Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Biomolecules ; 11(6)2021 06 17.
Article in English | MEDLINE | ID: mdl-34204306

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignancy with a five-year survival rate of only 9%. PDAC is characterized by a dense, fibrotic stroma composed of extracellular matrix (ECM) proteins. This desmoplastic stroma is a hallmark of PDAC, representing a significant physical barrier that is immunosuppressive and obstructs penetration of cytotoxic chemotherapy agents into the tumor microenvironment (TME). Additionally, dense ECM promotes hypoxia, making tumor cells refractive to radiation therapy and alters their metabolism, thereby supporting proliferation and survival. In this review, we outline the significant contribution of fibrosis to the pathogenesis of pancreatic cancer, with a focus on the cross talk between immune cells and pancreatic stellate cells that contribute to ECM deposition. We emphasize the cellular mechanisms by which neutrophils and macrophages, specifically, modulate the ECM in favor of PDAC-progression. Furthermore, we investigate how activated stellate cells and ECM influence immune cells and promote immunosuppression in PDAC. Finally, we summarize therapeutic strategies that target the stroma and hinder immune cell promotion of fibrogenesis, which have unfortunately led to mixed results. An enhanced understanding of the complex interactions between the pancreatic tumor ECM and immune cells may uncover novel treatment strategies that are desperately needed for this devastating disease.


Subject(s)
Carcinoma, Pancreatic Ductal/immunology , Extracellular Matrix/immunology , Immune Tolerance , Macrophages/immunology , Neutrophils/immunology , Pancreatic Neoplasms/immunology , Tumor Microenvironment , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/therapy , Humans , Macrophages/pathology , Neutrophils/pathology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , Pancreatic Stellate Cells/immunology , Pancreatic Stellate Cells/pathology
2.
Mol Cell ; 81(11): 2290-2302.e7, 2021 06 03.
Article in English | MEDLINE | ID: mdl-33831358

ABSTRACT

Cancer cells adapt their metabolism to support elevated energetic and anabolic demands of proliferation. Folate-dependent one-carbon metabolism is a critical metabolic process underpinning cellular proliferation supplying carbons for the synthesis of nucleotides incorporated into DNA and RNA. Recent research has focused on the nutrients that supply one-carbons to the folate cycle, particularly serine. Tryptophan is a theoretical source of one-carbon units through metabolism by IDO1, an enzyme intensively investigated in the context of tumor immune evasion. Using in vitro and in vivo pancreatic cancer models, we show that IDO1 expression is highly context dependent, influenced by attachment-independent growth and the canonical activator IFNγ. In IDO1-expressing cancer cells, tryptophan is a bona fide one-carbon donor for purine nucleotide synthesis in vitro and in vivo. Furthermore, we show that cancer cells release tryptophan-derived formate, which can be used by pancreatic stellate cells to support purine nucleotide synthesis.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Pancreatic Neoplasms/genetics , Pancreatic Stellate Cells/metabolism , Tumor Escape/drug effects , Allografts , Animals , Antineoplastic Agents/pharmacology , Carbon/immunology , Carbon/metabolism , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/mortality , Cell Line, Tumor , Formates/immunology , Formates/metabolism , Gene Expression Regulation, Neoplastic , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/genetics , Mice , Mice, Inbred C57BL , Mice, Nude , Oximes/pharmacology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/mortality , Pancreatic Stellate Cells/drug effects , Pancreatic Stellate Cells/immunology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/immunology , Serine/immunology , Serine/metabolism , Serine/pharmacology , Signal Transduction , Sulfonamides/pharmacology , Tryptophan/immunology , Tryptophan/metabolism , Tryptophan/pharmacology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology
3.
Int J Mol Sci ; 21(22)2020 Nov 20.
Article in English | MEDLINE | ID: mdl-33233631

ABSTRACT

The purinergic signaling has an important role in regulating pancreatic exocrine secretion. The exocrine pancreas is also a site of one of the most serious cancer forms, the pancreatic ductal adenocarcinoma (PDAC). Here, we explore how the network of purinergic and adenosine receptors, as well as ecto-nucleotidases regulate normal pancreatic cells and various cells within the pancreatic tumor microenvironment. In particular, we focus on the P2X7 receptor, P2Y2 and P2Y12 receptors, as well as A2 receptors and ecto-nucleotidases CD39 and CD73. Recent studies indicate that targeting one or more of these candidates could present new therapeutic approaches to treat pancreatic cancer. In pancreatic cancer, as much as possible of normal pancreatic function should be preserved, and therefore physiology of purinergic signaling in pancreas needs to be considered.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Gene Expression Regulation, Neoplastic/drug effects , Pancreatic Neoplasms/drug therapy , Signal Transduction/genetics , 5'-Nucleotidase/genetics , 5'-Nucleotidase/immunology , Animals , Apyrase/genetics , Apyrase/immunology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Clinical Trials as Topic , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Gene Expression Regulation, Neoplastic/immunology , Humans , Immunotherapy/methods , Pancreas/drug effects , Pancreas/immunology , Pancreas/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/drug effects , Pancreatic Stellate Cells/immunology , Pancreatic Stellate Cells/pathology , Receptors, Adenosine A2/genetics , Receptors, Adenosine A2/immunology , Receptors, Purinergic P2X7/genetics , Receptors, Purinergic P2X7/immunology , Receptors, Purinergic P2Y12/genetics , Receptors, Purinergic P2Y12/immunology , Receptors, Purinergic P2Y2/genetics , Receptors, Purinergic P2Y2/immunology , Signal Transduction/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
4.
J Cancer Res Clin Oncol ; 146(12): 3233-3240, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32865617

