Neoadjuvant selicrelumab, an agonist CD40 antibody, induces changes in the tumor microenvironment in patients with resectable pancreatic cancer

KT Byrne, CB Betts, R Mick, S Sivagnanam… - Clinical Cancer …, 2021 - AACR
KT Byrne, CB Betts, R Mick, S Sivagnanam, DL Bajor, DA Laheru, EG Chiorean, MH O'Hara…
Clinical Cancer Research, 2021AACR
Purpose: CD40 activation is a novel clinical opportunity for cancer immunotherapy. Despite
numerous active clinical trials with agonistic CD40 monoclonal antibodies (mAb), biological
effects and treatment-related modulation of the tumor microenvironment (TME) remain
poorly understood. Patients and Methods: Here, we performed a neoadjuvant clinical trial of
agonistic CD40 mAb (selicrelumab) administered intravenously with or without
chemotherapy to 16 patients with resectable pancreatic ductal adenocarcinoma (PDAC) …
Purpose
CD40 activation is a novel clinical opportunity for cancer immunotherapy. Despite numerous active clinical trials with agonistic CD40 monoclonal antibodies (mAb), biological effects and treatment-related modulation of the tumor microenvironment (TME) remain poorly understood.
Patients and Methods
Here, we performed a neoadjuvant clinical trial of agonistic CD40 mAb (selicrelumab) administered intravenously with or without chemotherapy to 16 patients with resectable pancreatic ductal adenocarcinoma (PDAC) before surgery followed by adjuvant chemotherapy and CD40 mAb.
Results
The toxicity profile was acceptable, and overall survival was 23.4 months (95% confidence interval, 18.0–28.8 months). Based on a novel multiplexed immunohistochemistry platform, we report evidence that neoadjuvant selicrelumab leads to major differences in the TME compared with resection specimens from treatment-naïve PDAC patients or patients given neoadjuvant chemotherapy/chemoradiotherapy only. For selicrelumab-treated tumors, 82% were T-cell enriched, compared with 37% of untreated tumors (P = 0.004) and 23% of chemotherapy/chemoradiation-treated tumors (P = 0.012). T cells in both the TME and circulation were more active and proliferative after selicrelumab. Tumor fibrosis was reduced, M2-like tumor-associated macrophages were fewer, and intratumoral dendritic cells were more mature. Inflammatory cytokines/sec CXCL10 and CCL22 increased systemically after selicrelumab.
Conclusions
This unparalleled examination of CD40 mAb therapeutic mechanisms in patients provides insights for design of subsequent clinical trials targeting CD40 in cancer.
AACR