Abstract
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.Free full text
Activated Pancreatic Stellate Cells Sequester CD8+ T-Cells to Reduce Their Infiltration of the Juxtatumoral Compartment of Pancreatic Ductal Adenocarcinoma
Associated Data
Abstract
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.
Method
We used tissue microarray analysis to compare immune cell infiltration of different pancreatico-biliary 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 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 PDAC patients had increased chemotaxis towards activated PSCs, which secrete CXCL12, compared with quiescent PSC or tumor cells. These effects could be reversed by knockdown of CXCL12 or treatment of PSCs with ATRA.
Conclusion
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 anti-tumor immune response.
INTRODUCTION
Despite our increased understanding of pancreatic ductal adenocarcinoma (PDAC) from transgenic mouse models,1 this human disease remains resistant to systemic therapies. Genomic and epigenetic alterations that are characteristic of all cancers provide a diverse set of antigens that immune cells can use to distinguish and selectively target malignant cells.2-4 The importance of tumor-infiltrating lymphocytes, in particular anti-tumor CD8+ cytotoxic T-cell location, has been highlighted by their prognostic value in many human cancers.2-7 However, similar immunohistological analysis investigating the correlation of T-cell infiltration with pancreatico-bilary cancer prognosis has been understudied to date. Increased infiltration of immunosuppressive cells such as tumor-associated macrophages (TAMs), myeloid derived suppressor cells (MDSCs) and regulatory T-cells (Tregs) has been detected in the inflammatory tumor microenvironment of genetic mouse models of PDAC (including ‘KPC’ mice: LSLKrasG12D/+; LSL-Trp53R172H/+; Pdx-1-Cre) during disease progression to invasive cancer.8 In human disease, increased Tregs infiltration was found to be increased in PDAC tissue when compared to non-malignant pancreas, whereas no differences were observed in peripheral blood mononuclear cells of healthy donors compared to cancer patients.9
PDAC is characterized by a pronounced desmoplastic reaction mediated by the activation of quiescent pancreatic stellate cells (PSC).10 We have previously demonstrated that this critical event is reversible.11 Importantly, in situ hybridization studies from other groups have demonstrated differential gene expression signatures when comparing defined stromal compartments in PDAC that include the juxtatumoral stroma (immediately adjacent to the tumor epithelial cells) and the panstromal compartment (non-adjacent to tumor cells).12, 13 In this present study, we show for the first time that there is differential immune cell infiltrate unique to these PDAC stromal compartments and that enhanced juxtatumoral CD8+ T-cell infiltrate correlates with better patient survival. Our experimental assays, including the use of the KPC PDAC mouse model, show that CD8+ T-cells are prevented from targeting PDAC cells by activated PSC that reside in the panstromal compartment. Of translational relevance, rendering activated PSC quiescent reduced PDAC patient CD8+ T-cell in vitro migration and adhesion towards PSC and allowed enhanced in vivo juxtatumoral infiltration of CD8+ T-cells.
MATERIALS AND METHODS
Tissue microarray, staining and analysis
Tissue microarrays were constructed with pancreatic tissues collected at cancer resection or biopsy (Supplementary Tables 1-2) at Barts Health NHS Trust (City and East London Research Ethics Committee (REC) 07/0705/87) as described before.14 Regions of tumor, stroma and normal pancreas were marked on Hematoxylin and Eosin (H&E) stained slides of the donor tissue blocks and three 1mm cores of each region were sampled per patient using the Tissue Arrayer Minicore® 3 (Alphelys, Plaisir, France). The TMA’s and whole sections (all formalin-fixed paraffin embedded 4μm sections) were stained with various antibodies (Supplementary Table 3).
Ariol imaging analysis system (Genetix, New Milton, UK) was used to determine immune cell densities. Briefly, software is trained by the user to distinguish and quantify positive (brown: DAB reaction) and negative (blue: hematoxylin counter-stain) cells by their color, shape, size and staining intensity characteristics as described before.15 The juxtatumoral compartment was delineated by using the marker tool to draw around cancer cells (approximately 100μm) in all TMA cores. Panstromal density of immune cell infiltrate was calculated by subtracting the density of the juxtatumoral compartment from the density of the whole TMA core. The median of all analyzed tumor cores (n=6) for each patient was used. Different methods were used to determine immune cell density (Supplementary Figures 1-4). Statistical tests were carried out in Prism software (Graphpad). The X-tile software v.3.6.116, 17 (Yale University) was used to analyze the impact of immune cell infiltrates on patient prognosis by the minimum p-value method.15 Miller-Seigmund p-value corrections were applied.18
Mouse model of PDAC
KPC mice were treated with all-trans retinoic acid (ATRA) dissolved in sesame oil at a dose of 10mg/kg administered daily for 5 days (n=6) or vehicle (n=4), as described before (Supplementary Table 4).11 The whole diseased pancreas, spleen (positive control) and salivary gland (negative control) were stained for immune cells on a single slide.