ABSTRACT

INTRODUCTION: The immunosuppressive tumor microenvironment promotes progression of pancreatic ductal adenocarcinoma (PDAC). γδ T cells infiltrate the pancreatic tumor stroma and support tumorigenesis through αß T cell inhibition. Pancreatic stellate cell (PSC) activation contributes to pancreatic fibrosis in PDAC, limiting the delivery and efficacy of therapeutic agents. Whether γδ T cells have direct effects on PSC activation is unknown. METHODS: In this study, we analyzed tumor tissue from 68 patients with PDAC and determined the frequency and location of γδ T cells using immunohistochemistry and immunofluorescence. PDAC samples from the TCGA database with low and high TRGC2 expression were correlated with the expression of extracellular matrix genes. Further, PSCs were isolated from pancreatic tumor tissue and co-cultured with γδ T cells for 48 hours and cytokine production was measured using a cytometric bead array. RESULTS: γδ T cells infiltrated the pancreatic tumor stroma and were located in proximity to PSCs. A high infiltration of γδ T cells was associated with increased expression of several extracellular matrix genes in human PDAC. In vitro, γδ T cells stimulated IL-6 production by PDAC-derived PSCs. CONCLUSION: γδ T cells activated PSCs and modulation of this interaction may enhance the efficacy of combinational therapies in human PDAC.


Subject(s)
Adenocarcinoma/immunology , Carcinoma, Pancreatic Ductal/immunology , Interleukin-6/genetics , Intraepithelial Lymphocytes/immunology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Carcinogenesis/genetics , Carcinogenesis/immunology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation , Coculture Techniques , Extracellular Matrix/genetics , Gene Expression Regulation, Neoplastic , Humans , Pancreatic Stellate Cells/immunology , Pancreatic Stellate Cells/metabolism , Tumor Microenvironment/genetics
5.
BMC Cancer ; 20(1): 651, 2020 Jul 13.
Article in English | MEDLINE | ID: mdl-32660466

ABSTRACT

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive forms of malignancies with a nearly equal incidence and mortality rates in patients. Pancreatic stellate cells (PSCs) are critical players in PDAC microenvironment to promote the aggressiveness and pathogenesis of the disease. Dysregulation of microRNAs (miRNAs) have been shown to play a significant role in progression of PDAC. Earlier, we observed a PSC-specific downregulation of miR-29a in PDAC pancreas, however, the mechanism of action of the molecule in PSCs is still to be elucidated. The current study aims to clarify the regulation of miR-29a in PSCs and identifies functionally important downstream targets that contribute to tumorigenic activities during PDAC progression. METHODS: In this study, using RNAseq approach, we performed transcriptome analysis of paired miR-29a overexpressing and control human PSCs (hPSCs). Enrichment analysis was performed with the identified differentially expressed genes (DEGs). miR-29a targets in the dataset were identified, which were utilized to create network interactions. Western blots were performed with the top miR-29a candidate targets in hPSCs transfected with miR-29a mimic or scramble control. RESULTS: RNAseq analysis identified 202 differentially expressed genes, which included 19 downregulated direct miR-29a targets. Translational repression of eight key pro-tumorigenic and -fibrotic targets namely IGF-1, COL5A3, CLDN1, E2F7, MYBL2, ITGA6 and ADAMTS2 by miR-29a was observed in PSCs. Using pathway analysis, we find that miR-29a modulates effectors of IGF-1-p53 signaling in PSCs that may hinder carcinogenesis. We further observe a regulatory role of the molecule in pathways associated with PDAC ECM remodeling and tumor-stromal crosstalk, such as INS/IGF-1, RAS/MAPK, laminin interactions and collagen biosynthesis. CONCLUSIONS: Together, our study presents a comprehensive understanding of miR-29a regulation of PSCs, and identifies essential pathways associated with PSC-mediated PDAC pathogenesis. The findings suggest an anti-tumorigenic role of miR-29a in the context of PSC-cancer cell crosstalk and advocates for the potential of the molecule in PDAC targeted therapies.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Pancreatic Ductal/pathology , MicroRNAs/genetics , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/pathology , Transcriptome , Tumor Microenvironment/genetics , Apoptosis , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , Pancreatic Stellate Cells/immunology , Pancreatic Stellate Cells/metabolism , Tumor Cells, Cultured , Tumor Microenvironment/immunology , Pancreatic Neoplasms
6.
Pancreas ; 49(1): 120-127, 2020 01.
Article in English | MEDLINE | ID: mdl-31856087

ABSTRACT

OBJECTIVES: Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal forms of cancer with poor prognosis. Pancreatic stellate cells (PSCs) play a vital role in PDAC development. The aim of this study was to explore tumor microenvironment response to PSCs in an orthotopic pancreatic cancer mouse model and to assess if PSCs secreted factors that can facilitate an immunosuppressive microenvironment. METHODS: Pancreatic ductal adenocarcinoma orthotopic tumor model, derived from coinjection of Panc02 cells plus PSCs, was used to investigate tumor proliferation, metastasis, and the population of immune cells in vivo, including regulatory T cells, M2-type macrophages, myeloid-derived suppressor cells, CD8 T cells, CD4 T cells, M1-type macrophages, natural killer (NK), and NK T cells. RESULTS: Pancreatic stellate cells promoted PDAC growth not only induced cell proliferation and metastasis, but also significantly increased the suppressive immune cell population of regulatory T cells, M2-type macrophages, and myeloid-derived suppressor cells. In addition, PSCs decreased the immune cell population of CD8 T, CD4 T cells, and M1-type macrophages in the spleen and tumor tissues of the tumor-bearing mice. Moreover, PSCs decreased the population of NK and NK T cells in the tumor tissues. CONCLUSIONS: Our findings support PSCs playing multiple roles in PDAC development via promoting immunosuppressive microenvironment.


