Pancreatic Ductal Adenocarcinoma: A Strong Imbalance of Good and Bad Immunological Cops in the Tumor Microenvironment

Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive and lethal cancers with very few available treatments. For many decades, gemcitabine was the only treatment for patients with PDAC. A recent attempt to improve patient survival by combining this chemotherapy with FOLFIRINOX and nab-paclitaxel failed and instead resulted in increased toxicity. Novel therapies are urgently required to improve PDAC patient survival. New treatments in other cancers such as melanoma, non-small-cell lung cancer, and renal cancer have emerged, based on immunotherapy targeting the immune checkpoints cytotoxic T-lymphocyte-associated antigen 4 or programmed death 1 ligand. However, the first clinical trials using such immune checkpoint inhibitors in PDAC have had limited success. Resistance to immunotherapy in PDAC remains unclear but could be due to tissue components (cancer-associated fibroblasts, desmoplasia, hypoxia) and to the imbalance between immunosuppressive and effector immune populations in the tumor microenvironment. In this review, we analyzed the presence of “good and bad immunological cops” in PDAC and discussed the significance of changes in their balance.

[1]  M. Azuma,et al.  Clinical Significance and Therapeutic Potential of the Programmed Death-1 Ligand/Programmed Death-1 Pathway in Human Pancreatic Cancer , 2007, Clinical Cancer Research.

[2]  S. Riddell,et al.  Pancreatic Ductal Adenocarcinoma Contains an Effector and Regulatory Immune Cell Infiltrate that Is Altered by Multimodal Neoadjuvant Treatment , 2014, PloS one.

[3]  S. Hugues,et al.  Th17 Cell Plasticity and Functions in Cancer Immunity , 2015, BioMed research international.

[4]  M. Korsten,et al.  Periacinar stellate shaped cells in rat pancreas: identification, isolation, and culture , 1998, Gut.

[5]  M. Honda,et al.  Inflammatory features of pancreatic cancer highlighted by monocytes/macrophages and CD4+ T cells with clinical impact , 2015, Cancer science.

[6]  M. Braga,et al.  Intratumor T helper type 2 cell infiltrate correlates with cancer-associated fibroblast thymic stromal lymphopoietin production and reduced survival in pancreatic cancer , 2011, The Journal of experimental medicine.

[7]  K. Felix,et al.  Neutrophil-Derived Proteases in the Microenvironment of Pancreatic Cancer -Active Players in Tumor Progression , 2016, International journal of biological sciences.

[8]  S. Mehrotra,et al.  Th17 Cells in Cancer: The Ultimate Identity Crisis , 2014, Front. Immunol..

[9]  S. Natsugoe,et al.  Significance of M2-polarized tumor-associated macrophage in pancreatic cancer. , 2011, The Journal of surgical research.

[10]  C. Logsdon,et al.  Roles for KRAS in pancreatic tumor development and progression. , 2013, Gastroenterology.

[11]  Y. Morine,et al.  Clinical Roles of Increased Populations of Foxp3+CD4+ T Cells in Peripheral Blood from Advanced Pancreatic Cancer Patients , 2006, Pancreas.

[12]  Y. Miao,et al.  High expression of Galectin‐1 in pancreatic stellate cells plays a role in the development and maintenance of an immunosuppressive microenvironment in pancreatic cancer , 2012, International journal of cancer.

[13]  Xiaofang Wang,et al.  Changes of Th17/Treg cell and related cytokines in pancreatic cancer patients. , 2015, International journal of clinical and experimental pathology.

[14]  E. Furth,et al.  Induction of T-cell Immunity Overcomes Complete Resistance to PD-1 and CTLA-4 Blockade and Improves Survival in Pancreatic Carcinoma , 2015, Cancer Immunology Research.

[15]  Xianjun Yu,et al.  CD8+ T Cells Are Compromised In Human Pancreatic Cancer , 2012 .

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

[17]  M. Miyazaki,et al.  Effects of perioperative immunonutrition on cell-mediated immunity, T helper type 1 (Th1)/Th2 differentiation, and Th17 response after pancreaticoduodenectomy. , 2010, Surgery.

[18]  P. Lin,et al.  Expansion and functions of myeloid-derived suppressor cells in the tumor microenvironment. , 2016, Cancer letters.