Culture conditions
Stellate cells (PS1) and primary PSC (pPSC) were rendered quiescent (qPSC) by daily treatment with 1μM ATRA under subdued light conditions over seven days.11 PSCs treated with vehicle (ethanol) were used as activated PSC (aPSC) phenotype. Conditioned media from cancer cells lines (AsPc1, Capan1) or PSC was collected.
Migration assay
Healthy donor (National Blood Service (Brentwood, UK): North London REC) or PDAC patient (Brent REC 05/Q0408/65) lymphocytes were obtained from buffy coats. CD3+, CD4+ CD19+, CD56+ and CD8+ T-cells were negatively selected using magnetic-activated cell sorting cell isolation kits (Miltenyi Biotec, Surrey, UK).
1 × 105 CD3+, CD4+ CD8+, CD19+ or CD56+ cells in 200 μl of FBS-replete RPMI-1640 were incubated for four-eight hours in sterile, 5.0μm pore Transwells™ (Corning) to assess the differential migration of T-cells to the conditioned media (500 μl). The migrated cells were then harvested after eight hours of incubation and cells counted with the Casy counter (Roche, UK). Background migration (unsupplemented media) was subtracted. All migration was relative to media with fetal bovine serum (FBS) to enable comparison across biological repeats.
Adhesion assays
Activated or quiescent PSC were incubated with 5×104 CD3+ at 37°C for one hour, washed repeatedly, fixed with Cellstor (4% PFA, 20 minutes; Cellpath, Newton, UK) and the number of adherent T-cells relative to PSC were counted based on cell morphologies and staining patterns. A median of counts from six fields gave a data point for one patient. Each experiment was repeated in triplicate.
Gene-expression
Re-analysis of gene expression microarrays of qPSC compared to aPSC11 for significantly differentially expressed genes performed using DAVID.19
Enzyme linked immunosorbent assay (ELISA) and qPCR
Quantikine® ELISA kits (R & D Systems, Abingdon, UK) were used to measure the concentrations of candidate chemokines (manufacturer’s instructions). qPCR were carried out as described.20
RNAi
5nm CXCL12 GeneSolution siRNA (Qiagen, West Sussex, UK) was used alongside All-stars Negative Control siRNA (Qiagen, West Sussex, UK) with INTERFERin™ (Polyplus-transfection SA, France) in Opti-MEM® reduced serum media (Life Technologies Ltd. Paisley, UK) as per manufacturer’s instructions to perform RNAi experiments. Recombinant human CXCL12 (R&D Systems, Abingdon, UK) was used as a positive control.
RESULTS
Immune cell infiltration in pancreatico-biliary diseases (PBD)
We performed an unbiased, automated evaluation of immune cell infiltrate for pancreatico-biliary cancers such as PDAC, ampullary carcinoma (AC) and cholangiocarcinoma (CC) using dedicated tissue micro-arrays (TMA). These tumors were compared alongside other PBD including borderline malignant condition: mucinous cystic neoplasm (MCN); chronic inflammation: chronic pancreatitis (CP); and normal donor control TMAs (Supplementary Tables 1-2). Our immune cell density measurements were validated in whole tissue sections and by two distinct methods: as a percentage of all cells and by cell numbers per unit area (Supplementary Figures 1-4). Our results showed that immune cell densities were significantly increased in malignant (PDAC, CC, AC) tumors that are associated with an adverse prognosis,21 compared to other PBD and healthy controls (Figure 1). Increased Treg (FoxP3+) infiltrate in human PDAC was confirmed as reported before.9 In contrast, the density of CD8+ T-cell infiltrate in PDAC was comparable to chronic pancreatitis but significantly reduced compared to AC and CC (P<0.01). These findings in human PDAC when compared with other PBD, reported for the first time, are consistent with reduced infiltration of CD8+ cytotoxic T-cells recently reported in the KPC mouse model of PDAC.8, 22, 23
Differential immune cell infiltrate in stromal sub-compartments
We next characterized the immune infiltrate within stromal sub-compartments of PDAC and identified that cytotoxic effector T-cells (CD8+), Tregs (FoxP3+), B-cells (CD20+) and natural killer or NK-cells (CD56+) were significantly reduced in juxtatumoral compartments compared to the panstromal areas. This was in contrast to neutrophil (myeloperoxidase+) and macrophage (CD68+) stromal infiltration (Figure 2, Supplementary Figure 5).13 This differential immune cell infiltrate, particularly for CD8+ T-cells, was confirmed in an independent TMA cohort of PDAC patients composed mainly of advanced cancers (Supplementary Figure 6). Interestingly, this differential immune cell infiltrate was not observed in AC (Supplementary Figure 7) or CC (Supplementary Figure 8). Our analysis showed that tissue immune cell infiltrate had no correlation with circulating immune cell counts (Supplementary Figures 9-11). The paucity of functional vasculature close to the PDAC tumor cells (juxtatumoral) is well established.24 Taken together these observations strongly suggest a hitherto unreported PDAC-specific CD8+ T-cell trafficking defect within human PDAC tissues.
