201
|
Porciuncula A, Hajdu C, David G. The Dual Role of Senescence in Pancreatic Ductal Adenocarcinoma. Adv Cancer Res 2016; 131:1-20. [PMID: 27451122 DOI: 10.1016/bs.acr.2016.05.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The role of senescence as a tumor suppressor is well established; however, recent evidence has revealed novel paracrine functions for senescent cells in relation to their microenvironment, most notably protumorigenic roles in certain contexts. Senescent cells are capable of altering the inflammatory microenvironment through the senescence-associated secretory phenotype, which could have important consequences for tumorigenesis. The role of senescent cells in a highly inflammatory cancer like pancreatic cancer is still largely undefined, apart from the fact that senescence abrogation increases tumorigenesis in vivo. This review will summarize our current knowledge of the phenomenon of cellular senescence in pancreatic ductal adenocarcinoma, its overlapping link with inflammation, and some urgent unanswered questions in the field.
Collapse
Affiliation(s)
- A Porciuncula
- NYU Cancer Institute, New York University School of Medicine, New York, NY, United States
| | - C Hajdu
- New York University School of Medicine, New York, NY, United States
| | - G David
- NYU Cancer Institute, New York University School of Medicine, New York, NY, United States.
| |
Collapse
|
202
|
Mei L, Du W, Ma WW. Targeting stromal microenvironment in pancreatic ductal adenocarcinoma: controversies and promises. J Gastrointest Oncol 2016; 7:487-94. [PMID: 27284483 DOI: 10.21037/jgo.2016.03.03] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Pancreatic cancer is a highly lethal disease. Conventional therapeutics targeting pancreas cancer cell compartment using cytotoxics improved patient survival but at the expense of significant toxicity. Microscopically, the tumor is characterized by thick desmoplastic stroma that surrounds islands of pancreatic cancer cells. The tumor microenvironment has been found to play important roles in carcinogenesis, the development of drug resistance, and mediating immunosuppression. The understanding the tumor-stromal interaction has led to the development of novel therapeutic approaches. Here, we review the strategies that are currently in (or, near to) clinical evaluation and the underlying preclinical rationales.
Collapse
Affiliation(s)
- Lin Mei
- 1 Department of Medicine, 2 Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Wei Du
- 1 Department of Medicine, 2 Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Wen Wee Ma
- 1 Department of Medicine, 2 Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| |
Collapse
|
203
|
Hamada S, Masamune A, Yoshida N, Takikawa T, Shimosegawa T. IL-6/STAT3 Plays a Regulatory Role in the Interaction Between Pancreatic Stellate Cells and Cancer Cells. Dig Dis Sci 2016; 61:1561-71. [PMID: 26738736 DOI: 10.1007/s10620-015-4001-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 12/11/2015] [Indexed: 12/18/2022]
Abstract
BACKGROUND Pancreatic stellate cells (PSCs) play a pivotal role in pancreatic fibrosis, a characteristic feature of pancreatic cancer. Although it is still controversial, previous studies have suggested that PSCs promote the progression of pancreatic cancer by regulating the cell functions of cancer cells. PSCs produce large amounts of IL-6, which promotes the accumulation of myeloid-derived suppressor cells via a signal transducers and activator of transcription 3 (STAT3)-dependent mechanism. But the role of IL-6/STAT3 pathway in the interaction between PSCs and pancreatic cancer cells remains largely unknown. AIMS To clarify the role of IL-6/STAT3 in the interaction between PSCs and cancer cells. METHODS Human pancreatic cancer cells (Panc-1 and SUIT-2 cells) were treated with conditioned medium of immortalized human PSCs (PSC-CM). The effects of PSC-CM and IL-6 neutralization on the mRNA expression profiles were examined using Agilent's microarray. Activation of STAT3 was assessed by Western blotting using an anti-phospho-specific antibody. Cellular migration was examined by a two-chamber assay. The expression of markers related to epithelial-mesenchymal transition (EMT) was assessed by real-time reverse transcription PCR. RESULTS PSC-CM induced the activation of STAT3 in pancreatic cancer cells. Neutralization of IL-6 suppressed the PSC-CM-induced upregulation of genes including complement factor B, lipocalin, and chemokine (C-C motif) ligand 20. Inhibition of IL-6/STAT3 pathway by anti-IL-6 antibody or a STAT3 inhibitor (NSC74859) inhibited the PSC-CM-induced migration and the expression of EMT-related markers (Snail and cadherin-2) in pancreatic cancer cells. CONCLUSION IL-6/STAT3 pathway regulates the PSC-induced EMT and alterations in gene expression in pancreatic cancer cells.
Collapse
Affiliation(s)
- Shin Hamada
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan
| | - Atsushi Masamune
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan.
| | - Naoki Yoshida
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan
| | - Tetsuya Takikawa
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan
| | - Tooru Shimosegawa
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan
| |
Collapse
|
204
|
Abstract
The local extension of cancer cells along nerves is a frequent clinical finding for various tumours. Traditionally, nerve invasion was assumed to occur via the path of least resistance; however, recent animal models and human studies have revealed that cancer cells have an innate ability to actively migrate along axons in a mechanism called neural tracking. The tendency of cancer cells to track along nerves is supported by various cell types in the perineural niche that secrete multiple growth factors and chemokines. We propose that the perineural niche should be considered part of the tumour microenvironment, describe the molecular cues that facilitate neural tracking and suggest methods for its inhibition.