[19]  Neus Martínez-Bosch,et al.  Immune Evasion in Pancreatic Cancer: From Mechanisms to Therapy , 2018, Cancers.

[20]  L. Chow,et al.  The Evolving Role of Immune Checkpoint Inhibitors in Cancer Treatment. , 2015, The oncologist.

[21]  M. Fernández-Zapico,et al.  Pancreatic cancer microenvironment, to target or not to target? , 2016, EMBO molecular medicine.

[22]  G. Beatty,et al.  Immune Escape Mechanisms as a Guide for Cancer Immunotherapy , 2014, Clinical Cancer Research.

[23]  S. Chouaib,et al.  Hypoxic stress: obstacles and opportunities for innovative immunotherapy of cancer , 2017, Oncogene.

[24]  R. DePinho,et al.  Genetics and biology of pancreatic ductal adenocarcinoma , 2006, Genes & development.

[25]  G. Cheng,et al.  Polarization of tumor-associated neutrophil phenotype by TGF-beta: "N1" versus "N2" TAN. , 2009, Cancer cell.

[26]  George Coukos,et al.  Cancer immunotherapy comes of age , 2011, Nature.

[27]  P. Conti,et al.  Mast cells: the Jekyll and Hyde of tumor growth. , 2004, Trends in immunology.

[28]  M. Stein,et al.  Targeting tumor-associated macrophages to combat pancreatic cancer , 2016, Oncotarget.

[29]  Yang Li,et al.  Hypoxia Inducible Factor 1 (HIF-1) Recruits Macrophage to Activate Pancreatic Stellate Cells in Pancreatic Ductal Adenocarcinoma , 2016, International journal of molecular sciences.

[30]  D. Gabrilovich,et al.  Antigen-Specific Inhibition of CD8+ T Cell Response by Immature Myeloid Cells in Cancer Is Mediated by Reactive Oxygen Species1 , 2004, The Journal of Immunology.

[31]  Songchuan Guo,et al.  Immunotherapy in pancreatic cancer: Unleash its potential through novel combinations , 2017, World journal of clinical oncology.

[32]  Davalyn R. Powell,et al.  Neutrophils in the Tumor Microenvironment. , 2016, Trends in immunology.

[33]  M. Korsten,et al.  Pancreatic stellate cells are activated by proinflammatory cytokines: implications for pancreatic fibrogenesis , 1999, Gut.

[34]  H. Abken,et al.  T cells that target carcinoembryonic antigen eradicate orthotopic pancreatic carcinomas without inducing autoimmune colitis in mice. , 2012, Gastroenterology.

[35]  R. Hwang,et al.  Dynamic mast cell-stromal cell interactions promote growth of pancreatic cancer. , 2013, Cancer research.

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

[37]  Pierre Michel,et al.  FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. , 2011, The New England journal of medicine.

[38]  Steven J. M. Jones,et al.  Integrated Genomic Characterization of Pancreatic Ductal Adenocarcinoma. , 2017, Cancer cell.

[39]  Alberto Mantovani,et al.  Tumour-associated macrophages as treatment targets in oncology , 2017, Nature Reviews Clinical Oncology.

[40]  A. Koong,et al.  Pancreatic tumors show high levels of hypoxia. , 2000, International journal of radiation oncology, biology, physics.

[41]  G. Miller,et al.  Myeloid-derived suppressor cells and their role in pancreatic cancer , 2016, Cancer Gene Therapy.

[42]  Se Hoon Kim,et al.  Prognostic impact of the tumor-infiltrating regulatory T-cell (Foxp3+)/activated cytotoxic T lymphocyte (granzyme B+) ratio on resected left-sided pancreatic cancer , 2016, Oncology letters.

[43]  R. Schreiber,et al.  The three Es of cancer immunoediting. , 2004, Annual review of immunology.

[44]  W. Zou,et al.  TH17 cells in tumour immunity and immunotherapy , 2010, Nature Reviews Immunology.

[45]  Xuetao Cao,et al.  The origin and function of tumor-associated macrophages , 2014, Cellular and Molecular Immunology.

[46]  Yong-jian Jiang,et al.  Role of immune cells in pancreatic cancer from bench to clinical application , 2016, Medicine.