Our data further demonstrates for the first time that a higher CD8+ T-cell juxtatumoral infiltrate is associated with improved survival post-surgical resection as analyzed by X-Tile software (Figure 3, Supplementary Table 5).17, 25 Of interest, a lower juxtatumoral Treg (FoxP3+) infiltrate also demonstrated a pro-survival effect in PDAC patients. Taken together, this data suggests that reduced infiltration of anti-tumor CD8+ T-cells to target tumor epithelial cells in the PDAC microenvironment has important clinical impact.
Reversal of the juxtatumoral exclusion of cytotoxic T-cells in murine PDAC
We next identified similar juxtatumoral exclusion of CD8+ T-cells and CD45R+ B-cells, but not F4/80+ macrophages in the PDAC microenvironment of KPC mice (Figure 4, Supplementary Figure 12). Previously we have demonstrated that treating these PDAC mice with ATRA renders activated pancreatic stellate cells (PSC) quiescent leading to stromal collapse and a detrimental effect on resident tumor cells through a multitude of signaling cascades.11 In the present study, we found that ATRA treatment enhanced juxtatumoral CD8+ T-cell infiltration but not CD45R+ B-cells or F4/80+ macrophages. Recent studies suggest that MDSCs have an important role in the PDAC-associated CD8+ T-cell infiltration defect.22, 23 Thus, we examined this population of immune cells in our ATRA-treated KPC mice experiments but did not observe any changes in MDSC (CD11b+) infiltrate. Thus, it is conceivable that activated PSC in PDAC sequester anti-tumor CD8+ T-cell migratory and adhesive function preventing access to the tumoral cells. Since it is not technically feasible to study the dynamic spatial interactions between PSC and immune cells by current in vivo models, we designed novel in vitro chemotaxis assays examining the possible immunomodulatory role of PSC.
CD8+ T-cells migrate preferentially towards activated PSC
Quiescent (normal pancreas) and activated (PDAC) PSCs have distinct phenotypes.11 Our chemotaxis assays showed that healthy donor CD3+ T-cells adhered and migrated preferentially to activated PSC (aPSC) conditioned medium compared to quiescent PSC (qPSC) or cancer cell (Capan1 and AsPc1) conditioned medium that contained serum (Figure 5, Supplementary Figure 13). Notably, quiescent PSC induced chemotactic responses were remarkably similar to baseline levels.
In line with our in vivo histological observations, the differential migration of CD4+ helper T-cells towards cancer cells or PSC was not as significant when compared to CD8+ T-cell experiments (Figure 5C,D). CD56+ NK-cells showed preferential migration to aPSC compared to qPSC, while CD19+ B-cells did not (Figure 5E,F). Together with our earlier KPC mouse study findings, we believe that these results suggest that panstromal-associated aPSC attract and adhere to CD8+ T-cells preventing their effective juxtatumoral infiltration.
CXCL12 mediates the increased T-cell migration towards aPSC
Re-analysis of our previous Affymetrix gene expression data of PSC in PDAC revealed that aPSC alter the expression of a number of cytokines/chemokines and adhesion-mediating molecules compared to qPSC (Figure 6A, Supplementary Figure 14).11 The transcriptomic changes for a number of these molecules was validated at the protein level both in vitro and in vivo in human PDAC by others26 and us (Figure 6B-E).11, 20 Notably, fibronectin expression was markedly increased in human PDAC (especially in the panstromal compartment compared to normal pancreas, Figure 6B). Fibronectin expression in murine PDAC was markedly reduced following ATRA treatment (Figure 6C).