Collapse
Affiliation(s)
- Moran Amit
- Laboratory for Applied Cancer Research, Department of Otolaryngology Head and Neck Surgery, Head and Neck Center, Rambam Healthcare Campus, Clinical Research Institute at Rambam, Rappaport Institute of Medicine and Research, The Technion-Israel Institute of Technology, Haalia Street No. 8, Haifa, Israel
| | - Shorook Na'ara
- Laboratory for Applied Cancer Research, Department of Otolaryngology Head and Neck Surgery, Head and Neck Center, Rambam Healthcare Campus, Clinical Research Institute at Rambam, Rappaport Institute of Medicine and Research, The Technion-Israel Institute of Technology, Haalia Street No. 8, Haifa, Israel
| | - Ziv Gil
- Laboratory for Applied Cancer Research, Department of Otolaryngology Head and Neck Surgery, Head and Neck Center, Rambam Healthcare Campus, Clinical Research Institute at Rambam, Rappaport Institute of Medicine and Research, The Technion-Israel Institute of Technology, Haalia Street No. 8, Haifa, Israel
| |
Collapse
|
205
|
Alexander J, Cukierman E. Stromal dynamic reciprocity in cancer: intricacies of fibroblastic-ECM interactions. Curr Opin Cell Biol 2016; 42:80-93. [PMID: 27214794 DOI: 10.1016/j.ceb.2016.05.002] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 05/03/2016] [Accepted: 05/04/2016] [Indexed: 12/18/2022]
Abstract
Stromal dynamic reciprocity (SDR) consists of the biophysical and biochemical interplay between connective tissue elements that regulate and maintain organ homeostasis. In epithelial cancers, chronic alterations of SDR result in the once tumor-restrictive stroma evolving into a 'new' tumor-permissive environment. This altered stroma, known as desmoplasia, is initiated and maintained by cancer associated fibroblasts (CAFs) that remodel the extracellular matrix (ECM). Desmoplasia fuels a vicious cycle of stromal dissemination enriching both CAFs and desmoplastic ECM. Targeting specific drivers of desmoplasia, such as CAFs, either enhances or halts tumor growth and progression. These conflicting effects suggest that stromal interactions are not fully understood. This review highlights known fibroblastic-ECM interactions in an effort to encourage therapies that will restore cancer-restrictive stromal cues.
Collapse
Affiliation(s)
- Jennifer Alexander
- Fox Chase Cancer Center, Cancer Biology, Temple Health, 333 Cottman Ave, Philadelphia, PA 19111, USA; Drexel University College of Medicine, Department of Molecular Biology and Biochemistry, 245 N 15(th) St, Philadelphia, PA 19102, USA
| | - Edna Cukierman
- Fox Chase Cancer Center, Cancer Biology, Temple Health, 333 Cottman Ave, Philadelphia, PA 19111, USA.
| |
Collapse
|
206
|
Oh SY, Irani S, Kozarek RA. What are the current and potential future roles for endoscopic ultrasound in the treatment of pancreatic cancer? World J Gastrointest Endosc 2016; 8:319-329. [PMID: 27076870 PMCID: PMC4823670 DOI: 10.4253/wjge.v8.i7.319] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 12/15/2015] [Accepted: 02/16/2016] [Indexed: 02/06/2023] Open
Abstract
Pancreatic adenocarcinoma is the fourth leading cause of cancer-related death in the United States. Due to the aggressive tumor biology and late manifestations of the disease, long-term survival is extremely uncommon and the current 5-year survival rate is 7%. Over the last two decades, endoscopic ultrasound (EUS) has evolved from a diagnostic modality to a minimally invasive therapeutic alternative to radiologic procedures and surgery for pancreatic diseases. EUS-guided celiac plexus intervention is a useful adjunct to conventional analgesia for patients with pancreatic cancer. EUS-guided biliary drainage has emerged as a viable option in patients who have failed endoscopic retrograde cholangiopancreatography. Recently, the use of lumen-apposing metal stent to create gastrojejunal anastomosis under EUS and fluoroscopic guidance in patients with malignant gastric outlet obstruction has been reported. On the other hand, anti-tumor therapies delivered by EUS, such as the injection of anti-tumor agents, brachytherapy and ablations are still in the experimental stage without clear survival benefit. In this article, we provide updates on well-established EUS-guided interventions as well as novel techniques relevant to pancreatic cancer.
Collapse
|
207
|
Zhang H, Wu H, Guan J, Wang L, Ren X, Shi X, Liang Z, Liu T. Paracrine SDF-1α signaling mediates the effects of PSCs on GEM chemoresistance through an IL-6 autocrine loop in pancreatic cancer cells. Oncotarget 2016; 6:3085-97. [PMID: 25609203 PMCID: PMC4413639 DOI: 10.18632/oncotarget.3099] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Accepted: 12/25/2014] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer exhibits the poorest prognosis among all tumors and is characterized by high resistance to the currently available chemotherapeutic agents. Our previous studies have suggested that stromal components could promote the chemoresistance of pancreatic cancer cells (PCCs). Here, we explored the roles of pancreatic stellate cells (PSCs) and the SDF-1α/CXCR4 axis in pancreatic cancer chemoresitance. Our results showed that primary PSCs typically expressed SDF-1α, whereas its receptor CXCR4 was highly expressed in PCCs. PSC-conditioned medium (PSC-CM) inhibited Gemcitabine (GEM)-induced cytotoxicity and apoptosis in the human PCC line Panc-1, which was antagonized by an SDF-1α neutralizing Ab. Recombinant human SDF-1α (rhSDF-1α) increased IL-6 expression and secretion in Panc-1 cells in a time and dose-dependent manner, and this effect was suppressed by the CXCR4 antagonist AMD3100. rhSDF-1α protected Panc-1 cells from GEM-induced apoptosis, and the protective effect was significantly reduced by blocking IL-6 using a neutralizing antibody. Moreover, rhSDF-1α increased FAK, ERK1/2, AKT and P38 phosphorylation in Panc-1 cells, and either FAK or ERK1/2 inhibition suppressed SDF-1α-upregulated IL-6 expression. SDF-1α-induced AKT activation was almost completely blocked by FAK inhibition. In conclusion, we demonstrate for the first time that PSCs promote the chemoresistance of PCCs to GEM, and this effect is mediated by paracrine SDF-1α/CXCR4 signaling-induced activation of the intracellular FAK-AKT and ERK1/2 signaling pathways and a subsequent IL-6 autocrine loop in PCCs. Our findings indicate that blocking the PSC-PCC interaction by inhibiting SDF-1α/CXCR4 signaling may be a promising therapeutic strategy for overcoming chemoresistance in pancreatic cancer.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, PR China
