Stratification of Pancreatic Ductal Adenocarcinoma: Combinatorial Genetic, Stromal, and Immunologic Markers

Purpose: Pancreatic ductal adenocarcinoma (PDAC) is associated with an immunosuppressive milieu that supports immune system evasion and disease progression. Here, we interrogated genetic, stromal, and immunologic features of PDAC to delineate impact on prognosis and means to more effectively employ immunotherapy. Experimental Design: A cohort of 109 PDAC cases annotated for overall survival was utilized as a primary discovery cohort. Gene expression analysis defined immunologic subtypes of PDAC that were confirmed in the Cancer Genome Atlas dataset. Stromal and metabolic characteristics of PDAC cases were evaluated by histologic analysis and immunostaining. Enumeration of lymphocytes, as well as staining for CD8, FOXP3, CD68, CD163, PDL1, and CTLA4 characterized immune infiltrate. Neoantigens were determined by analysis of whole-exome sequencing data. Random-forest clustering was employed to define multimarker subtypes, with univariate and multivariate analyses interrogating prognostic significance. Results: PDAC cases exhibited distinct stromal phenotypes that were associated with prognosis, glycolytic and hypoxic biomarkers, and immune infiltrate composition. Immune infiltrate was diverse among PDAC cases and enrichment for M2 macrophages and select immune checkpoints regulators were specifically associated with survival. Composite analysis with neoantigen burden, immunologic, and stromal features defined novel subtypes of PDAC that could have bearing on sensitivity to immunologic therapy approaches. In addition, a subtype with low levels of neoantigens and minimal lymphocyte infiltrate was associated with improved overall survival. Conclusions: The mutational burden of PDAC is associated with distinct immunosuppressive mechanisms that are conditioned by the tumor stromal environment. The defined subtypes have significance for utilizing immunotherapy in the treatment of PDAC. Clin Cancer Res; 23(15); 4429–40. ©2017 AACR.

[1]  S. Haferkamp,et al.  LDHA-Associated Lactic Acid Production Blunts Tumor Immunosurveillance by T and NK Cells. , 2016, Cell metabolism.

[2]  S. Pandol,et al.  Macrophages and pancreatic ductal adenocarcinoma. , 2016, Cancer letters.

[3]  E. Knudsen,et al.  Unique metabolic features of pancreatic cancer stroma: relevance to the tumor compartment, prognosis, and invasive potential , 2016, Oncotarget.

[4]  T. Chan,et al.  Personalized Oncology Meets Immunology: The Path toward Precision Immunotherapy. , 2016, Cancer discovery.

[5]  Jaime Rodriguez-Canales,et al.  An Expression Signature as an Aid to the Histologic Classification of Non–Small Cell Lung Cancer , 2016, Clinical Cancer Research.

[6]  R. Gillies,et al.  Neutralization of Tumor Acidity Improves Antitumor Responses to Immunotherapy. , 2016, Cancer research.

[7]  C. Link,et al.  Pancreatic cancer: Update on immunotherapies and algenpantucel-L , 2016, Human vaccines & immunotherapeutics.

[8]  R. Gibbs,et al.  Genomic analyses identify molecular subtypes of pancreatic cancer , 2016, Nature.

[9]  J. Hutcheson,et al.  Immunologic and Metabolic Features of Pancreatic Ductal Adenocarcinoma Define Prognostic Subtypes of Disease , 2016, Clinical Cancer Research.

[10]  C. Link,et al.  Algenpantucel-L immunotherapy in pancreatic adenocarcinoma. , 2016, Immunotherapy.

[11]  Hai Hu,et al.  The M2 phenotype of tumor-associated macrophages in the stroma confers a poor prognosis in pancreatic cancer , 2016, Tumor Biology.

[12]  Somnath Mukherjee,et al.  The prognostic role of desmoplastic stroma in pancreatic ductal adenocarcinoma , 2015, Oncotarget.

[13]  R. Salgia,et al.  Randomized Phase Ib/II Study of Gemcitabine Plus Placebo or Vismodegib, a Hedgehog Pathway Inhibitor, in Patients With Metastatic Pancreatic Cancer , 2015, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[14]  Bin Shang,et al.  Prognostic value of tumor-infiltrating FoxP3+ regulatory T cells in cancers: a systematic review and meta-analysis , 2015, Scientific Reports.

[15]  R. Schreiber,et al.  Metabolic Competition in the Tumor Microenvironment Is a Driver of Cancer Progression , 2015, Cell.

[16]  Jen Jen Yeh,et al.  Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma , 2015, Nature Genetics.

[17]  G. Freeman,et al.  Combination cancer immunotherapy and new immunomodulatory targets , 2015, Nature Reviews Drug Discovery.

[18]  D. V. Von Hoff,et al.  Orchestrating the Tumor Microenvironment to Improve Survival for Patients With Pancreatic Cancer: Normalization, Not Destruction , 2015, Cancer journal.

[19]  Antoni Ribas,et al.  Classifying Cancers Based on T-cell Infiltration and PD-L1. , 2015, Cancer research.

[20]  Michael A. Choti,et al.  Whole-exome sequencing of pancreatic cancer defines genetic diversity and therapeutic targets , 2015, Nature Communications.

[21]  D. Fearon,et al.  T cell exclusion, immune privilege, and the tumor microenvironment , 2015, Science.

[22]  P. Sharma,et al.  The future of immune checkpoint therapy , 2015, Science.

[23]  T. Schumacher,et al.  Neoantigens in cancer immunotherapy , 2015, Science.

[24]  S. Rosenberg,et al.  Adoptive cell transfer as personalized immunotherapy for human cancer , 2015, Science.