Of the multitude of signaling cascades/adhesion molecules deregulated with an activated PSC phenotype, we chose to study upregulated CXCL12 in aPSC because of its well-established chemotactic properties for T-cells.27 CXCL12 knock-down in both in vitro aPSC and primary PDAC patient PSC resulted in a reduction of healthy donor and PDAC patient CD8+ and CD4+ T-cells T-cell migration to levels comparable to qPSC (Figure 7, Supplementary Figure 17). Our flow cytometric analysis showed that whilst the circulating effector/memory T-cell ratio28, 29 was similar in PDAC patients compared to age-matched healthy donors (Supplementary Figures 15-16), the CXCR4 expression was markedly upregulated in PDAC patient T-cells (Figure 7F, Supplementary Figure 17A). Taken together, this data suggests an important role for the CXCL12/CXCR4 axis in regulating T-cell trafficking.
DISCUSSION
PDAC has a 5-year overall patient survival of 3-5% 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 PBD. We show an increasing immune cell infiltrate from normal (healthy pancreas tissue) to inflammatory (CP), borderline (MCN) and to malignant (AC, PDAC and CC) diseases. To our knowledge, we identify for the first time that higher CD8+ T-cell infiltration in PDAC tumors correlates with better patient survival, a finding that that provides evidence for anti-tumor CD8+ T-cell cancer immunoediting in human PDAC.2
The PDAC stromal microenvironment is composed of activated myo-fibroblasts including aPSC, extracellular matrix, blood vessels and various immune cells, whose individual contribution to tumor progression remains elusive.26 The PDAC stroma has been classified into two compartments based on differential gene expression:13, 30 the juxtatumoral and panstromal compartments. Our immunohistochemistry analysis revealed that whilst some immune cells (CD68+ macrophages, myeloperoxidase+ neutrophils) infiltrate the juxtatumoral compartment, others (CD8+ T-cells, FoxP3+ Tregs CD56+ NK-cells, CD20+ B-cells) are sequestrated in the panstromal compartment. The use of the KPC mouse model of PDAC that exhibits an intact in situ cellular desmoplastic component allowed us to confirm panstromal sequestration of CD8+ T-cells and CD45R+ B-cells, but not CD4+ T-cells or F4/80+ macrophages, in agreement with the human data.
More importantly, we could demonstrate that in vivo ATRA treatment, that converts aPSC to a qPSC phenotype11, specifically enhanced CD8+ T-cell infiltration towards tumor cells but no change was demonstrable for the CD4+ T-cells, CD45R+ B-cells, F4/80+ macrophages and MDSC (CD11b+) infiltrate. In the absence of appropriate tools we were unable to define MDSC infiltrate in stromal tissue compartments in human PDAC.31 MDSCs have been postulated to drive CD8+ T-cells exclusion recently in murine KPC-derived subcutaneous PDAC models devoid of in situ cellular desmoplastic component, particularly stellate cells.22, 23 Of interest, in vitro but not in vivo studies demonstrate the PSC may modulate differentiation of MDSC in an IL6-STAT3 dependent manner.32 Depletion of FAPα+ stromal cells in a subcutaneous mouse model of PDAC resulted in immunological control (CD8+ T-cell-mediated) of tumor growth, a phenomenon not seen in Rag-2 deficient mice.33 The relationship between FAPα cells and PSC remains to be established. Furthermore, in a cohort of surgically incurable PDAC patients and in KPC mice, combining gemcitabine therapy with an agonist CD40 antibody led to significant tumor regression. CD40-activated macrophages were found to infiltrate the tumors to facilitate the depletion of the stroma. This depletion of tumor stroma was associated with tumor immunoclearance.34 Our data suggest CD8+ T-cells may be involved in tumor immunoclearance if their trafficking is unimpeded by the ‘PDAC stromal barrier’, a finding substantiated by murine PDAC investigations.8, 34
Our ex vivo migratory bioassays using both healthy donor and PDAC patient cytotoxic CD8+, CD4+ helper T-cells and NK-cells showed that aPSC conditioned media attracted increased numbers of these lymphocytes compared to tumor cells or qPSC. Taken together, these data provide evidence that aPSC are key regulators of the immune cell infiltrate in the PDAC stromal microenvironment. Understanding the complex temporal (disease progression) and spatial (stromal compartment) relationship between aPSC, FAPα+ stromal cells, CD8+ T-cells, MDSC, Tregs and NK-cells in the PDAC disease process will be important for identifying novel cellular and molecular targets for therapy.35