| | - Huanwen Wu
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, PR China
| | - Jian Guan
- Department of Pathology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences, Beijing, PR China
| | - Li Wang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, PR China
| | - Xinyu Ren
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, PR China
| | - Xiaohua Shi
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, PR China
| | - Zhiyong Liang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, PR China
| | - Tonghua Liu
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, PR China
| |
Collapse
|
208
|
Toste PA, Nguyen AH, Kadera BE, Duong M, Wu N, Gawlas I, Tran LM, Bikhchandani M, Li L, Patel SG, Dawson DW, Donahue TR. Chemotherapy-Induced Inflammatory Gene Signature and Protumorigenic Phenotype in Pancreatic CAFs via Stress-Associated MAPK. Mol Cancer Res 2016; 14:437-47. [PMID: 26979711 DOI: 10.1158/1541-7786.mcr-15-0348] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 03/01/2016] [Indexed: 12/18/2022]
Abstract
UNLABELLED Pancreatic ductal adenocarcinoma (PDAC) has a characteristically dense stroma comprised predominantly of cancer-associated fibroblasts (CAF). CAFs promote tumor growth, metastasis, and treatment resistance. This study aimed to investigate the molecular changes and functional consequences associated with chemotherapy treatment of PDAC CAFs. Chemoresistant immortalized CAFs (R-CAF) were generated by continuous incubation in gemcitabine. Gene expression differences between treatment-naïve CAFs (N-CAF) and R-CAFs were compared by array analysis. Functionally, tumor cells (TC) were exposed to N-CAF- or R-CAF-conditioned media and assayed for migration, invasion, and viability in vitro Furthermore, a coinjection (TC and CAF) model was used to compare tumor growth in vivo R-CAFs increased TC viability, migration, and invasion compared with N-CAFs. In vivo, TCs coinjected with R-CAFs grew larger than those accompanied by N-CAFs. Genomic analysis demonstrated that R-CAFs had increased expression of various inflammatory mediators, similar to the previously described senescence-associated secretory phenotype (SASP). In addition, SASP mediators were found to be upregulated in response to short duration treatment with gemcitabine in both immortalized and primary CAFs. Inhibition of stress-associated MAPK signaling (P38 MAPK or JNK) attenuated SASP induction as well as the tumor-supportive functions of chemotherapy-treated CAFs in vitro and in vivo These results identify a negative consequence of chemotherapy on the PDAC microenvironment that could be targeted to improve the efficacy of current therapeutic regimens. IMPLICATIONS Chemotherapy treatment of pancreatic cancer-associated fibroblasts results in a proinflammatory response driven by stress-associated MAPK signaling that enhances tumor cell growth and invasiveness. Mol Cancer Res; 14(5); 437-47. ©2016 AACR.
Collapse
Affiliation(s)
- Paul A Toste
- Department of Surgery, Division of General Surgery, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California
| | - Andrew H Nguyen
- Department of Surgery, Division of General Surgery, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California
| | - Brian E Kadera
- Department of Surgery, Division of General Surgery, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California
| | - Mindy Duong
- Department of Surgery, Division of General Surgery, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California
| | - Nanping Wu
- Department of Surgery, Division of General Surgery, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California
| | - Irmina Gawlas
- Department of Surgery, Division of General Surgery, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California
| | - Linh M Tran
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California
| | - Mihir Bikhchandani
- Department of Surgery, Division of General Surgery, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California
| | - Luyi Li
- Department of Surgery, Division of General Surgery, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California
| | - Sanjeet G Patel
- Department of Surgery, Division of General Surgery, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California
| | - David W Dawson
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California. Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California
| | - Timothy R Donahue
- Department of Surgery, Division of General Surgery, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California. Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California. Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California.
| |
Collapse
|
209
|
Duluc C, Moatassim-Billah S, Chalabi-Dchar M, Perraud A, Samain R, Breibach F, Gayral M, Cordelier P, Delisle MB, Bousquet-Dubouch MP, Tomasini R, Schmid H, Mathonnet M, Pyronnet S, Martineau Y, Bousquet C. Pharmacological targeting of the protein synthesis mTOR/4E-BP1 pathway in cancer-associated fibroblasts abrogates pancreatic tumour chemoresistance. EMBO Mol Med 2016; 7:735-53. [PMID: 25834145 PMCID: PMC4459815 DOI: 10.15252/emmm.201404346] [Citation(s) in RCA: 159] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is extremely stroma-rich. Cancer-associated fibroblasts (CAFs) secrete proteins that activate survival and promote chemoresistance of cancer cells. Our results demonstrate that CAF secretome-triggered chemoresistance is abolished upon inhibition of the protein synthesis mTOR/4E-BP1 regulatory pathway which we found highly activated in primary cultures of α-SMA-positive CAFs, isolated from human PDAC resections. CAFs selectively express the sst1 somatostatin receptor. The SOM230 analogue (Pasireotide) activates the sst1 receptor and inhibits the mTOR/4E-BP1 pathway and the resultant synthesis of secreted proteins including IL-6. Consequently, tumour growth and chemoresistance in nude mice xenografted with pancreatic cancer cells and CAFs, or with pieces of resected human PDACs, are reduced when chemotherapy (gemcitabine) is combined with SOM230 treatment. While gemcitabine alone has marginal effects, SOM230 is permissive to gemcitabine-induced cancer cell apoptosis and acts as an antifibrotic agent. We propose that selective inhibition of CAF protein synthesis with sst1-directed pharmacological compounds represents an anti-stromal-targeted therapy with promising chemosensitization potential.