Subject(s)
Carcinoma, Pancreatic Ductal/immunology , Disease Models, Animal , Pancreatic Neoplasms/immunology , Pancreatic Stellate Cells/immunology , Tumor Microenvironment/immunology , Animals , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Disease Progression , Female , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/metabolism , Pancreatic Stellate Cells/pathology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
7.
Gastroenterology ; 155(3): 880-891.e8, 2018 09.
Article in English | MEDLINE | ID: mdl-29909021

ABSTRACT

BACKGROUND & AIMS: Immunotherapies are ineffective against pancreatic cancer. We investigated whether the activity of nuclear factor (NF)κB in pancreatic stromal cells contributes to an environment that suppresses antitumor immune response. METHODS: Pancreata of C57BL/6 or Rag1-/- mice were given pancreatic injections of a combination of KrasG12D/+; Trp53 R172H/+; Pdx-1cre (KPC) pancreatic cancer cells and pancreatic stellate cells (PSCs) extracted from C57BL/6 (control) or mice with disruption of the gene encoding the NFκB p50 subunit (Nfkb1 or p50-/- mice). Tumor growth was measured as an endpoint. Other mice were given injections of Lewis lung carcinoma (LLC) lung cancer cells or B16-F10 melanoma cells with control or p50-/- fibroblasts. Cytotoxic T cells were depleted from C57BL/6 mice by administration of antibodies against CD8 (anti-CD8), and growth of tumors from KPC cells, with or without control or p50-/- PSCs, was measured. Some mice were given an inhibitor of CXCL12 (AMD3100) and tumor growth was measured. T-cell migration toward cancer cells was measured using the Boyden chamber assay. RESULTS: C57BL/6 mice coinjected with KPC cells (or LLC or B16-F10 cells) and p50-/- PSCs developed smaller tumors than mice given injections of the cancer cells along with control PSCs. Tumors that formed when KPC cells were injected along with p50-/- PSCs had increased infiltration by activated cytotoxic T cells along with decreased levels of CXCL12, compared with tumors grown from KPC cells injected along with control PSCs. KPC cells, when coinjected with control or p50-/- PSCs, developed the same-size tumors when CD8+ T cells were depleted from C57BL/6 mice or in Rag1-/- mice. The CXCL12 inhibitor slowed tumor growth and increased tumor infiltration by cytotoxic T cells. In vitro expression of p50 by PSCs reduced T-cell migration toward and killing of cancer cells. When cultured with cancer cells, control PSCs expressed 10-fold higher levels of CXCL12 than p50-/- PSCs. The CXCL12 inhibitor increased migration of T cells toward KPC cells in culture. CONCLUSIONS: In studies of mice and cell lines, we found that NFκB activity in PSCs promotes tumor growth by increasing expression of CXCL12, which prevents cytotoxic T cells from infiltrating the tumor and killing cancer cells. Strategies to block CXCL12 in pancreatic tumor cells might increase antitumor immunity.


Subject(s)
Chemokine CXCL12/physiology , Lymphocytes, Tumor-Infiltrating/physiology , NF-kappa B/physiology , Pancreatic Neoplasms/metabolism , Pancreatic Stellate Cells/metabolism , T-Lymphocytes, Cytotoxic/physiology , Animals , Carcinogenesis/metabolism , Cell Line, Tumor , Immunity, Cellular , Mice , Mice, Inbred C57BL , Pancreatic Neoplasms/immunology , Pancreatic Stellate Cells/immunology , Up-Regulation
8.
Gastroenterology ; 154(5): 1524-1537.e6, 2018 04.
Article in English | MEDLINE | ID: mdl-29274868

