Back to Search Start Over

Vaccine-Induced Intratumoral Lymphoid Aggregates Correlate with Survival Following Treatment with a Neoadjuvant and Adjuvant Vaccine in Patients with Resectable Pancreatic Adenocarcinoma

Authors :
Jessica Hoare
Katrina Purtell
Elizabeth A. Sugar
Raka Bhattacharya
Arsen Osipov
Daniel A. Laheru
Katherine M. Bever
Rose Parkinson
Beth Onner
Matthew J. Weiss
Jin He
Ding Ding
Qingfeng Zhu
Elizabeth M. Jaffee
Guanglan Mo
Elizabeth D. Thompson
Amy Hacker-Prietz
Victoria M. Kim
Martin A. Makary
Hongyan Cai
Barish H. Edil
Richard D. Schulick
Robert A. Anders
John L. Cameron
Joseph M. Herman
Lindsey Manos
Nilofer S. Azad
Christopher L. Wolfgang
Dung T. Le
Dwayne L. Thomas
Jennifer N. Durham
Kevin C. Soares
Carol Judkins
Alex B. Blair
Ana De Jesus-Acosta
Sara Solt
Lei Zheng
Source :
Clin Cancer Res
Publication Year :
2021
Publisher :
American Association for Cancer Research (AACR), 2021.

Abstract

Purpose: Immunotherapy is currently ineffective for nearly all pancreatic ductal adenocarcinomas (PDAC), largely due to its tumor microenvironment (TME) that lacks antigen-experienced T effector cells (Teff). Vaccine-based immunotherapies are known to activate antigen-specific Teffs in the peripheral blood. To evaluate the effect of vaccine therapy on the PDAC TME, we designed a neoadjuvant and adjuvant clinical trial of an irradiated, GM-CSF-secreting, allogeneic PDAC vaccine (GVAX). Patients and Methods: Eighty-seven eligible patients with resectable PDAC were randomly assigned (1:1:1) to receive GVAX alone or in combination with two forms of low-dose cyclophosphamide. Resected tumors following neoadjuvant immunotherapy were assessed for the formation of tertiary lymphoid aggregates (TLA) in response to treatment. The clinical endpoints are disease-free survival (DFS) and overall survival (OS). Results: The neoadjuvant treatment with GVAX either alone or with two forms of low-dose cyclophosphamide is safe and feasible without adversely increasing the surgical complication rate. Patients in Arm A who received neoadjuvant and adjuvant GVAX alone had a trend toward longer median OS (35.0 months) than that (24.8 months) in the historical controls who received adjuvant GVAX alone. However, Arm C, who received low-dose oral cyclophosphamide in addition to GVAX, had a significantly shorter DFS than Arm A. When comparing patients with OS > 24 months to those with OS < 15 months, longer OS was found to be associated with higher density of intratumoral TLA. Conclusions: It is safe and feasible to use a neoadjuvant immunotherapy approach for PDACs to evaluate early biologic responses. In-depth analysis of TLAs is warranted in future neoadjuvant immunotherapy clinical trials.

Details

ISSN :
15573265 and 10780432
Volume :
27
Database :
OpenAIRE
Journal :
Clinical Cancer Research
Accession number :
edsair.doi.dedup.....534abf5113ad35ad967526b033d79e86