Collapse
Affiliation(s)
- Camille Duluc
- INSERM UMR-1037, Cancer Research Center of Toulouse (CRCT), Equipe labellisée Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer (TOUCAN), Université de Toulouse, Toulouse, France
| | - Siham Moatassim-Billah
- INSERM UMR-1037, Cancer Research Center of Toulouse (CRCT), Equipe labellisée Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer (TOUCAN), Université de Toulouse, Toulouse, France Biochemistry-Immunology Laboratory, Faculty of Sciences Rabat, University Mohammed V - Agdal, Agdal, Morocco
| | - Mounira Chalabi-Dchar
- INSERM UMR-1037, Cancer Research Center of Toulouse (CRCT), Equipe labellisée Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer (TOUCAN), Université de Toulouse, Toulouse, France
| | - Aurélie Perraud
- EA 3842 Laboratory, Medicine and Pharmacy Faculties, Limoges University, Limoges, France
| | - Rémi Samain
- INSERM UMR-1037, Cancer Research Center of Toulouse (CRCT), Equipe labellisée Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer (TOUCAN), Université de Toulouse, Toulouse, France
| | | | - Marion Gayral
- INSERM UMR-1037, Cancer Research Center of Toulouse (CRCT), Equipe labellisée Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer (TOUCAN), Université de Toulouse, Toulouse, France
| | - Pierre Cordelier
- INSERM UMR-1037, Cancer Research Center of Toulouse (CRCT), Equipe labellisée Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer (TOUCAN), Université de Toulouse, Toulouse, France
| | | | - Marie-Pierre Bousquet-Dubouch
- CNRS UMR-5089, Institut de Pharmacologie et de Biologie structurale (IPBS), Université de Toulouse, Toulouse, France
| | - Richard Tomasini
- CRCM, INSERM, U1068; Paoli-Calmettes Institute; Aix-Marseille University, UM105; CNRS, UMR7258, Marseille, France
| | | | - Muriel Mathonnet
- EA 3842 Laboratory, Medicine and Pharmacy Faculties, Limoges University, Limoges, France
| | - Stéphane Pyronnet
- INSERM UMR-1037, Cancer Research Center of Toulouse (CRCT), Equipe labellisée Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer (TOUCAN), Université de Toulouse, Toulouse, France
| | - Yvan Martineau
- INSERM UMR-1037, Cancer Research Center of Toulouse (CRCT), Equipe labellisée Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer (TOUCAN), Université de Toulouse, Toulouse, France
| | - Corinne Bousquet
- INSERM UMR-1037, Cancer Research Center of Toulouse (CRCT), Equipe labellisée Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer (TOUCAN), Université de Toulouse, Toulouse, France
| |
Collapse
|
210
|
Li X, Roife D, Kang Y, Dai B, Pratt M, Fleming JB. Extracellular lumican augments cytotoxicity of chemotherapy in pancreatic ductal adenocarcinoma cells via autophagy inhibition. Oncogene 2016; 35:4881-90. [PMID: 26876211 DOI: 10.1038/onc.2016.20] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 10/27/2015] [Accepted: 12/04/2015] [Indexed: 12/17/2022]
Abstract
Lumican, an extracellular matrix proteoglycan overexpressed by pancreatic stellate cells (PSCs) and pancreatic ductal adenocarcinoma cells (PDACs), drives the formation of a tumor-specific microenvironment. We recently showed that extracellular lumican inhibits pancreatic cancer cell growth and is associated with prolonged survival after surgery. Here we investigated the role of extracellular lumican in chemotherapy-mediated cancer therapy. Lumican secretion was increased by chemotherapeutic agents in PDAC, and especially in PSCs, and appeared to be linked to the extent of cells' response to chemotherapy-induced growth inhibition. In multiple PDAC models, including cell lines, patient-derived xenografts and lumican knockout mice, lumican significantly increased antitumor effect of chemotherapy. This effect was associated with DNA damage, apoptosis and inhibition of cell viability, glucose consumption, lactate production and vascular endothelial growth factor secretion. In PDAC cells, chemotherapeutic agents triggered autophagosome formation and increased LC3 expression through the reactive oxygen species-mediated AMP-activated kinase (AMPK) signaling pathway. Inhibition of gemcitabine-induced autophagy in cancer cells by treatment with AMPK inhibitor compound C, lysosomal inhibitor chloroquine or autophagy inhibitor 3MA enhanced gemcitabine-induced apoptosis, suggesting that autophagy is a protective cellular response to gemcitabine treatment. Importantly, lumican dramatically decreased AMPK activity, inhibiting chemotherapy-induced autophagy in both in vitro and in vivo PDAC models. Co-treatment of PDAC cells with lumican and gemcitabine increased mitochondrial damage, reactive oxygen species (ROS) production and cytochrome c release, indicating that lumican-induced disruption of mitochondrial function may be the mechanism of sensitization to gemcitabine. Together, our findings demonstrate that extracellular lumican augments cytotoxicity of chemotherapy in PDAC cells through inhibition of chemotherapeutic agent-induced autophagy.
Collapse
Affiliation(s)
- X Li
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - D Roife
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Y Kang
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - B Dai
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - M Pratt
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - J B Fleming
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
211
|
Suppression of CD51 in pancreatic stellate cells inhibits tumor growth by reducing stroma and altering tumor-stromal interaction in pancreatic cancer. Int J Oncol 2016; 48:1499-508. [DOI: 10.3892/ijo.2016.3374] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 01/05/2016] [Indexed: 11/05/2022] Open
|
212
|
Pothula SP, Xu Z, Goldstein D, Biankin AV, Pirola RC, Wilson JS, Apte MV. Hepatocyte growth factor inhibition: a novel therapeutic approach in pancreatic cancer. Br J Cancer 2016; 114:269-80. [PMID: 26766740 PMCID: PMC4742591 DOI: 10.1038/bjc.2015.478] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 12/04/2015] [Accepted: 12/16/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Pancreatic stellate cells (PSCs, which produce the stroma of pancreatic cancer (PC)) interact with cancer cells to facilitate PC growth. A candidate growth factor pathway that may mediate this interaction is the HGF-c-MET pathway. METHODS Effects of HGF inhibition (using a neutralising antibody AMG102) alone or in combination with gemcitabine were assessed (i) in vivo using an orthotopic model of PC, and (ii) in vitro using cultured PC cells (AsPC-1) and human PSCs. RESULTS We have shown that human PSCs (hPSCs) secrete HGF but do not express the receptor c-MET, which is present predominantly on cancer cells. HGF inhibition was as effective as standard chemotherapy in inhibiting local tumour growth but was significantly more effective than gemcitabine in reducing tumour angiogenesis and metastasis. HGF inhibition has resulted in reduced metastasis; however, interestingly this antimetastatic effect was lost when combined with gemcitabine. This suggests that gemcitabine treatment selects out a subpopulation of cancer cells with increased epithelial-mesenchymal transition (EMT) and stem-cell characteristics, as supported by our findings of increased expression of EMT and stem-cell markers in tumour sections from our animal model. In vitro studies showed that hPSC secretions induced proliferation and migration, but inhibited apoptosis, of cancer cells. These effects were countered by pretreatment of hPSC secretions with a HGF-neutralising antibody but not by gemcitabine, indicating a key role for HGF in PSC-PC interactions. CONCLUSIONS Our studies suggest that targeted therapy to inhibit stromal-tumour interactions mediated by the HGF-c-MET pathway may represent a novel therapeutic approach in PC that will require careful modelling for optimal integration with existing treatment modalities.
