4AD2-BE74-79F8184C788A Data Availability StatementThe datasets analyzed during the current study are available from the corresponding author on reasonable request. Qualified researchers may request access to individual patient-level data through the clinical study data request platform (www.clinicalstudydatarequest.com). Further details on Roches criteria for eligible studies are available here at https://clinicalstudydatarequest.com/Study-Sponsors/Study-Sponsors-Roche.aspx. For further details on Roches Global Policy on the Sharing of Clinical Information and how to request access to related clinical study documents, see https://www.roche.com/research_and_development/who_we_are_how_we_work/clinical_trials/our_commitment_to_data_sharing.htm. Abstract Background We investigated the effect of crenezumab, a humanized anti-amyloid-beta (A) immunoglobulin (Ig)G4 monoclonal antibody, on biomarkers of amyloid pathology, neurodegeneration, and disease progression in patients with mild-to-moderate Alzheimers disease (AD). Methods This double-blind, placebo-controlled, randomized phase II study enrolled patients with mild-to-moderate AD and a Mini-Mental State Examination (MMSE) score of 18C26. In part 1 of the study, patients were 2:1 randomized to receive low-dose subcutaneous (SC) 300?mg crenezumab every 2?weeks (q2w) or placebo for 68?weeks; in part 2, patients were 2:1 randomized to receive high-dose intravenous (IV) 15?mg/kg crenezumab every 4?weeks (q4w) or placebo for 68?weeks. The primary endpoint was change in amyloid burden from baseline to week 69 assessed by florbetapir positron emission tomography (PET) in the modified intent-to-treat population. Secondary endpoints were change from baseline to week 69 in cerebrospinal fluid (CSF) biomarkers and fluorodeoxyglucose PET, and change from baseline to week 73 in 12-point Alzheimers Disease Assessment Scale cognitive subscale (ADAS-Cog12) and Clinical Dementia Rating Sum of Boxes (CDR-SB). Safety was assessed in patients who received at least one dose of study treatment. Results From August 2011 to September 2012, 91 patients were enrolled and randomized (low-dose SC cohort: crenezumab (The IgG4 Rabbit Polyclonal to SFRS4 backbone confers reduced binding of Fc-gamma receptors (FcRs) compared with an IgG1 backbone, and was shown in vitro to preserve FcR-mediated microglial phagocytosis and 4-Pyridoxic acid removal of oligomers while minimizing FcR-mediated inflammatory activation of microglia and release of proinflammatory cytokines [4]. This reduced effector function of crenezumab is usually hypothesized to reduce cytokine-mediated neurotoxicity and reduce inflammation at sites of A plaque deposition, particularly involving blood vessels. The latter was hypothesized to increase the risk of drug-induced amyloid-related imaging abnormalities (ARIA) in studies investigating anti-amyloid antibodies with full effector function [4]. This phase II, multicenter, randomized, double-blind, placebo-controlled, parallel-group study was designed to evaluate the effects of crenezumab on brain amyloid plaque load as assessed by florbetapir positron emission tomography (PET) and other biomarkers in patients with mild-to-moderate AD (ABE4955g, BLAZE; “type”:”clinical-trial”,”attrs”:”text”:”NCT01397578″,”term_id”:”NCT01397578″NCT01397578). The prespecified analysis of the florbetapir PET data used a cerebellar reference 4-Pyridoxic acid region for calculating standard uptake value ratios (SUVRs). Although this was the widely accepted reference region at the time this study was designed, more recent evidence published near the conclusion of this study showed that using a subcortical white matter reference region reduces longitudinal.

This entry was posted in DNA-Dependent Protein Kinase. Bookmark the permalink.