ABSTRACT

BACKGROUND & AIMS: Pancreatic ductal adenocarcinoma (PDAC) is characterized by activated pancreatic stellate cells (PSCs), abundance of extracellular matrix (ECM), and production of cytokines and chemokines. Galectin 3 (GAL3), a ß-galactoside-specific lectin, contributes to PDAC development but its effects on the stroma and cytokine production are unclear. METHODS: The effect of recombinant human GAL3 (rGAL3) on activation of PSCs, production of cytokines, and ECM proteins was determined by proliferation, invasion, cytokine array, and quantitative polymerase chain reaction. We assessed co-cultures of PDAC cells with GAL3 genetic alterations with PSCs. Production of interleukin 8 (IL8) and activities of nuclear factor (NF)-κB were determined by enzyme-linked immunosorbent assay and luciferase reporter analyses. We studied the effects of inhibitors of NF-κB and integrin-linked kinase (ILK) on pathways activated by rGAL3. RESULTS: In analyses of the Gene Expression Omnibus database and our dataset, we observed higher levels of GAL3, IL8, and other cytokines in PDAC than in nontumor tissues. Production of IL8, granulocyte-macrophage colony-stimulating factor, chemokine ligand 1, and C-C motif chemokine ligand 2 increased in PSCs exposed to rGAL3 compared with controls. Culture of PSCs with PDAC cells that express different levels of GAL3 resulted in proliferation and invasion of PSCs that increased with level of GAL3. GAL3 stimulated transcription of IL8 through integrin subunit beta 1 (ITGB1) on PSCs, which activates NF-κB through ILK. Inhibitors of ILK or NF-κB or a neutralizing antibody against ITGB1 blocked transcription and production of IL8 from PSCs induced by rGAL3. The GAL3 inhibitor significantly reduced growth and metastases of orthotopic tumors that formed from PDAC and PSC cells co-implanted in mice. CONCLUSION: GAL3 activates PSC cells to produce inflammatory cytokines via ITGB1signaling to ILK and activation of NF-κB. Inhibition of this pathway reduced growth and metastases of pancreatic orthotopic tumors in mice.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Cytokines/metabolism , Galectin 3/metabolism , Integrin beta1/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Stellate Cells/metabolism , Paracrine Communication , Stromal Cells/metabolism , Tumor Microenvironment , Animals , Antineoplastic Agents/pharmacology , Blood Proteins , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/secondary , Cell Line, Tumor , Cell Movement , Cell Proliferation , Coculture Techniques , Cytokines/genetics , Extracellular Matrix Proteins/metabolism , Galectin 3/antagonists & inhibitors , Galectins , Gene Expression Regulation, Neoplastic , Humans , Mice, Nude , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Neoplasm Invasiveness , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/drug effects , Pancreatic Stellate Cells/immunology , Pancreatic Stellate Cells/pathology , Paracrine Communication/drug effects , Phenotype , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Stromal Cells/drug effects , Stromal Cells/immunology , Stromal Cells/pathology , Time Factors , Transcription, Genetic , Xenograft Model Antitumor Assays
9.
Lab Invest ; 98(1): 27-40, 2018 01.
Article in English | MEDLINE | ID: mdl-29155423

ABSTRACT

Desmoplasia is a fibro-inflammatory process and a well-established feature of pancreatic cancer. A key contributor to pancreatic cancer desmoplasia is the pancreatic stellate cell. Various in vitro and in vivo methods have emerged for the isolation, characterization, and use of pancreatic stellate cells in models of cancer-associated fibrosis. In addition to cell culture models, genetically engineered animal models have been established that spontaneously develop pancreatic cancer with desmoplasia. These animal models are currently being used for the study of pancreatic cancer pathogenesis and for evaluating therapeutics against pancreatic cancer. Here, we review various in vitro and in vivo models that are being used or have the potential to be used to study desmoplasia in pancreatic cancer.


Subject(s)
Biomedical Research/methods , Disease Models, Animal , Fibroma/etiology , Pancreatic Neoplasms/physiopathology , Animals , Animals, Genetically Modified , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomedical Research/trends , Cell Line, Tumor , Drugs, Investigational/pharmacology , Drugs, Investigational/therapeutic use , Female , Fibroma/drug therapy , Fibroma/immunology , Fibroma/pathology , Fibrosis , Humans , Male , Mice , Neoplasm Transplantation/methods , Neoplasm Transplantation/trends , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/drug effects , Pancreatic Stellate Cells/immunology , Pancreatic Stellate Cells/pathology , Pancreatic Stellate Cells/transplantation , Rats , Tumor Cells, Cultured , Xenograft Model Antitumor Assays/methods
10.
Gut ; 67(2): 320-332, 2018 02.
Article in English | MEDLINE | ID: mdl-27797936

ABSTRACT

OBJECTIVE: Limited efficacy of immune checkpoint inhibitors in pancreatic ductal adenocarcinoma (PDAC) has prompted investigation into combination therapy. We hypothesised that interleukin 6 (IL-6) blockade would modulate immunological features of PDAC and enhance the efficacy of anti-programmed death-1-ligand 1 (PD-L1) checkpoint inhibitor therapy. DESIGN: Transcription profiles and IL-6 secretion from primary patient-derived pancreatic stellate cells (PSCs) were analyzed via Nanostring and immunohistochemistry, respectively. In vivo efficacy and mechanistic studies were conducted with antibodies (Abs) targeting IL-6, PD-L1, CD4 or CD8 in subcutaneous or orthotopic models using Panc02, MT5 or KPC-luc cell lines; and the aggressive, genetically engineered PDAC model (KrasLSL-G12D, Trp53LSL-R270H, Pdx1-cre, Brca2F/F (KPC-Brca2 mice)). Systemic and local changes in immunophenotype were measured by flow cytometry or immunohistochemical analysis. RESULTS: PSCs (n=12) demonstrated prominent IL-6 expression, which was localised to stroma of tumours. Combined IL-6 and PD-L1 blockade elicited efficacy in mice bearing subcutaneous MT5 (p<0.02) and Panc02 tumours (p=0.046), which was accompanied by increased intratumoural effector T lymphocytes (CD62L-CD44-). CD8-depleting but not CD4-depleting Abs abrogated the efficacy of combined IL-6 and PD-L1 blockade in mice bearing Panc02 tumours (p=0.0016). This treatment combination also elicited significant antitumour activity in mice bearing orthotopic KPC-luc tumours and limited tumour progression in KPC-Brca2 mice (p<0.001). Histological analysis revealed increased T-cell infiltration and reduced α-smooth muscle actin cells in tumours from multiple models. Finally, IL-6 and PD-L1 blockade increased overall survival in KPC-Brca2 mice compared with isotype controls (p=0.0012). CONCLUSIONS: These preclinical results indicate that targeted inhibition of IL-6 may enhance the efficacy of anti-PD-L1 in PDAC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Carcinoma, Pancreatic Ductal/drug therapy , Interleukin-6/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Actins/metabolism , Animals , Antineoplastic Agents, Immunological/administration & dosage , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/metabolism , Disease Models, Animal , Disease Progression , Female , Humans , Hyaluronan Receptors/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Janus Kinases/metabolism , L-Selectin/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Mice, Inbred C57BL , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/metabolism , Pancreatic Stellate Cells/immunology , Pancreatic Stellate Cells/metabolism , STAT Transcription Factors/metabolism , Survival Rate , Th1 Cells/metabolism , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
11.
Clin Cancer Res ; 23(7): 1656-1669, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28373364