[47]  E. Meuillet,et al.  Improved Treatment of Pancreatic Cancer With Drug Delivery Nanoparticles Loaded With a Novel AKT/PDK1 Inhibitor , 2016, Pancreas.

[48]  D. Olive,et al.  BTN3A is a prognosis marker and a promising target for Vγ9Vδ2 T cells based-immunotherapy in pancreatic ductal adenocarcinoma (PDAC) , 2018, Oncoimmunology.

[49]  L. De Monte,et al.  Immune infiltrates as predictive markers of survival in pancreatic cancer patients , 2013, Front. Physiol..

[50]  T. Whiteside The role of regulatory T cells in cancer immunology , 2015, ImmunoTargets and therapy.

[51]  D. V. Von Hoff,et al.  Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. , 1997, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[52]  D. Bar-Sagi,et al.  γδ T Cells Support Pancreatic Oncogenesis by Restraining αβ T Cell Activation , 2016, Cell.

[53]  G. Zhu,et al.  Tumor-associated B7-H1 promotes T-cell apoptosis: A potential mechanism of immune evasion , 2002, Nature Medicine.

[54]  Jonathan B. Mitchem,et al.  Induction of Th17 Cells in the Tumor Microenvironment Improves Survival in a Murine Model of Pancreatic Cancer , 2010, The Journal of Immunology.

[55]  A. Mantovani,et al.  Smoldering and polarized inflammation in the initiation and promotion of malignant disease. , 2005, Cancer cell.

[56]  Y. Kanai,et al.  Immune cell infiltration as an indicator of the immune microenvironment of pancreatic cancer , 2013, British Journal of Cancer.

[57]  Y. Miao,et al.  Altered expression of CD226 and CD96 on natural killer cells in patients with pancreatic cancer , 2016, Oncotarget.

[58]  Chun-tao Wu,et al.  Infiltrating immune cells and gene mutations in pancreatic ductal adenocarcinoma , 2016, The British journal of surgery.

[59]  M. Fei,et al.  Distribution and Clinical Significance of Th17 Cells in the Tumor Microenvironment and Peripheral Blood of Pancreatic Cancer Patients , 2011, International journal of molecular sciences.

[60]  M. Ebsen,et al.  Tumor‐associated macrophages exhibit pro‐ and anti‐inflammatory properties by which they impact on pancreatic tumorigenesis , 2014, International journal of cancer.

[61]  P. Allavena,et al.  Role of tumor-associated macrophages in tumor progression and invasion , 2006, Cancer and Metastasis Reviews.

[62]  M. Lesina,et al.  The immune network in pancreatic cancer development and progression , 2014, Oncogene.

[63]  J. Gribben,et al.  Activated pancreatic stellate cells sequester CD8+ T cells to reduce their infiltration of the juxtatumoral compartment of pancreatic ductal adenocarcinoma. , 2013, Gastroenterology.

[64]  P. Allavena,et al.  Cancer-related inflammation , 2008, Nature.

[65]  H. Nishikawa,et al.  Roles of regulatory T cells in cancer immunity. , 2016, International immunology.

[66]  J. Humm,et al.  A Combination of Radiation and the Hypoxia-Activated Prodrug Evofosfamide (TH-302) is Efficacious against a Human Orthotopic Pancreatic Tumor Model12 , 2017, Translational oncology.

[67]  I. Mellman,et al.  Elements of cancer immunity and the cancer–immune set point , 2017, Nature.

[68]  B. Baradaran,et al.  The paradox of Th17 cell functions in tumor immunity. , 2017, Cellular immunology.

[69]  S. Song,et al.  Differences in Immune Cells Engaged in Cell-Mediated Immunity After Chemotherapy for Far Advanced Pancreatic Cancer , 2006, Pancreas.

[70]  E. Thorsby,et al.  Vaccination with mutant ras peptides and induction of T-cell responsiveness in pancreatic carcinoma patients carrying the corresponding RAS mutation , 1995, The Lancet.

[71]  Janice M. Y. Brown,et al.  The hypoxic cell: a target for selective cancer therapy--eighteenth Bruce F. Cain Memorial Award lecture. , 1999, Cancer research.

[72]  G. Sammarco,et al.  Mast Cells Density Positive to Tryptase Correlates with Angiogenesis in Pancreatic Ductal Adenocarcinoma Patients Having Undergone Surgery , 2014, Gastroenterology research and practice.