Collapse
Affiliation(s)
- Srinivasa P Pothula
- Pancreatic Research Group, South Western Sydney Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
| | - Zhihong Xu
- Pancreatic Research Group, South Western Sydney Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
| | - David Goldstein
- Department of Medical Oncology, Prince of Wales Hospital, Sydney, New South Wales, Australia
| | - Andrew V Biankin
- Cancer Research Division, The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Romano C Pirola
- Pancreatic Research Group, South Western Sydney Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
| | - Jeremy S Wilson
- Pancreatic Research Group, South Western Sydney Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
| | - Minoti V Apte
- Pancreatic Research Group, South Western Sydney Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
| |
Collapse
|
213
|
Tang D, Gao J, Wang S, Ye N, Chong Y, Huang Y, Wang J, Li B, Yin W, Wang D. Cancer-associated fibroblasts promote angiogenesis in gastric cancer through galectin-1 expression. Tumour Biol 2016; 37:1889-99. [PMID: 26323258 DOI: 10.1007/s13277-015-3942-9] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 08/17/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Galectin-1, an evolutionarily conserved glycan-binding protein with angiogenic potential, was recently identified as being overexpressed in cancer-associated fibroblasts (CAFs) of gastric cancer. The role of endogenous CAF-derived galectin-1 on angiogenesis in gastric cancer and the mechanism involved remain unknown. METHODS Immunohistochemical staining was used to investigate the correlation between galectin-1 and vascular endothelial growth factor (VEGF) and CD31 expression in gastric cancer tissues and normal gastric tissues. Galectin-1 was knocked down in CAFs isolated from gastric cancer using small interfering ribonucleic acid (RNA), or overexpressed using recombinant lentiviruses, and the CAFs were co-cultured with human umbilical vein endothelial cells (HUVECs) or cancer cells. Subsequently, proliferation, migration, tube formation, and VEGF/VEGF receptor (VEGFR) 2 expression were detected. The role of CAF-derived galectin-1 in tumor angiogenesis in vivo was studied using the chick chorioallantoic membrane (CAM) assay. RESULTS Galectin-1 was highly expressed in the CAFs and was positively associated with VEGF and CD31 expression. In the co-culture, high expression of galectin-1 in the CAFs increased HUVEC proliferation, migration, tube formation, and VEGFR2 phosphorylation and enhanced VEGF expression in gastric cancer cells. The CAM assay indicated that high expression of galectin-1 in the CAFs accelerated tumor growth and promoted angiogenesis. In contrast, galectin-1 knockdown in the CAFs significantly inhibited this effect. CONCLUSION CAF-derived galectin-1 significantly promotes angiogenesis in gastric cancer and may be a target for angiostatic therapy.
Collapse
Affiliation(s)
- Dong Tang
- Department of Gastrointestinal Surgery, Clinical Medical College of Yangzhou University (Subei People's Hospital of Jiangsu Province), Yangzhou, 225001, People's Republic of China
| | - Jun Gao
- Department of Gastrointestinal Surgery, Clinical Medical College of Yangzhou University (Subei People's Hospital of Jiangsu Province), Yangzhou, 225001, People's Republic of China
| | - Sen Wang
- College of Clinical Medicine, Nanjing Medical University (the First Affiliated Hospital of Nanjing Medical University), Nanjing, 211166, People's Republic of China
| | - Nianyuan Ye
- Department of Gastrointestinal Surgery, Clinical Medical College of Yangzhou University (Subei People's Hospital of Jiangsu Province), Yangzhou, 225001, People's Republic of China
| | - Yang Chong
- Department of Gastrointestinal Surgery, Clinical Medical College of Yangzhou University (Subei People's Hospital of Jiangsu Province), Yangzhou, 225001, People's Republic of China
| | - Yuqin Huang
- Department of Gastrointestinal Surgery, Clinical Medical College of Yangzhou University (Subei People's Hospital of Jiangsu Province), Yangzhou, 225001, People's Republic of China
| | - Jie Wang
- Department of Gastrointestinal Surgery, Clinical Medical College of Yangzhou University (Subei People's Hospital of Jiangsu Province), Yangzhou, 225001, People's Republic of China
| | - Bin Li
- Department of Gastrointestinal Surgery, Clinical Medical College of Yangzhou University (Subei People's Hospital of Jiangsu Province), Yangzhou, 225001, People's Republic of China
| | - Wei Yin
- Department of Gastrointestinal Surgery, Clinical Medical College of Yangzhou University (Subei People's Hospital of Jiangsu Province), Yangzhou, 225001, People's Republic of China
| | - Daorong Wang
- Department of Gastrointestinal Surgery, Clinical Medical College of Yangzhou University (Subei People's Hospital of Jiangsu Province), Yangzhou, 225001, People's Republic of China.
| |
Collapse
|
214
|
Hah N, Sherman MH, Yu RT, Downes M, Evans RM. Targeting Transcriptional and Epigenetic Reprogramming in Stromal Cells in Fibrosis and Cancer. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2016; 80:249-55. [PMID: 26801159 DOI: 10.1101/sqb.2015.80.027185] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The basis of many human diseases arises from both genetic and epigenetic regulation. Recent advances in the understanding of the mechanisms underlying transcriptional and epigenetic regulation and their prevalence as contributors to a diverse range of human diseases have led us to focus on transcription and epigenetic changes in a variety of human disease conditions. Specifically, our recent studies in liver fibrosis and pancreatic cancer have demonstrated that the epigenetic regulation in hepatic stellate cells (HSCs) and pancreatic stellate cells (PSCs) significantly contributes to the progress in such diseases and presents great therapeutic potential. We show that the vitamin D receptor (VDR) acts as a master genomic suppressor in both HSC and PSC activation. The studies also have demonstrated that the VDR ligand reduces fibrosis and inflammation in a murine liver fibrosis and pancreatitis model. Although our current studies focus on characterizing the roles of VDR and regulatory regions within gene promoters and regulatory enhancers, we have expanded our effort to epigenetic mechanisms as major determinants of gene activation and repression in order to develop potential therapeutics to modulate stroma-associated pathologies including inflammation, fibrosis, and cancer.