[25]  E. Jaffee,et al.  The prognostic value of stroma in pancreatic cancer in patients receiving adjuvant therapy. , 2015, HPB : the official journal of the International Hepato Pancreato Biliary Association.

[26]  J. Kench,et al.  Whole genomes redefine the mutational landscape of pancreatic cancer , 2015, Nature.

[27]  Caroline H. Diep,et al.  Desmoplasia in Primary Tumors and Metastatic Lesions of Pancreatic Cancer , 2015, Clinical Cancer Research.

[28]  R. Deberardinis,et al.  MCT4 defines a glycolytic subtype of pancreatic cancer with poor prognosis and unique metabolic dependencies. , 2014, Cell reports.

[29]  J. Wolchok,et al.  Genetic basis for clinical response to CTLA-4 blockade in melanoma. , 2014, The New England journal of medicine.

[30]  Molin Wang,et al.  Prognostic value of tumor-infiltrating lymphocytes in triple-negative breast cancers from two phase III randomized adjuvant breast cancer trials: ECOG 2197 and ECOG 1199. , 2014, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[31]  S Michiels,et al.  Tumor infiltrating lymphocytes are prognostic in triple negative breast cancer and predictive for trastuzumab benefit in early breast cancer: results from the FinHER trial. , 2014, Annals of oncology : official journal of the European Society for Medical Oncology.

[32]  R. Hruban,et al.  Immunotherapy Converts Nonimmunogenic Pancreatic Tumors into Immunogenic Foci of Immune Regulation , 2014, Cancer Immunology Research.

[33]  Stephen A. Sastra,et al.  Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. , 2014, Cancer cell.

[34]  J. Wolchok,et al.  Immune modulation in cancer with antibodies. , 2014, Annual review of medicine.

[35]  Derek S. Chan,et al.  Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti–PD-L1 immunotherapy in pancreatic cancer , 2013, Proceedings of the National Academy of Sciences.

[36]  Henning Hermjakob,et al.  The Reactome pathway knowledgebase , 2013, Nucleic Acids Res..

[37]  J. Pouysségur,et al.  Disrupting proton dynamics and energy metabolism for cancer therapy , 2013, Nature Reviews Cancer.

[38]  Z. Husain,et al.  Tumor-Derived Lactate Modifies Antitumor Immune Response: Effect on Myeloid-Derived Suppressor Cells and NK Cells , 2013, The Journal of Immunology.

[39]  M. Tempero,et al.  Therapeutic advances in pancreatic cancer. , 2013, Gastroenterology.

[40]  Jonathan B. Mitchem,et al.  Targeting tumor-infiltrating macrophages decreases tumor-initiating cells, relieves immunosuppression, and improves chemotherapeutic responses. , 2013, Cancer research.

[41]  J. Berlin,et al.  Changing the Way We Do Business: Recommendations to Accelerate Biomarker Development in Pancreatic Cancer , 2013, Clinical Cancer Research.

[42]  C. Payne,et al.  Tumor-targeted TNFα stabilizes tumor vessels and enhances active immunotherapy , 2012, Proceedings of the National Academy of Sciences.

[43]  A. Corti,et al.  Targeting TNF-α to Neoangiogenic Vessels Enhances Lymphocyte Infiltration in Tumors and Increases the Therapeutic Potential of Immunotherapy , 2012, The Journal of Immunology.

[44]  Drew A. Torigian,et al.  CD40 Agonists Alter Tumor Stroma and Show Efficacy Against Pancreatic Carcinoma in Mice and Humans , 2011, Science.

[45]  J. Rathmell,et al.  Cutting Edge: Distinct Glycolytic and Lipid Oxidative Metabolic Programs Are Essential for Effector and Regulatory CD4+ T Cell Subsets , 2011, The Journal of Immunology.

[46]  Malte Buchholz,et al.  Stromal biology and therapy in pancreatic cancer , 2010, Gut.

[47]  R. Vonderheide,et al.  Immunosurveillance of pancreatic adenocarcinoma: insights from genetically engineered mouse models of cancer. , 2009, Cancer letters.

[48]  David Allard,et al.  Inhibition of Hedgehog Signaling Enhances Delivery of Chemotherapy in a Mouse Model of Pancreatic Cancer , 2009, Science.

[49]  Murray Korc,et al.  Pancreatic cancer-associated stroma production. , 2007, American journal of surgery.

[50]  Helmut Friess,et al.  Pancreatic cancer microenvironment , 2007, International journal of cancer.

[51]  P. Murawa,et al.  Erlotinib Plus Gemcitabine Compared With Gemcitabine Alone in Patients With Advanced Pancreatic Cancer: A Phase III Trial of the National Cancer Institute of Canada Clinical Trials Group , 2023, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[52]  D. V. Von Hoff,et al.  Tumor-stroma interactions in pancreatic ductal adenocarcinoma , 2007, Molecular Cancer Therapeutics.

[53]  G. Mills,et al.  Reverse phase protein array: validation of a novel proteomic technology and utility for analysis of primary leukemia specimens and hematopoietic stem cells , 2006, Molecular Cancer Therapeutics.

[54]  S. Quezada,et al.  Principles and use of anti-CTLA4 antibody in human cancer immunotherapy. , 2006, Current opinion in immunology.

[55]  John Condeelis,et al.  Macrophages: Obligate Partners for Tumor Cell Migration, Invasion, and Metastasis , 2006, Cell.

[56]  G. Coukos,et al.  Regulatory CD4(+)CD25(+) T cells in tumors from patients with early-stage non-small cell lung cancer and late-stage ovarian cancer. , 2001, Cancer research.

[57]  G. Prendergast,et al.  Cancer Vaccines: A Brief Overview. , 2016, Methods in molecular biology.