ABSTRACT

Immunotherapy has changed the standard of care for multiple deadly cancers, including lung, head and neck, gastric, and some colorectal cancers. However, single-agent immunotherapy has had little effect in pancreatic ductal adenocarcinoma (PDAC). Increasing evidence suggests that the PDAC microenvironment is comprised of an intricate network of signals between immune cells, PDAC cells, and stroma, resulting in an immunosuppressive environment resistant to single-agent immunotherapies. In this review, we discuss differences between immunotherapy-sensitive cancers and PDAC, the complex interactions between PDAC stroma and suppressive tumor-infiltrating cells that facilitate PDAC development and progression, the immunologic targets within these complex networks that are druggable, and data supporting combination drug approaches that modulate multiple PDAC signals, which should lead to improved clinical outcomes. Clin Cancer Res; 23(7); 1656-69. ©2017 AACRSee all articles in this CCR Focus section, "Pancreatic Cancer: Challenge and Inspiration."


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Drug Resistance, Neoplasm/immunology , Immunotherapy , Tumor Microenvironment/immunology , Adenocarcinoma/immunology , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Humans , Pancreatic Stellate Cells/immunology , Pancreatic Stellate Cells/pathology
12.
World J Gastroenterol ; 23(3): 382-405, 2017 Jan 21.
Article in English | MEDLINE | ID: mdl-28210075

ABSTRACT

Pancreatic stellate cells (PSCs) were identified in the early 1980s, but received much attention after 1998 when the methods to isolate and culture them from murine and human sources were developed. PSCs contribute to a small proportion of all pancreatic cells under physiological condition, but are essential for maintaining the normal pancreatic architecture. Quiescent PSCs are characterized by the presence of vitamin A laden lipid droplets. Upon PSC activation, these perinuclear lipid droplets disappear from the cytosol, attain a myofibroblast like phenotype and expresses the activation marker, alpha smooth muscle actin. PSCs maintain their activated phenotype via an autocrine loop involving different cytokines and contribute to progressive fibrosis in chronic pancreatitis (CP) and pancreatic ductal adenocarcinoma (PDAC). Several pathways (e.g., JAK-STAT, Smad, Wnt signaling, Hedgehog etc.), transcription factors and miRNAs have been implicated in the inflammatory and profibrogenic function of PSCs. The role of PSCs goes much beyond fibrosis/desmoplasia in PDAC. It is now shown that PSCs are involved in significant crosstalk between the pancreatic cancer cells and the cancer stroma. These interactions result in tumour progression, metastasis, tumour hypoxia, immune evasion and drug resistance. This is the rationale for therapeutic preclinical and clinical trials that have targeted PSCs and the cancer stroma.


Subject(s)
Carcinoma, Pancreatic Ductal/physiopathology , Pancreas, Exocrine/pathology , Pancreatic Neoplasms/physiopathology , Pancreatic Stellate Cells/metabolism , Pancreatic Stellate Cells/pathology , Pancreatitis, Chronic/physiopathology , Animals , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/immunology , Cytokines/metabolism , Extracellular Matrix Proteins/metabolism , Fibrosis , Humans , MicroRNAs/metabolism , Neoplasm Invasiveness , Pancreas, Exocrine/cytology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/immunology , Pancreatic Stellate Cells/drug effects , Pancreatic Stellate Cells/immunology , Pancreatitis, Chronic/drug therapy , Pancreatitis, Chronic/immunology , Signal Transduction , Tumor Microenvironment
13.
Int J Mol Sci ; 17(6)2016 Jun 03.
Article in English | MEDLINE | ID: mdl-27271610