Collapse
Affiliation(s)
- Nasun Hah
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037
| | - Mara H Sherman
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037
| | - Ruth T Yu
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037
| | - Michael Downes
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037
| | - Ronald M Evans
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037 Howard Hughes Medical Institute, The Salk Institute for Biological Studies, La Jolla, California 92037
| |
Collapse
|
215
|
Liang D, Shi S, Xu J, Zhang B, Qin Y, Ji S, Xu W, Liu J, Liu L, Liu C, Long J, Ni Q, Yu X. New insights into perineural invasion of pancreatic cancer: More than pain. Biochim Biophys Acta Rev Cancer 2016; 1865:111-22. [PMID: 26794395 DOI: 10.1016/j.bbcan.2016.01.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 12/26/2015] [Accepted: 01/11/2016] [Indexed: 01/01/2023]
Abstract
Pancreatic cancer is one of the most malignant human tumors. Perineural invasion, whereby a cancer cell invades the perineural spaces surrounding nerves, is acknowledged as a gradual contributor to cancer aggressiveness. Furthermore, perineural invasion is considered one of the root causes of the recurrence and metastasis observed after pancreatic resection, and it is also an independent predictor of prognosis. Advanced research has demonstrated that the neural microenvironment is closely associated with perineural invasion in pancreatic cancer. Therapy targeting the molecular mechanism of perineural invasion may enable the durable clinical treatment of this formidable disease. This review provides an overview of the present status of perineural invasion, the relevant molecular mechanisms of perineural invasion, pain and hyperglycemia associated with perineural invasion in pancreatic cancer, and the targeted therapeutics based on these studies.
Collapse
Affiliation(s)
- Dingkong Liang
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Si Shi
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jin Xu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Bo Zhang
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Yi Qin
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Shunrong Ji
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Wenyan Xu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jiang Liu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Liang Liu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Chen Liu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jiang Long
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Quanxing Ni
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xianjun Yu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| |
Collapse
|
216
|
Abstract
OBJECTIVES Pancreatic cancer (PaC) frequently results in death despite surgical resection. We sought to evaluate whether inflammation in the primary tumor was associated with early death after surgical resection. METHODS In this case-control study, we identified 21 individuals who died less than 12 months after surgery for PaC and 42 controls who survived more than 36 months after surgery. Differences in the composition of host inflammatory response between the groups were evaluated with univariate comparisons and odds ratios for early death were calculated using logistic regression modeling. RESULTS Cases were more likely to have a high tumor grade (90.5% vs 26.2%; P < 0.01). The odds of early death were increased in those with a high-grade tumor (unadjusted odds ratio, 26.77; 95% confidence interval, 5.35-134.07; P < 0.01). Conversely, the density (high vs low) of inflammatory cells in tumors was similar between the groups, and odds of early death were not associated with any inflammatory marker. CONCLUSIONS High tumor grade, but not altered density of inflammatory cells in the intratumoral compartment, is associated with increased odds of early death after PaC resection. Future studies evaluating the host response in multiple tumor compartments with advanced histologic techniques is needed to further elucidate the role of inflammation in PaC.
Collapse
|
217
|
Mettu NB, Abbruzzese JL. Clinical Insights Into the Biology and Treatment of Pancreatic Cancer. J Oncol Pract 2016; 12:17-23. [DOI: 10.1200/jop.2015.009092] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Pancreatic cancer is a devastating disease with a universally poor prognosis. In 2015, it is estimated that there will be 48,960 new cases of pancreatic cancer and that 40,560 people will die of the disease. The 5-year survival rate is 7.2% for all patients with pancreatic cancer; however, survival depends greatly on the stage at diagnosis. Unfortunately, 53% of patients already have metastatic disease at diagnosis, which corresponds to a 5-year survival rate of 2.4%. Even for the 9% of patients with localized disease confined to the pancreas, the 5-year survival is still modest at only 27.1%. These grim statistics highlight the need for ways to identify cohorts of individuals at highest risk, methods to screen those at highest risk to identify preinvasive pathologic precursors, and development of effective systemic therapies. Recent clinical and translational progress has emphasized the relationship with diabetes, the role of the stroma, and the interplay of each of these with inflammation in the pathobiology of pancreatic cancer. In this article, we will discuss these relationships and how they might translate into novel management strategies for the treatment of this disease.
Collapse
|
218
|
Strobel O, Dadabaeva N, Felix K, Hackert T, Giese NA, Jesenofsky R, Werner J. Isolation and culture of primary human pancreatic stellate cells that reflect the context of their tissue of origin. Langenbecks Arch Surg 2015; 401:89-97. [PMID: 26712717 DOI: 10.1007/s00423-015-1343-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 09/18/2015] [Indexed: 02/02/2023]
Abstract
BACKGROUND Pancreatic stellate cells (PSCs) play a critical role in pancreatic ductal adenocarcinoma (PDAC). Activated PSCs are the main source of fibrosis in chronic pancreatitis and of desmoplasia in PDAC. The majority of studies on PSC are based on in vitro experiments relying on immortalized cell lines derived from diseased human pancreas or from animal models. These PSCs are usually activated and may not represent the biological context of their tissue of origin. PURPOSE (1) To isolate and culture primary human PSC from different disease contexts with minimal impact on their state of activation. (2) To perform a comparative analysis of phenotypes of PSC derived from different contexts. METHODS PSCs were isolated from normal pancreas, chronic pancreatitis, and PDAC using a hybrid method of digestion and outgrowth. To minimize activation by serum compounds, cells were cultured in a low-serum environment (2.5 % fetal bovine serum (FBS)). Expression patterns of commonly used markers for PSC phenotype and activity were compared between primary PSC lines derived from different contexts and correlated to expression in their original tissues. RESULTS Isolation was successful from 14 of 17 tissues (82 %). Isolated PSC displayed stable viability and phenotype in low-serum environment. Expression profiles of isolated PSC and matched original tissues were closely correlated. PDAC-derived PSC tended to have a higher status of activation if compared to PSC derived from non-cancerous tissues. CONCLUSIONS Primary human PSCs isolated from different contexts and cultured in a low-serum environment maintain a phenotype that reflects the stromal activity present in their tissue of origin.
