CXCR3 and Cognate Ligands are Associated with Immune Cell Alteration and Aggressiveness of Pancreatic Ductal Adenocarcinoma

Purpose: The cytokine milieu in pancreatic ductal adenocarcinoma (PDAC) promotes tumor progression and immune suppression, contributing to the dismal prognosis of patients with PDAC. The roles of many of these cytokines, however, have not been thoroughly investigated in PDAC. Experimental Design: PDAC microarray and The Cancer Genome Atlas datasets were analyzed to identify cytokines and cognate receptors overexpressed in PDAC and associated with survival. Pathway and CIBERSORT analyses were used to elucidate potential mechanisms of altered patient survival. Comparative analysis of cytokine expression in KPC (K-rasG12D; TP53R172H; Pdx-1cre) and KC (K-rasG12D; Pdx-1cre) PDAC models and multicolor immunofluorescence (IF) staining of human PDAC–resected samples were used to validate these findings. Results: CXCL9 and CXCL10 were among the most highly overexpressed cytokines by bioinformatics analyses, while their receptor, CXCR3, was significantly overexpressed by IHC analysis. Higher CXCR3 ligand expression was associated with shorter overall survival, while high CXCR3 expression was associated with better survival. The CXCR3 ligands, CXCL4, 9, and 10, were overexpressed in KPC compared with KC mice. Pathway analysis of CXCR3- and CXCR3 ligand–associated genes showed that CXCR3 is a marker of antitumor immunity, while its ligands may promote immunosuppression. CIBERSORT and IF studies of PDAC tissues demonstrated that high CXCR3 expression was associated with increased CD8+ T-cell and naïve B-cell signatures and loss of plasma cell signatures. CXCR3 ligand expression was associated with increased CD8+ T-cell signatures and loss of natural killer–cell signatures. Conclusions: CXCR3 ligands are overexpressed in PDAC and are associated with poor survival likely related to alterations in tumor immune infiltrate/activity.

[1]  M. Ebert,et al.  CXCL10 and CCL21 Promote Migration of Pancreatic Cancer Cells Toward Sensory Neurons and Neural Remodeling in Tumors in Mice, Associated With Pain in Patients. , 2020, Gastroenterology.

[2]  J. Wargo,et al.  B cells are associated with survival and immunotherapy response in sarcoma , 2020, Nature.

[3]  Jeffrey E. Lee,et al.  B cells and tertiary lymphoid structures promote immunotherapy response , 2020, Nature.

[4]  D. Schadendorf,et al.  Tertiary lymphoid structures improve immunotherapy and survival in melanoma , 2020, Nature.

[5]  Wei Li,et al.  Inflammation Promotes Progression of Pancreatic Cancer Through WNT/β-Catenin Pathway-Dependent Manner. , 2019, Pancreas.

[6]  E. Collisson,et al.  Blockade of leukemia inhibitory factor as a therapeutic approach to KRAS driven pancreatic cancer , 2019, Nature Communications.

[7]  D. Jackson,et al.  Cooperation between Constitutive and Inducible Chemokines Enables T Cell Engraftment and Immune Attack in Solid Tumors. , 2019, Cancer cell.

[8]  D. Kuang,et al.  Plasma Cell Polarization to the Immunoglobulin G Phenotype in Hepatocellular Carcinomas Involves Epigenetic Alterations and Promotes Hepatoma Progression in Mice. , 2019, Gastroenterology.

[9]  Hesham H. Ali,et al.  Uncovering and characterizing splice variants associated with survival in lung cancer patients , 2019, bioRxiv.

[10]  Galina A. Erikson,et al.  Targeting LIF-mediated paracrine interaction for pancreatic cancer therapy and monitoring , 2019, Nature.

[11]  Mingyuan Wu,et al.  Non-platelet-derived CXCL4 differentially regulates cytotoxic and regulatory T cells through CXCR3 to suppress the immune response to colon cancer. , 2019, Cancer letters.

[12]  A. Califano,et al.  Experimental microdissection enables functional harmonisation of pancreatic cancer subtypes , 2019, Gut.

[13]  R. Searcy,et al.  IL35 Hinders Endogenous Antitumor T-cell Immunity and Responsiveness to Immunotherapy in Pancreatic Cancer , 2018, Cancer Immunology Research.

[14]  Ce Li,et al.  CXCL9/10/11, a regulator of PD-L1 expression in gastric cancer , 2018, BMC Cancer.

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

[16]  Rob Patro,et al.  Salmon provides fast and bias-aware quantification of transcript expression , 2017, Nature Methods.

[17]  Hans Clevers,et al.  Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer , 2017, The Journal of experimental medicine.

[18]  Kebin Liu,et al.  JAK-STAT-mediated chronic inflammation impairs cytotoxic T lymphocyte activation to decrease anti-PD-1 immunotherapy efficacy in pancreatic cancer , 2017, Oncoimmunology.

[19]  L. Moldawer,et al.  Human Pancreatic Cancer Cells Induce a MyD88-Dependent Stromal Response to Promote a Tumor-Tolerant Immune Microenvironment. , 2017, Cancer research.

[20]  C. Billottet,et al.  Dual Roles for CXCL4 Chemokines and CXCR3 in Angiogenesis and Invasion of Pancreatic Cancer. , 2016, Cancer research.