ABSTRACT

Hypoxia inducible factor 1 (HIF-1) is a transcription factor composed of two subunits, namely, HIF-1α and HIF-1ß, in which HIF-1ß is constitutively expressed. HIF-1 upregulates several hypoxia-responsive proteins, including angiogenesis factors, glycolysis solution enzymes, and cell survival proteins. HIF-1 is also associated with the degree of inflammation in the tumor region, but the exact mechanism remains unclear. This study aims to identify the molecular mechanism of recruiting monocytes/macrophages by HIF-1α in pancreatic ductal adenocarcinoma (PDAC) and the effects of macrophages on pancreatic stellate cells (PSCs). Immunohistochemistry (IHC) was performed for cluster of differentiation 68 (CD68), HIF-1α, and chemical chemokines 2 (CCL2). Western blot, real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR), chromatin immunoprecipitation assay, and The Cancer Genome Atlas (TCGA) were used to verify the correlation between HIF-1α and CCL2 at protein and nucleic acid levels. Monocytes/macrophages were co-cultured with PSCs to observe their interaction. Samples showed significant correlation between CD68 and HIF-1α (t-test, p < 0.05). HIF-1α recruited monocytes/macrophages by promoting CCL2 secretion. Moreover, macrophages could accelerate the activation of PSCs. HIF-1α might promote inflammation and fibrosis of PDAC through CCL2 secretion, which may provide a novel target to treat PDAC patients.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Hypoxia-Inducible Factor 1/metabolism , Macrophages/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Stellate Cells/metabolism , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Biomarkers , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Chemokine CCL2 , Chemotaxis, Leukocyte , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Macrophages/immunology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/immunology
14.
Pancreas ; 45(1): 21-31, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26658038

ABSTRACT

Even though a strong association between inflammation and cancer has been widely accepted, the underlying precise molecular mechanisms are still largely unknown. A complex signaling network between tumor and stromal cells is responsible for the infiltration of inflammatory cells into the cancer microenvironment. Tumor stromal cells such as pancreatic stellate cells (PSCs) and immune cells create a microenvironment that protects cancer cells through a complex interaction, ultimately facilitating their local proliferation and their migration to different sites. Furthermore, PSCs have multiple functions related to local immunity, angiogenesis, inflammation, and fibrosis. Recently, many studies have shown that members of the phosphoinositol-3-phosphate kinase (PI3K) family are activated in tumor cells, PSCs, and tumor-infiltrating inflammatory cells to promote cancer growth. Proinflammatory cytokines and chemokines secreted by immune cells and fibroblasts within the tumor environment can activate the PI3K pathway both in cancer and inflammatory cells. In this review, we focus on the central role of the PI3K pathway in regulating the cross talk between immune/stromal cells and cancer cells. Understanding the role of the PI3K pathway in the development of chronic pancreatitis and cancer is crucial for the discovery of novel and efficacious treatment options.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Inflammation Mediators/metabolism , Pancreatic Neoplasms/enzymology , Pancreatic Stellate Cells/enzymology , Pancreatitis, Chronic/enzymology , Phosphatidylinositol 3-Kinase/metabolism , Animals , Cell Transformation, Neoplastic/pathology , Humans , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/immunology , Pancreatic Stellate Cells/immunology , Pancreatitis, Chronic/complications , Pancreatitis, Chronic/immunology , Prognosis , Risk Factors , Signal Transduction
15.
Hepatobiliary Pancreat Dis Int ; 14(6): 651-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26663014

ABSTRACT

BACKGROUND: Pancreatic stellate cells (PSCs) play a critical role in the pathogenesis of pancreatic fibrosis and have emerging functions as progenitor cells, immune cells or intermediaries in pancreatic exocrine secretion. Increasing evidence has shown that desmin as an exclusive cytoskeleton marker of PSC is only expressed in part of these cells. This study was to establish a desmin-positive PSC cell line and evaluate its actions on pancreatic fibrosis, inflammation and immunity. METHODS: The presence of cytoskeletal proteins, integrin α5ß1 or TLR4, was determined by immunocytochemistry while the production of desmin, collagen I, MMP-1, MMP-2, TIMP-2, or CD14 was evaluated by Western blotting. The levels of desmin, collagen I, IL-1 and IL-6 mRNA were determined by real-time quantitative PCR. The secretion of cytokines was detected by ELISA. Cell function was assessed using adhesion, migration, or proliferation assays. RESULTS: A stable activated rat PSC cell line (designated as RP-2) was established by RSV promoter/enhancer-driven SV40 large T antigen expression. RP-2 cells retained typical PSC properties, exhibited a myofibroblast-like phenotype and persistently produced desmin. The cells produced collagen I protein, matrix metalloproteinases and inhibitors thereof. RP-2 cells demonstrated typical PSC functions, including proliferation, adherence, and migration, the latter two of which occurred in response to fibronectin and were mediated by integrin α5ß1. TLR4 and its response genes including proinflammatory cytokines (IL-1, IL-6, TNF-alpha) and chemotactic cytokines (MCP-1, MIP-1α, Rantes) were produced by RP-2 cells and activated by LPS. LPS-induced IL-1 or IL-6 mRNA expression in this cell line was fully blocked with MyD88 inhibitor. CONCLUSION: RP-2 cells provide a novel tool for analyzing the properties and functions of PSCs in the pathogenesis of fibrosis, inflammation and immunity in the pancreas.