Recently, we demonstrated that aPSC modulate a range of cellular process in tumor cells in a dose-dependent manner. These findings suggest a vital role for aPSC in PDAC disease progression mediated through perturbation of multitude of signaling cascades.20 Our analysis of the transcriptome of PDAC-associated aPSC revealed deregulated adhesion and chemokine/cytokine genes.11 We hypothesized that these molecules likely contribute to stromal microenvironment-specific regulation of immune cell trafficking and infiltration into the tumor.35,36 In this present study, we focused on CXCL12 that was upregulated in aPSC and is known to be an important chemoattractant for T-cells.27 Our chemotaxis assays showed that both donor and PDAC patient CD8+ and CD4+ T-cells showed elevated migration towards aPSC compared to qPSC that was CXCL12-dependent in CD8+ T-cells. CXCL12-siRNA treated aPSC cells or conversion of aPSC to qPSC reduced CXCL12 expression and T-cell chemotaxis. Notably, we show that PDAC patient T-cells express higher levels of the CXCL12 receptor (CXCR4) compared to healthy donor T-cells, but with no change in the proportion of effector or memory T-cells. Taken together, this data suggests an important role for stromal PSC:T-cell interaction and the CXCL12/CXCR4 axis in PDAC. Moreover, CXCL12 has been implicated in the maintenance of PDAC cancer stem cells in a PSC niche by regulating the expression of sonic hedgehog.37, 38
Further work remains to be done to delineate the relative contribution of NK-cells and Treg cells, both of which are also be sequestered in the panstromal compartment in human PDAC. Of note, it has been suggested that this intra-stromal cross-talk (tumor immuno-suppressive network) can further impede CD8+ T-cell trafficking and immuno-editing.35 The relative contribution of CXCR4/CXCL12 and other cytokines/chemokines/adhesion molecules such as IL-8, fibronectin amongst others will need to be precisely defined to refine therapeutic strategies.
Taken together, these data suggest that the complex tumor microenvironment, in particular, the increasingly recognized PSCs11, 20, 26 regulate the tumor immune response. We demonstrate that PDAC-associated aPSC orchestrate many such interactions by sequestration of CD8+, CD4+ T-cells, NK-cells and Tregs in the panstromal compartment. Our data provides the basis for further studies to explore this clinically significant, yet unrecognized, stromal-immune cell cross-talk in PDAC. Successful targeting of desmoplastic and immune stroma cells may provide an interesting approach in tackling human PDAC in an attempt to improve survival in this incurable disease.
Acknowledgements
We thank Dr Christine Feig (Cambridge Research Institute, UK) and Prof David A Tuveson (Cold Spring Harbor Laboratory, NY, USA) for helpful comments regarding murine experiments. We are very grateful to members of our laboratories for helpful discussions and suggestions. We also thank Dr Lisa Mears (Department of Pathology, Barts and the London) and Dr Moffadal Moonim (Department of Pathology, St Thomas’ Hospital) with human PDAC samples. We thank Prof Federica Marelli-Berg for her critical insight.
Grant Support: AEO is supported by a PhD studentship from Cross-River University of Technology, Nigeria. JGG acknowledges funding from National Institutes of Health, USA and Cancer Research UK. AJC, JW and AGR acknowledge support of Barts and the London Charity. AGR acknowledges funding from the European Hematology Association. HMK acknowledges support from National Institute of Health Research, UK.
Abbreviations
AC | Ampullary carcinoma |
aPSC | Activated Pancreatic Stellate Cells |
ATRA | All-Trans Retinoic Acid |
CC | Cholangiocarcinoma |
CCL2 | Chemokine (C-C motif) Ligand 2 |
CM | Conditioned Media |
CP | Chronic Pancreatitis |
CXCL12 | Chemokine (C-X-C Motif) Ligand 12 |
CXCR4 | Chemokine (C-X-C Motif) Receptor 4 |
FBS | Fetal Bovine Serum |
IL8 | Interleukin 8 |
KPC | LSL-KrasG12D/+; LSL-Trp53R172H/+; Pdx-1-Cre; |
MCN | Mucinous cystic neoplasm |
MCP1 | Monocyte Chemo-attractant Protein 1 |
MDSC | Myeloid Derived Suppressor Cells |
PDAC | Pancreatic Ductal Adenocarcinoma |
PSC | Pancreatic Stellate Cells |
qPSC | Quiescent Pancreatic Stellate Cells |
pPSC | Primary Pancreatic Stellate Cells |
SDF1α | Stromal cell Derived Factor 1 alpha |
TAMs | Tumor Associated Macrophages |
TMA | Tissue Micro-Array |
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Disclosures: All authors have nothing to disclose.