[21]  Achim Krüger,et al.  Systemic Ablation of MMP-9 Triggers Invasive Growth and Metastasis of Pancreatic Cancer via Deregulation of IL6 Expression in the Bone Marrow , 2016, Molecular Cancer Research.

[22]  R. Jain,et al.  Obesity-Induced Inflammation and Desmoplasia Promote Pancreatic Cancer Progression and Resistance to Chemotherapy. , 2016, Cancer discovery.

[23]  A. Schetter,et al.  A Novel MIF Signaling Pathway Drives the Malignant Character of Pancreatic Cancer by Targeting NR3C2. , 2016, Cancer research.

[24]  A. Biankin,et al.  CXCR2 Inhibition Profoundly Suppresses Metastases and Augments Immunotherapy in Pancreatic Ductal Adenocarcinoma , 2016, Cancer cell.

[25]  M. Reni,et al.  Basophil Recruitment into Tumor-Draining Lymph Nodes Correlates with Th2 Inflammation and Reduced Survival in Pancreatic Cancer Patients. , 2016, Cancer research.

[26]  W. Bamlet,et al.  GSK-3β Governs Inflammation-Induced NFATc2 Signaling Hubs to Promote Pancreatic Cancer Progression , 2016, Molecular Cancer Therapeutics.

[27]  Ash A. Alizadeh,et al.  Robust enumeration of cell subsets from tissue expression profiles , 2015, Nature Methods.

[28]  S. Batra,et al.  NCOA3-mediated upregulation of mucin expression via transcriptional and post-translational changes during the development of pancreatic cancer , 2014, Oncogene.

[29]  N. Jamieson,et al.  IP-10/CXCL10 induction in human pancreatic cancer stroma influences lymphocytes recruitment and correlates with poor survival , 2014, Oncotarget.

[30]  Lianyu Chen,et al.  The IL-8/CXCR1 axis is associated with cancer stem cell-like properties and correlates with clinical prognosis in human pancreatic cancer cases , 2014, Scientific Reports.

[31]  C. Hajdu,et al.  Senescence-associated SIN3B promotes inflammation and pancreatic cancer progression. , 2014, The Journal of clinical investigation.

[32]  W. Bamlet,et al.  Inflammation-induced NFATc1-STAT3 transcription complex promotes pancreatic cancer initiation by KrasG12D. , 2014, Cancer discovery.

[33]  Andreas Krämer,et al.  Causal analysis approaches in Ingenuity Pathway Analysis , 2013, Bioinform..

[34]  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.

[35]  T. Roskams,et al.  Pancreatic cancer circulating tumour cells express a cell motility gene signature that predicts survival after surgery , 2012, BMC Cancer.

[36]  N. Jhala,et al.  Tumor-derived granulocyte-macrophage colony-stimulating factor regulates myeloid inflammation and T cell immunity in pancreatic cancer. , 2012, Cancer cell.

[37]  Jochen Gaedcke,et al.  DPEP1 Inhibits Tumor Cell Invasiveness, Enhances Chemosensitivity and Predicts Clinical Outcome in Pancreatic Ductal Adenocarcinoma , 2012, PloS one.

[38]  Hong Wu,et al.  Integrative Survival-Based Molecular Profiling of Human Pancreatic Cancer , 2012, Clinical Cancer Research.

[39]  Wei Hu,et al.  Priming microenvironments dictate cytokine requirements for T helper 17 cell lineage commitment. , 2011, Immunity.

[40]  Gerald C. Chu,et al.  PTEN is a major tumor suppressor in pancreatic ductal adenocarcinoma and regulates an NF-κB-cytokine network. , 2011, Cancer discovery.

[41]  M. Barbacid,et al.  Pancreatitis-induced inflammation contributes to pancreatic cancer by inhibiting oncogene-induced senescence. , 2011, Cancer cell.

[42]  Krishna R. Kalari,et al.  FKBP51 affects cancer cell response to chemotherapy by negatively regulating Akt. , 2009, Cancer cell.

[43]  Liviu Badea,et al.  Combined gene expression analysis of whole-tissue and microdissected pancreatic ductal adenocarcinoma identifies genes specifically overexpressed in tumor epithelia. , 2008, Hepato-gastroenterology.

[44]  C. Heeschen,et al.  Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. , 2007, Cell stem cell.

[45]  T. Manabe,et al.  Chemokine receptor CXCR3 promotes colon cancer metastasis to lymph nodes , 2007, Oncogene.

[46]  Pablo Tamayo,et al.  Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[47]  Sergio Romagnani,et al.  An Alternatively Spliced Variant of CXCR3 Mediates the Inhibition of Endothelial Cell Growth Induced by IP-10, Mig, and I-TAC, and Acts as Functional Receptor for Platelet Factor 4 , 2003, The Journal of experimental medicine.

[48]  James G. Boyd,et al.  Interferon–inducible T Cell Alpha Chemoattractant (I-TAC): A Novel Non-ELR CXC Chemokine with Potent Activity on Activated T Cells through Selective High Affinity Binding to CXCR3 , 1998, The Journal of experimental medicine.

[49]  Simon A. Jones,et al.  Chemokine receptor specific for IP10 and mig: structure, function, and expression in activated T-lymphocytes , 1996, The Journal of experimental medicine.