Subject(s)
Pancreatic Stellate Cells/metabolism , Pancreatitis/metabolism , Animals , Antigens, Polyomavirus Transforming/genetics , Cell Adhesion , Cell Line, Transformed , Cell Movement , Cell Proliferation , Collagen Type I/genetics , Collagen Type I/metabolism , Desmin/genetics , Desmin/metabolism , Fibrosis , Gene Expression Regulation , Integrin alpha5beta1/metabolism , Interleukin-1/genetics , Interleukin-1/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Lipopolysaccharide Receptors/metabolism , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 2/metabolism , Pancreatic Stellate Cells/immunology , Pancreatic Stellate Cells/pathology , Pancreatitis/immunology , Pancreatitis/pathology , Phenotype , Rats , Respiratory Syncytial Viruses/genetics , Signal Transduction , Tissue Inhibitor of Metalloproteinase-2/metabolism , Toll-Like Receptor 4/metabolism
16.
Gastroenterology ; 149(3): 765-76.e10, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26001927

ABSTRACT

BACKGROUND & AIMS: Little is known about the pathogenic mechanisms of chronic pancreatitis. We investigated the roles of complement component 5 (C5) in pancreatic fibrogenesis in mice and patients. METHODS: Chronic pancreatitis was induced by ligation of the midpancreatic duct, followed by a single supramaximal intraperitoneal injection of cerulein, in C57Bl6 (control) and C5-deficient mice. Some mice were given injections of 2 different antagonists of the receptor for C5a over 21 days. In a separate model, mice were given injections of cerulein for 10 weeks to induce chronic pancreatitis. Direct effects of C5 were studied in cultured primary cells. We performed genotype analysis for the single-nucleotide polymorphisms rs 17611 and rs 2300929 in C5 in patients with pancreatitis and healthy individuals (controls). Blood cells from 976 subjects were analyzed by transcriptional profiling. RESULTS: During the initial phase of pancreatitis, levels of pancreatic damage were similar between C5-deficient and control mice. During later stages of pancreatitis, C5-deficient mice and mice given injections of C5a-receptor antagonists developed significantly less pancreatic fibrosis than control mice. Primary pancreatic stellate cells were activated in vitro by C5a. There were no differences in the rs 2300929 SNP between subjects with or without pancreatitis, but the minor allele rs17611 was associated with a significant increase in levels of C5 in whole blood. CONCLUSIONS: In mice, loss of C5 or injection of a C5a-receptor antagonist significantly reduced the level of fibrosis of chronic pancreatitis, but this was not a consequence of milder disease in early stages of pancreatitis. C5 might be a therapeutic target for chronic pancreatitis.


Subject(s)
Complement C5/metabolism , Pancreatic Stellate Cells/metabolism , Pancreatitis, Chronic/metabolism , Aniline Compounds/pharmacology , Animals , Case-Control Studies , Ceruletide , Complement C5/deficiency , Complement C5/genetics , Disease Models, Animal , Fibrosis , Genetic Predisposition to Disease , Ligation , Mice, Inbred C57BL , Mice, Knockout , Pancreatic Ducts/surgery , Pancreatic Stellate Cells/drug effects , Pancreatic Stellate Cells/immunology , Pancreatic Stellate Cells/pathology , Pancreatitis, Chronic/chemically induced , Pancreatitis, Chronic/drug therapy , Pancreatitis, Chronic/genetics , Pancreatitis, Chronic/immunology , Pancreatitis, Chronic/pathology , Phenotype , Polymorphism, Single Nucleotide , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Receptor, Anaphylatoxin C5a/metabolism , Tetrahydronaphthalenes/pharmacology , Time Factors
17.
Oncotarget ; 5(22): 11064-80, 2014 Nov 30.
Article in English | MEDLINE | ID: mdl-25415223

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is characterized by an abundant desmoplastic reaction driven by pancreatic stellate cells (PSCs) that contributes to tumor progression. Here we sought to characterize the interactions between pancreatic cancer cells (PCCs) and PSCs that affect the inflammatory and immune response in pancreatic tumors. Conditioned media from mono- and cocultures of PSCs and PCCs were assayed for expression of cytokines and growth factors. IP-10/CXCL10 was the most highly induced chemokine in coculture of PSCs and PCCs. Its expression was induced in the PSCs by PCCs. IP-10 was elevated in human PDAC specimens, and positively correlated with high stroma content. Furthermore, gene expression of IP-10 and its receptor CXCR3 were significantly associated with the intratumoral presence of regulatory T cells (Tregs). In an independent cohort of 48 patients with resectable pancreatic ductal adenocarcinoma, high IP-10 expression levels correlated with decreased median overall survival. Finally, IP-10 stimulated the ex vivo recruitment of CXCR3+ effector T cells as well as CXCR3+ Tregs derived from patients with PDAC. Our findings suggest that, in pancreatic cancer, CXCR3+ Tregs can be recruited by IP-10 expressed by PSCs in the tumor stroma, leading to immunosuppressive and tumor-promoting effects.


Subject(s)
Carcinoma, Pancreatic Ductal/immunology , Chemokine CXCL10/biosynthesis , Pancreatic Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , HEK293 Cells , Humans , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Stellate Cells/immunology , Pancreatic Stellate Cells/metabolism , Pancreatic Stellate Cells/pathology , Stromal Cells/immunology , Stromal Cells/metabolism , Stromal Cells/pathology , Survival Analysis , T-Lymphocytes, Regulatory/pathology
18.
World J Gastroenterol ; 20(32): 11216-29, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-25170206

ABSTRACT

Pancreatic cancer is characterised by a prominent desmoplastic/stromal reaction that has received little attention until recent times. Given that treatments focusing on pancreatic cancer cells alone have failed to significantly improve patient outcome over many decades, research efforts have now moved to understanding the pathophysiology of the stromal reaction and its role in cancer progression. In this regard, our Group was the first to identify the cells (pancreatic stellate cells, PSCs) that produced the collagenous stroma of pancreatic cancer and to demonstrate that these cells interacted closely with cancer cells to facilitate local tumour growth and distant metastasis. Evidence is accumulating to indicate that stromal PSCs may also mediate angiogenesis, immune evasion and the well known resistance of pancreatic cancer to chemotherapy and radiotherapy. This review will summarise current knowledge regarding the critical role of pancreatic stellate cells and the stroma in pancreatic cancer biology and the therapeutic approaches being developed to target the stroma in a bid to improve the outcome of this devastating disease.


