Gastroenterology

Gastroenterology

Volume 145, Issue 5, November 2013, Pages 1121-1132
Gastroenterology

Original Research
Full Report: Basic and Translational—Pancreas
Activated Pancreatic Stellate Cells Sequester CD8+ T Cells to Reduce Their Infiltration of the Juxtatumoral Compartment of Pancreatic Ductal Adenocarcinoma

https://doi.org/10.1053/j.gastro.2013.07.025Get rights and content

Background & Aims

Pancreatic ductal adenocarcinoma (PDAC) is characterized by a prominent desmoplastic microenvironment that contains many different immune cells. Activated pancreatic stellate cells (PSCs) contribute to the desmoplasia. We investigated whether distinct stromal compartments are differentially infiltrated by different types of immune cells.

Methods

We used tissue microarray analysis to compare immune cell infiltration of different pancreaticobiliary diseased tissues (PDAC, ampullary carcinoma, cholangiocarcinoma, mucinous cystic neoplasm, chronic inflammation, and chronic pancreatitis) and juxtatumoral stromal (<100 μm from tumor) and panstromal compartments. We investigated the association between immune infiltrate and patient survival times. We also analyzed T-cell migration and tumor infiltration in LSL-KrasG12D/+; LSL-Trp53R172H/+; Pdx-1-Cre (KPC) mice and the effects of all-trans retinoic acid (ATRA) on these processes.

Results

Juxtatumoral compartments in PDAC samples from 2 independent groups of patients contained increased numbers of myeloperoxidase+ and CD68+ cells compared with panstromal compartments. However, juxtatumoral compartments of PDACs contained fewer CD8+, FoxP3+, CD56+, or CD20+ cells than panstromal compartments, a distinction absent in ampullary carcinomas and cholangiocarcinomas. Patients with PDACs that had high densities of CD8+ T cells in the juxtatumoral compartment had longer survival times than patients with lower densities. In KPC mice, administration of ATRA, which renders PSCs quiescent, increased numbers of CD8+ T cells in juxtatumoral compartments. We found that activated PSCs express cytokines, chemokines, and adhesion molecules that regulate T-cell migration. In vitro migration assays showed that CD8+ T cells, from patients with PDAC, had increased chemotaxis toward activated PSCs, which secrete CXCL12, compared with quiescent PSCs or tumor cells. These effects could be reversed by knockdown of CXCL12 or treatment of PSCs with ATRA.

Conclusions

Based on studies of human PDAC samples and KPC mice, activated PSCs appear to reduce migration of CD8+ T cells to juxtatumoral stromal compartments, preventing their access to cancer cells. Deregulated signaling by activated PSCs could prevent an effective antitumor immune response.

Section snippets

Tissue Microarray, Staining, and Analysis

Tissue microarrays were constructed with pancreatic tissues collected at cancer resection or biopsy (Supplementary Tables 1 and 2) at Barts Health NHS Trust (City and East London Research Ethics Committee 07/0705/87) as described previously.14 Regions of tumor, stroma, and normal pancreas were marked on H&E-stained slides of the donor tissue blocks, and three 1-mm cores of each region were sampled per patient using the Tissue Arrayer Minicore 3 (Alphelys, Plaisir, France). The tissue

Immune Cell Infiltration in Pancreaticobiliary Diseases

We performed an unbiased, automated evaluation of immune cell infiltrate for pancreaticobiliary cancers such as PDAC, ampullary carcinoma (AC), and cholangiocarcinoma (CC) using dedicated TMAs. These tumors were compared alongside other pancreaticobiliary diseases (PBDs), including borderline malignant conditions such as mucinous cystic neoplasm, chronic inflammation such as chronic pancreatitis, and normal donor control TMAs (Supplementary Tables 1 and 2). Our immune cell density measurements

Discussion

Patients with PDAC have a 5-year survival rate of 3% to 5%, making it the PBD with the worst prognosis.21 In this report, we performed a comprehensive global as well as stromal compartment–specific comparison of immune infiltrate in a variety of PBDs. We showed an increasing immune cell infiltrate from normal (healthy pancreas tissue) to inflammatory (chronic pancreatitis), borderline (mucinous cystic neoplasm), and malignant (AC, PDAC, and CC) diseases. To our knowledge, we identified for the

Acknowledgments

The authors thank Dr Christine Feig (Cambridge Research Institute) and Prof David A. Tuveson (Cold Spring Harbor Laboratory) for helpful comments regarding murine experiments, members of our laboratories for helpful discussions and suggestions, Dr Lisa Mears (Department of Pathology, Barts Health NHS Trust) and Dr Moffadal Moonim (Department of Pathology, St Thomas' Hospital) for human PDAC samples, and Prof Federica Marelli-Berg for her critical insight.

References (38)

  • R.D. Schreiber et al.

    Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion

    Science

    (2011)
  • M.D. Vesely et al.

    Natural innate and adaptive immunity to cancer

    Annu Rev Immunol

    (2011)
  • N.P. Restifo et al.

    Adoptive immunotherapy for cancer: harnessing the T cell response

    Nat Rev Immunol

    (2012)
  • H. Matsushita et al.

    Cancer exome analysis reveals a T-cell-dependent mechanism of cancer immunoediting

    Nature

    (2011)
  • L. Zhang et al.

    Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer

    N Engl J Med

    (2003)
  • J. Galon et al.

    Type, density, and location of immune cells within human colorectal tumors predict clinical outcome

    Science

    (2006)
  • C.E. Clark et al.

    Dynamics of the immune reaction to pancreatic cancer from inception to invasion

    Cancer Res

    (2007)
  • D. Nummer et al.

    Role of tumor endothelium in CD4+ CD25+ regulatory T cell infiltration of human pancreatic carcinoma

    J Natl Cancer Inst

    (2007)
  • A. Neesse et al.

    Stromal biology and therapy in pancreatic cancer

    Gut

    (2011)
  • Cited by (406)

    View all citing articles on Scopus

    Author names in bold designate shared co-first authorship.

    Conflicts of interest The authors disclose no conflicts.

    Funding Supported by a PhD studentship from Cross-River University of Technology, Nigeria (to A.E.-O), the National Institutes of Health and Cancer Research UK (J.G.G.), Barts and the London Charity (A.J.C., J.W., and A.G.R.), the European Hematology Association (A.G.R.), and the National Institute for Health Research (H.M.K.).

    View full text