Collapse
Affiliation(s)
- Oliver Strobel
- Department of General Surgery, University Hospital Heidelberg, Im Neuenheimer Feld 110, 69120, Heidelberg, Germany.
| | - Nigora Dadabaeva
- Department of General Surgery, University Hospital Heidelberg, Im Neuenheimer Feld 110, 69120, Heidelberg, Germany
| | - Klaus Felix
- Department of General Surgery, University Hospital Heidelberg, Im Neuenheimer Feld 110, 69120, Heidelberg, Germany
| | - Thilo Hackert
- Department of General Surgery, University Hospital Heidelberg, Im Neuenheimer Feld 110, 69120, Heidelberg, Germany
| | - Nathalia A Giese
- Department of General Surgery, University Hospital Heidelberg, Im Neuenheimer Feld 110, 69120, Heidelberg, Germany
| | - Ralf Jesenofsky
- Department of Internal Medicine 2, University Medicine Mannheim, Mannheim, Germany
| | - Jens Werner
- Department of General Surgery, University Hospital Heidelberg, Im Neuenheimer Feld 110, 69120, Heidelberg, Germany
| |
Collapse
|
219
|
Topalovski M, Brekken RA. Matrix control of pancreatic cancer: New insights into fibronectin signaling. Cancer Lett 2015; 381:252-8. [PMID: 26742464 DOI: 10.1016/j.canlet.2015.12.027] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 12/18/2015] [Accepted: 12/19/2015] [Indexed: 12/18/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDA) is a highly metastatic disease that resists most current therapies. A defining characteristic of PDA is an intense fibrotic response that promotes tumor cell invasion and chemoresistance. Efforts to understand the complex relationship between the tumor and its extracellular network and to therapeutically perturb tumor-stroma interactions are ongoing. Fibronectin (FN), a provisional matrix protein abundant in PDA stroma but not normal tissues, supports metastatic spread and chemoresistance of this deadly disease. FN also supports angiogenesis, which is required for even hypovascular tumors such as PDA to develop and progress. Targeting components of the tumor stroma, such as FN, can effectively reduce tumor growth and spread while also enhancing delivery of chemotherapy. Here, we review the molecular mechanisms by which FN drives angiogenesis, metastasis and chemoresistance in PDA. In light of these new findings, we also discuss therapeutic strategies to inhibit FN signaling.
Collapse
Affiliation(s)
- Mary Topalovski
- Hamon Center for Therapeutic Oncology Research and the Division of Surgical Oncology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rolf A Brekken
- Hamon Center for Therapeutic Oncology Research and the Division of Surgical Oncology, UT Southwestern Medical Center, Dallas, TX 75390, USA; Departments of Surgery and Pharmacology, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
220
|
Pothula SP, Xu Z, Goldstein D, Pirola RC, Wilson JS, Apte MV. Key role of pancreatic stellate cells in pancreatic cancer. Cancer Lett 2015; 381:194-200. [PMID: 26571462 DOI: 10.1016/j.canlet.2015.10.035] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 10/29/2015] [Accepted: 10/29/2015] [Indexed: 02/07/2023]
Abstract
Pancreatic stellate cells (PSCs) are responsible for producing the collagenous stroma in pancreatic cancer. Findings from the majority of in vitro and in vivo studies to date indicate that PSCs interact with cancer cells as well as with other cellular elements in the stroma including immune cells, endothelial cells and neuronal cells to set up a growth permissive microenvironment for pancreatic tumours. However, two recent studies reporting a protective effect of myofibroblasts in pancreatic cancer have served to remind researchers of the possibility that the role of PSCs in this disease may be context and time-dependent, such that any possible early protective role of PSCs is subverted in later stages by the ability of cancer cells to turn PSCs into cancer-promoting aides. This concept is supported by the development in recent years of several novel therapeutic approaches targeting the stroma that have been successfully applied in pre-clinical settings to inhibit disease progression. A multi-pronged approach aimed at tumour cells as well as stromal elements may be the key to achieving better clinical outcomes in patients with pancreatic cancer.
Collapse
Affiliation(s)
- Srinivasa P Pothula
- Pancreatic Research Group, South Western Sydney Clinical School, Faculty of Medicine, The University of New South Wales, Sydney, Australia; Ingham Institute for Applied Medical Research, Liverpool, NSW, Australia
| | - Zhihong Xu
- Pancreatic Research Group, South Western Sydney Clinical School, Faculty of Medicine, The University of New South Wales, Sydney, Australia; Ingham Institute for Applied Medical Research, Liverpool, NSW, Australia
| | - David Goldstein
- Pancreatic Research Group, South Western Sydney Clinical School, Faculty of Medicine, The University of New South Wales, Sydney, Australia
| | - Romano C Pirola
- Pancreatic Research Group, South Western Sydney Clinical School, Faculty of Medicine, The University of New South Wales, Sydney, Australia; Ingham Institute for Applied Medical Research, Liverpool, NSW, Australia
| | - Jeremy S Wilson
- Pancreatic Research Group, South Western Sydney Clinical School, Faculty of Medicine, The University of New South Wales, Sydney, Australia; Ingham Institute for Applied Medical Research, Liverpool, NSW, Australia
| | - Minoti V Apte
- Pancreatic Research Group, South Western Sydney Clinical School, Faculty of Medicine, The University of New South Wales, Sydney, Australia; Ingham Institute for Applied Medical Research, Liverpool, NSW, Australia.