Subject(s)
Cell Communication , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/pathology , Stromal Cells/pathology , Animals , Biomarkers, Tumor/metabolism , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Extracellular Matrix/metabolism , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , Neovascularization, Pathologic , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/therapy , Pancreatic Stellate Cells/immunology , Pancreatic Stellate Cells/metabolism , Prognosis , Signal Transduction , Stromal Cells/immunology , Stromal Cells/metabolism , Tumor Escape , Tumor Microenvironment
19.
Pancreas ; 41(3): 422-7, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22415667

ABSTRACT

OBJECTIVES: Major histocompatibility complex (MHC) class II molecules are expressed on professional antigen-presenting cells (APCs), and pancreatic stellate cells (PSCs) have endocytic and phagocytic functions and play a role in the immune responses of the pancreas. The aim of the present study was to investigate whether PSCs exhibit features of APCs. METHODS: Rat and human PSCs were cultured with interferon-γ (IFN-γ) or an exogenous antigen, ovalbumin (OVA), and they were analyzed for expression of MHC II molecules by flow cytometry and reverse transcription-polymerase chain reaction. RESULTS: The cells simulated with IFN-γ expressed very little or no MHC class II molecules or human leukocyte antigen (HLA)-DR at the transcriptional level. Stimulation with IFN-γ failed to induce expression of MHC class II molecules and HLA-DR molecules according to the results of flow cytometry. Dual-color flow cytometric analysis showed that approximately 95% of the PSCs took up OVA; however, none of the cells that took up OVA expressed MHC class II molecules or HLA-DR molecules. CONCLUSIONS: Pancreatic stellate cells do not seem to be responsible for the MHC class II-dependent pathway of antigen presentation, suggesting that PSCs do not play a role in adaptive immunity as APCs.


Subject(s)
Antigen Presentation , Antigen-Presenting Cells/immunology , Pancreatic Stellate Cells/immunology , Animals , Cells, Cultured , Endocytosis , Flow Cytometry , Gene Expression Regulation , HLA-DR Antigens/genetics , HLA-DR Antigens/metabolism , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Humans , Interferon-gamma/metabolism , Male , Ovalbumin/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
20.
Int J Cancer ; 130(10): 2337-48, 2012 May 15.
Article in English | MEDLINE | ID: mdl-21780106

ABSTRACT

Galectin-1 is implicated in making tumor cells immune privileged, in part by regulating the survival of infiltrating T cells. Galectin-1 is strongly expressed in activated pancreatic stellate cells (PSCs); however, whether this is linked to tumor cell immune escape in pancreatic cancer is unknown. Galectin-1 was knocked down in PSCs isolated from pancreatic tissues using small interfering RNA (siRNA), or overexpressed using recombinant lentiviruses, and the PSCs were cocultured with T cells. CD3(+) , CD4(+) and CD8(+) T cell apoptosis was detected by flow cytometry; T cell IL-2, IL-4, IL-5 and INF-γ production levels were quantified using ELISA. Immunohistochemical analysis showed increased numbers of PSCs expressed Galectin-1 (p < 0.01) and pancreatic cancers had increased CD3(+) T cell densities (p < 0.01) compared to normal pancreas or chronic pancreatitis samples. In coculture experiments, PSCs that overexpressed Galectin-1 induced apoptosis of CD4(+) T cells (p < 0.01) and CD8(+) T cells (p < 0.05) significantly, compared to normal PSCs. Knockdown of Galectin-1 in PSCs increased CD4(+) T cell (p < 0.01) and CD8(+) T cell viability (p < 0.05). Supernatants from T cells cocultured with PSCs that overexpressed Galectin-1 contained significantly increased levels of Th2 cytokines (IL-4 and IL-5, p < 0.01) and decreased Th1 cytokines (IL-2 and INF-γ, p < 0.01). However, the knockdown of PSC Galectin-1 had the opposite effect on Th1 and Th2 cytokine secretion. Our study suggests that the overexpression of Galectin-1 in PSCs induced T cell apoptosis and Th2 cytokine secretion, which may regulate PSC-dependent immunoprivilege in the pancreatic cancer microenvironment. Galectin-1 may provide a novel candidate target for pancreatic cancer immunotherapy.


Subject(s)
Galectin 1/metabolism , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/metabolism , Pancreatic Stellate Cells/metabolism , Tumor Escape , Adult , Aged , Aged, 80 and over , CD3 Complex/metabolism , Female , Galectin 1/genetics , Gene Knockdown Techniques , Humans , Interleukins/metabolism , Male , Middle Aged , Pancreatic Stellate Cells/immunology , T-Lymphocytes/metabolism , Tumor Microenvironment/immunology
SELECTION OF CITATIONS
SEARCH DETAIL