A phase 1b study of the anti-PD-1 monoclonal antibody BGB-A317 (A317) in combination with the PARP inhibitor BGB-290 (290) in advanced solid tumors.

Authors

null

Michael Friedlander

The Prince of Wales Hospital, Randwick, Australia

Michael Friedlander , Tarek Meniawy , Ben Markman , Linda R. Mileshkin , Paul R. Harnett , Michael Millward , Joanne Lundy , Alison E. Freimund , Christie Norris , Song Mu , John Wu , Virginia E. Paton , Lai Wang , Bo Gao

Organizations

The Prince of Wales Hospital, Randwick, Australia, Sir Charles Gairdner Hospital, Perth, Australia, Monash Cancer Centre, Melbourne, Australia, Division of Cancer Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia, Crown Princess Mary Cancer Centre Westmead, Sydney, Australia, School of Medicine and Pharmacology, Nedlands, WA, Australia, Launceston Hospital, Mordialloc, Australia, St Vincent's Hospital, Darlinghurst, Australia, Prince of Wales Hospital, Randwick, Australia, BeiGene, Fort Lee, NJ, Beigene USA, INC., Fort Lee, NJ, Genentech, Inc., San Francisco, CA, BeiGene, Beijing, China, Westmead Hospital, Westmead, NSW, Australia

Research Funding

Pharmaceutical/Biotech Company

Background: The release of tumor-associated antigens may enhance the response to immunotherapy. BGB-A317, a humanized IgG4 variant monoclonal antibody engineered to have no Fc gamma receptor binding, targets the programmed cell death-1 (PD-1) receptor. It is being developed in solid and hematologic malignancies at a dose of 200 mg IV Q3W. BGB-290, a potent inhibitor of PARP 1/2, is hypothesized to promote neoantigen release that will potentially increase the efficacy of BGB-A317. A phase 1 study identified 60mg BID as the recommended Phase 2 dose (RP2D) for BGB-290. This study consists of initial dose escalation to determine the maximum-tolerated dose (MTD), safety, PK profile, and preliminary anti-tumor activity of the combination, followed by expansion into ovarian, breast, prostate, gastric, bladder, pancreatic and small cell lung cancers. Methods: Cohorts of 6 -12 pts with advanced solid tumors were treated in each of 5 planned dose levels (DLs). In DLs 1-3, BGB-290 doses ranged between 20-60mg PO BID with BGB-A317 2mg/kg IV Q3W. In DLs 4 - 5, BGB-290 doses were 40 or 60 mg BID; A317 was given at 200 mg IV Q3W based on PK data from a single agent Phase 1 study. Results: As of 16 Jan 2017, 38 pts [median age 59 years (34-75)] were treated in DLs 1-4; enrollment to DL5 is ongoing. One DLT of persistent Gr 2 nausea was reported in DL 4. The most common adverse event (AE) considered related to both study drugs was fatigue (10.5%). Immune-related AEs were Gr 3 hypophysitis (n = 1), Gr 3 or 4 autoimmune hepatitis(n = 2), and Gr 2 elevated AST/ALT (n = 1). Decreases in tumor burden have been observed in 16 pts; 7 achieved a PR (5 with ovarian and one each with uterine and pancreatic cancer) and one CR was observed in ovarian cancer. Six pts had SD for > 6 months including 2 pts with pancreatic cancer who received BGB-A317+BGB-290 for 189 and 281 days. Plasma/serum exposure of BGB-290 and BGB-A317 were consistent with those in single-agent trials. Conclusions: BGB290 and BGB-A317 can be combined. Dose expansion in multiple tumor types is planned to commence in 2017 once the RP2D is determined. Clinical trial information: NCT02660034

Disclaimer

This material on this page is ©2024 American Society of Clinical Oncology, all rights reserved. Licensing available upon request. For more information, please contact licensing@asco.org

Abstract Details

Meeting

2017 ASCO Annual Meeting

Session Type

Poster Discussion Session

Session Title

Developmental Therapeutics—Immunotherapy

Track

Developmental Therapeutics and Translational Research,Immunotherapy

Sub Track

Immune Checkpoint Inhibitors

Clinical Trial Registration Number

NCT02660034

Citation

J Clin Oncol 35, 2017 (suppl; abstr 3013)

DOI

10.1200/JCO.2017.35.15_suppl.3013

Abstract #

3013

Poster Bd #

108

Abstract Disclosures

Similar Abstracts

First Author: Michael Friedlander

First Author: Michael Friedlander