Distinct biosignatures associate with survival after chemoimmunotherapy in a randomized, three-arm phase II study in patients with metastatic pancreatic cancer.

Authors

null

Lacey J. Padrón

Parker Institute for Cancer Immunotherapy, San Francisco, CA

Lacey J. Padrón , Deena M. Maurer , Mark H. O'Hara , Eileen Mary O'Reilly , Robert A. Wolff , Zev A. Wainberg , Andrew H. Ko , George A. Fisher Jr., Osama E. Rahma , Jaclyn P. Lyman , Christopher R. Cabanski , Jia Yu , Shannon M. Pfeiffer , Marko Spasic , Travis J Hollmann , Richard Chen , Jill O'Donnell-Tormey , Samantha Bucktrout , Theresa LaVallee , Robert H. Vonderheide

Organizations

Parker Institute for Cancer Immunotherapy, San Francisco, CA, Hospital of the University of Pennsylvania, Philadelphia, PA, Memorial Sloan Kettering Cancer Center, New York, NY, Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, Ronald Reagan UCLA Medical Center, Los Angeles, CA, University of California San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, Department of Medicine, Stanford University School of Medicine, Stanford, CA, Dana-Farber Cancer Institute, Boston, MA, Memor Sloan-Kettering Cancer Ctr, New York, NY, Cancer Research Institute, New York, NY, MSKCC, New York, NY, Personalis, Inc, Menlo Park, CA, The Parker Institute for Cancer Immunotherapy, San Francisco, CA, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, PA

Research Funding

Other Foundation
Pharmaceutical/Biotech Company

Background: Preclinical and small clinical studies of chemoimmunotherapy for metastatic pancreatic ductal adenocarcinoma (mPDAC) point to a yet unrealized potential of clinically significant immune activation. In our phase II study of the CD40 agonist antibody sotigalimab (sotiga) and/or nivolumab (nivo) with gemcitabine and nab-paclitaxel (chemo), we observed promising improvements in overall survival (OS) in 105 patients with newly diagnosed mPDAC (NCT03214250); the primary endpoint of 1-year OS rate was 57.7% (p = 0.006) in the nivo/chemo arm, 48.1% (p = 0.062) in the sotiga/chemo arm and 41.3% (p = 0.233) in the nivo/sotiga/chemo arm (O’Hara, ASCO 2021) as compared to a historical control of 35%. Here, we report results of multi-omic translational analyses designed to identify signatures predictive of OS benefit. Methods: Longitudinal blood and tumor tissue samples were collected for immune and tumor biomarker analysis. Tumor samples underwent RNA sequencing and multiplex immunofluorescence (mIF). Peripheral blood was analyzed by mass cytometry time of flight (CyTOF), high parameter flow cytometry, and proteomics. Machine learning (ML) algorithms were applied to the data to identify biosignatures related to OS in each arm. Results: Comprehensive multi-omic, multi-parameter immune and tumor biomarker analyses identified distinct pretreatment immune signatures predictive of longer OS specific to nivo/chemo or sotiga/chemo (Table, representative examples). Because patients in each arm received chemotherapy, these and other arm-unique biomarkers suggest a relationship to the immunotherapy rather than chemotherapy in this randomized study. There was evidence of immune exhaustion in the sotiga/nivo/chemo arm that may explain the lack of survival benefit. Conclusions: From in-depth translational and ML analyses of randomized phase II trial of first-line chemoimmunotherapy in mPDAC patients, we identified novel biomarkers that associated with OS distinctly in each arm. Clinical trials in first-line mPDAC exploiting these previously unappreciated biomarkers and aiming to enrich patients for response, are warranted to further advance chemoimmunotherapy in this disease. Clinical trial information: NCT03214250.

Selected pretreatment immune features associated with overall survival benefit.

Immune Feature
Sample
Nivo/Chemo

(p-value, by log rank)
Sotiga/Chemo

(p-value, by log rank)
Lower expression of TNF-α signaling via NFκB hallmark gene signature
Tumor
0.001
Not significant (ns)
Lower expression of the E2F targets hallmark gene signature
Tumor
ns
0.021
Higher frequencies of Tfh cells
Blood
< 0.0001
ns
Higher frequencies of PD-1+ CD39+ CD4 Central Memory T cells
Blood
0.037
ns
Higher frequencies of Tbet+ Eomes+ CD4 T cells
Blood
ns
0.013
Higher frequencies of PD-1+ Tbet+ CD4 T cells
Blood
ns
0.004

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

2022 ASCO Annual Meeting

Session Type

Clinical Science Symposium

Session Title

Can We Begin to Predict Responders to Targeted Therapy in Gastrointestinal Cancer?

Track

Gastrointestinal Cancer—Gastroesophageal, Pancreatic, and Hepatobiliary

Sub Track

Pancreatic Cancer

Clinical Trial Registration Number

NCT03214250

Citation

J Clin Oncol 40, 2022 (suppl 16; abstr 4010)

DOI

10.1200/JCO.2022.40.16_suppl.4010

Abstract #

4010

Abstract Disclosures

Similar Abstracts

First Author: Yan-Shen Shan

Abstract

2024 ASCO Gastrointestinal Cancers Symposium

Associations between BMI, morphomics and survival in 528 patients with metastatic pancreatic cancer.

First Author: Valerie Gunchick