Your browser doesn't support javascript.
loading
Neoadjuvant Chemotherapy Is Associated with Altered Immune Cell Infiltration and an Anti-Tumorigenic Microenvironment in Resected Pancreatic Cancer.
Dias Costa, Andressa; Väyrynen, Sara A; Chawla, Akhil; Zhang, Jinming; Väyrynen, Juha P; Lau, Mai Chan; Williams, Hannah L; Yuan, Chen; Morales-Oyarvide, Vicente; Elganainy, Dalia; Singh, Harshabad; Cleary, James M; Perez, Kimberly; Ng, Kimmie; Freed-Pastor, William; Mancias, Joseph D; Dougan, Stephanie K; Wang, Jiping; Rubinson, Douglas A; Dunne, Richard F; Kozak, Margaret M; Brais, Lauren; Reilly, Emma; Clancy, Thomas; Linehan, David C; Chang, Daniel T; Hezel, Aram F; Koong, Albert C; Aguirre, Andrew J; Wolpin, Brian M; Nowak, Jonathan A.
Affiliation
  • Dias Costa A; Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
  • Väyrynen SA; Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
  • Chawla A; Department of Surgery, Northwestern Medicine Regional Medical Group, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
  • Zhang J; Robert H. Lurie Comprehensive Cancer Center, Chicago, Illinois.
  • Väyrynen JP; Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
  • Lau MC; Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
  • Williams HL; Program in Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts.
  • Yuan C; Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland.
  • Morales-Oyarvide V; Program in Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts.
  • Elganainy D; Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
  • Singh H; Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
  • Cleary JM; Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
  • Perez K; Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
  • Ng K; Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
  • Freed-Pastor W; Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
  • Mancias JD; Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
  • Dougan SK; Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
  • Wang J; Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
  • Rubinson DA; Department of Radiation Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Boston, Massachusetts.
  • Dunne RF; Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
  • Kozak MM; Department of Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts.
  • Brais L; Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
  • Reilly E; Division of Hematology and Oncology, Department of Medicine, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York.
  • Clancy T; Department of Radiation Oncology, Stanford Cancer Institute, Stanford, California.
  • Linehan DC; Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
  • Chang DT; Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
  • Hezel AF; Department of Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts.
  • Koong AC; Department of General Surgery, University of Rochester Medical Center, Rochester, New York.
  • Aguirre AJ; Department of Radiation Oncology, Stanford Cancer Institute, Stanford, California.
  • Wolpin BM; Department of Cell, Developmental & Cancer Biology, Oregon Health and Science University, Portland, Oregon.
  • Nowak JA; Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
Clin Cancer Res ; 28(23): 5167-5179, 2022 12 01.
Article in En | MEDLINE | ID: mdl-36129461
ABSTRACT

PURPOSE:

Neoadjuvant chemotherapy is increasingly administered to patients with resectable or borderline resectable pancreatic ductal adenocarcinoma (PDAC), yet its impact on the tumor immune microenvironment is incompletely understood.

DESIGN:

We employed quantitative, spatially resolved multiplex immunofluorescence and digital image analysis to identify T-cell subpopulations, macrophage polarization states, and myeloid cell subpopulations in a multi-institution cohort of up-front resected primary tumors (n = 299) and in a comparative set of resected tumors after FOLFIRINOX-based neoadjuvant therapy (n = 36) or up-front surgery (n = 30). Multivariable-adjusted Cox proportional hazards models were used to evaluate associations between the immune microenvironment and patient outcomes.

RESULTS:

In the multi-institutional resection cohort, immune cells exhibited substantial heterogeneity across patient tumors and were located predominantly in stromal regions. Unsupervised clustering using immune cell densities identified four main patterns of immune cell infiltration. One pattern, seen in 20% of tumors and characterized by abundant T cells (T cell-rich) and a paucity of immunosuppressive granulocytes and macrophages, was associated with improved patient survival. Neoadjuvant chemotherapy was associated with a higher CD8CD4 ratio, greater M1M2-polarized macrophage ratio, and reduced CD15+ARG1+ immunosuppressive granulocyte density. Within neoadjuvant-treated tumors, 72% showed a T cell-rich pattern with low immunosuppressive granulocytes and macrophages. M1-polarized macrophages were located closer to tumor cells after neoadjuvant chemotherapy, and colocalization of M1-polarized macrophages and tumor cells was associated with greater tumor pathologic response and improved patient survival.

CONCLUSIONS:

Neoadjuvant chemotherapy with FOLFIRINOX shifts the PDAC immune microenvironment toward an anti-tumorigenic state associated with improved patient survival.
Subject(s)

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Pancreatic Neoplasms / Adenocarcinoma / Carcinoma, Pancreatic Ductal Type of study: Risk_factors_studies Limits: Humans Language: En Journal: Clin Cancer Res Journal subject: NEOPLASIAS Year: 2022 Document type: Article

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Pancreatic Neoplasms / Adenocarcinoma / Carcinoma, Pancreatic Ductal Type of study: Risk_factors_studies Limits: Humans Language: En Journal: Clin Cancer Res Journal subject: NEOPLASIAS Year: 2022 Document type: Article