Author contributions: Study concept and design: HMK, JFM, CC, JGG, AGR, AEO; acquisition of data: AEO, AJC, JeW, JuW, RF, JCR, JAC, HMK; analysis and interpretation of data: all authors; drafting of the manuscript: HMK, AGR, AEO; critical revision of the manuscript for important intellectual content: all authors; statistical analysis: AEO, JuW, CC, HMK; obtained funding: AEO, HMK, JGG, AGR; technical, or material support: HMK, AJC, AGR, JGG; study supervision: HMK.
*Author names in bold designate shared co-first authorship.
REFERENCES
Full text links
Read article at publisher's site: https://doi.org/10.1053/j.gastro.2013.07.025
Read article for free, from open access legal sources, via Unpaywall: http://www.gastrojournal.org/article/S0016508513010767/pdf
Citations & impact
Impact metrics
Citations of article over time
Alternative metrics
Smart citations by scite.ai
Explore citation contexts and check if this article has been
supported or disputed.
https://scite.ai/reports/10.1053/j.gastro.2013.07.025
Article citations
TAK1 Promotes an Immunosuppressive Tumor Microenvironment through Cancer-Associated Fibroblast Phenotypic Conversion in Pancreatic Ductal Adenocarcinoma.
Clin Cancer Res, 30(22):5138-5153, 01 Nov 2024
Cited by: 0 articles | PMID: 39264265 | PMCID: PMC11565170
Cold and hot tumors: from molecular mechanisms to targeted therapy.
Signal Transduct Target Ther, 9(1):274, 18 Oct 2024
Cited by: 0 articles | PMID: 39420203 | PMCID: PMC11491057
Review Free full text in Europe PMC
Multifaceted bioinformatic analysis of m6A-related ferroptosis and its link with gene signatures and tumour-infiltrating immune cells in gliomas.
J Cell Mol Med, 28(17):e70060, 01 Sep 2024
Cited by: 0 articles | PMID: 39248438 | PMCID: PMC11382363
The role of cancer-associated fibroblasts and exosomal miRNAs-mediated intercellular communication in the tumor microenvironment and the biology of carcinogenesis: a systematic review.
Cell Death Discov, 10(1):380, 26 Aug 2024
Cited by: 0 articles | PMID: 39187523 | PMCID: PMC11347635
Review Free full text in Europe PMC
Precision Targeting Strategies in Pancreatic Cancer: The Role of Tumor Microenvironment.
Cancers (Basel), 16(16):2876, 19 Aug 2024
Cited by: 0 articles | PMID: 39199647 | PMCID: PMC11352254
Review Free full text in Europe PMC
Go to all (329) article citations
Data
Data behind the article
This data has been text mined from the article, or deposited into data resources.
BioStudies: supplemental material and supporting data
Quick GO
- (1 citation) GO - GO0007155
Similar Articles
To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.
Retinoic acid-induced pancreatic stellate cell quiescence reduces paracrine Wnt-β-catenin signaling to slow tumor progression.
Gastroenterology, 141(4):1486-97, 1497.e1-14, 24 Jun 2011
Cited by: 228 articles | PMID: 21704588
NFκB in Pancreatic Stellate Cells Reduces Infiltration of Tumors by Cytotoxic T Cells and Killing of Cancer Cells, via Up-regulation of CXCL12.
Gastroenterology, 155(3):880-891.e8, 29 Jul 2018
Cited by: 90 articles | PMID: 29909021 | PMCID: PMC6679683
Pancreatic stellate cell secreted IL-6 stimulates STAT3 dependent invasiveness of pancreatic intraepithelial neoplasia and cancer cells.
Oncotarget, 7(40):65982-65992, 01 Oct 2016
Cited by: 63 articles | PMID: 27602757 | PMCID: PMC5323208
Pancreatic stellate cells and pancreas cancer: current perspectives and future strategies.
Eur J Cancer, 50(15):2570-2582, 01 Aug 2014
Cited by: 64 articles | PMID: 25091797
Review
Funding
Funders who supported this work.
Cancer Research UK (1)
Genomics & Molecular Pathology of Pancreatic Cancer
Professor Nick Lemoine, Queen Mary, University of London
Grant ID: 12008
NCI NIH HHS (1)
Grant ID: P20 CA081534