| |
Collapse
|
221
|
Han S, Delitto D, Zhang D, Sorenson HL, Sarosi GA, Thomas RM, Behrns KE, Wallet SM, Trevino JG, Hughes SJ. Primary outgrowth cultures are a reliable source of human pancreatic stellate cells. J Transl Med 2015; 95:1331-1340. [PMID: 26322418 DOI: 10.1038/labinvest.2015.117] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 07/20/2015] [Indexed: 01/18/2023] Open
Abstract
Recent advances demonstrate a critical yet poorly understood role for the pancreatic stellate cell (PSC) in the pathogenesis of chronic pancreatitis (CP) and pancreatic cancer (PC). Progress in this area has been hampered by the availability, fidelity, and/or reliability of in vitro models of PSCs. We examined whether outgrowth cultures from human surgical specimens exhibited reproducible phenotypic and functional characteristics of PSCs. PSCs were cultured from surgical specimens of healthy pancreas, CP and PC. Growth dynamics, phenotypic characteristics, soluble mediator secretion profiles and co-culture with PC cells both in vitro and in vivo were assessed. Forty-seven primary cultures were established from 52 attempts, demonstrating universal α-smooth muscle actin and glial fibrillary acidic protein but negligible epithelial surface antigen expression. Modification of culture conditions consistently led to cytoplasmic lipid accumulation, suggesting induction of a quiescent phenotype. Secretion of growth factors, chemokines and cytokines did not significantly differ between donor pathologies, but did evolve over time in culture. Co-culture of PSCs with established PC cell lines resulted in significant changes in levels of multiple secreted mediators. Primary PSCs co-inoculated with PC cells in a xenograft model led to augmented tumor growth and metastasis. Therefore, regardless of donor pathology, outgrowth cultures produce PSCs that demonstrate consistent growth and protein secretion properties. Primary cultures from pancreatic surgical specimens, including malignancies, may represent a reliable source of human PSCs.
Collapse
Affiliation(s)
- Song Han
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Gainesville, FL, USA
| | - Daniel Delitto
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Gainesville, FL, USA
| | - Dongyu Zhang
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Gainesville, FL, USA
| | - Heather L Sorenson
- Department of Periodontology and Oral Biology, College of Dentistry, University of Florida Health Science Center, Gainesville, FL, USA
| | - George A Sarosi
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Gainesville, FL, USA
- Department of Surgery, North Florida/South Georgia Veterans Health System, University of Florida College of Medicine, Gainesville, FL, USA
| | - Ryan M Thomas
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Gainesville, FL, USA
- Department of Surgery, North Florida/South Georgia Veterans Health System, University of Florida College of Medicine, Gainesville, FL, USA
| | - Kevin E Behrns
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Gainesville, FL, USA
| | - Shannon M Wallet
- Department of Periodontology and Oral Biology, College of Dentistry, University of Florida Health Science Center, Gainesville, FL, USA
| | - Jose G Trevino
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Gainesville, FL, USA
| | - Steven J Hughes
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Gainesville, FL, USA
| |
Collapse
|
222
|
Delitto D, Black BS, Sorenson HL, Knowlton AE, Thomas RM, Sarosi GA, Moldawer LL, Behrns KE, Liu C, George TJ, Trevino JG, Wallet SM, Hughes SJ. The inflammatory milieu within the pancreatic cancer microenvironment correlates with clinicopathologic parameters, chemoresistance and survival. BMC Cancer 2015; 15:783. [PMID: 26498838 PMCID: PMC4619553 DOI: 10.1186/s12885-015-1820-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 10/16/2015] [Indexed: 01/05/2023] Open
Abstract
Background The tumor microenvironment impacts pancreatic cancer (PC) development, progression and metastasis. How intratumoral inflammatory mediators modulate this biology remains poorly understood. We hypothesized that the inflammatory milieu within the PC microenvironment would correlate with clinicopathologic findings and survival. Methods Pancreatic specimens from normal pancreas (n = 6), chronic pancreatitis (n = 9) and pancreatic adenocarcinoma (n = 36) were homogenized immediately upon resection. Homogenates were subjected to multiplex analysis of 41 inflammatory mediators. Results Twenty-three mediators were significantly elevated in adenocarcinoma specimens compared to nonmalignant controls. Increased intratumoral IL-8 concentrations associated with larger tumors (P = .045) and poor differentiation (P = .038); the administration of neoadjuvant chemotherapy associated with reduced IL-8 concentrations (P = .003). Neoadjuvant therapy was also associated with elevated concentrations of Flt-3 L (P = .005). Elevated levels of pro-inflammatory cytokines IL-1β (P = .017) and TNFα (P = .033) were associated with a poor histopathologic response to neoadjuvant therapy. Elevated concentrations of G-CSF (P = .016) and PDGF-AA (P = .012) correlated with reduced overall survival. Conversely, elevated concentrations of FGF-2 (P = .038), TNFα (P = .031) and MIP-1α (P = .036) were associated with prolonged survival. Conclusion The pancreatic cancer microenvironment harbors a unique inflammatory milieu with potential diagnostic and prognostic value.
Collapse
Affiliation(s)
- Daniel Delitto
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Room 6116, Shands Hospital, 1600 SW Archer Rd, Gainesville, FL, 32610, USA.
| | - Brian S Black
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Room 6116, Shands Hospital, 1600 SW Archer Rd, Gainesville, FL, 32610, USA.
| | - Heather L Sorenson
- Department of Oral Biology, College of Dentistry, University of Florida Health Science Center, Gainesville, FL, 32610, USA.
| | - Andrea E Knowlton
- Department of Oral Biology, College of Dentistry, University of Florida Health Science Center, Gainesville, FL, 32610, USA.
| | - Ryan M Thomas
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Room 6116, Shands Hospital, 1600 SW Archer Rd, Gainesville, FL, 32610, USA. .,North Florida/South Georgia Veterans Health System, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, 32610, USA.
| | - George A Sarosi
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Room 6116, Shands Hospital, 1600 SW Archer Rd, Gainesville, FL, 32610, USA. .,North Florida/South Georgia Veterans Health System, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, 32610, USA.
| | - Lyle L Moldawer
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Room 6116, Shands Hospital, 1600 SW Archer Rd, Gainesville, FL, 32610, USA.
| | - Kevin E Behrns
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Room 6116, Shands Hospital, 1600 SW Archer Rd, Gainesville, FL, 32610, USA.
| | - Chen Liu
- Department of Pathology, College of Medicine, University of Florida Health Science Center, Gainesville, FL, 32610, USA.
| | - Thomas J George
- Department of Medicine, College of Medicine, University of Florida Health Science Center, Gainesville, FL, 32610, USA.
| | - Jose G Trevino
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Room 6116, Shands Hospital, 1600 SW Archer Rd, Gainesville, FL, 32610, USA.
| | - Shannon M Wallet
- Department of Oral Biology, College of Dentistry, University of Florida Health Science Center, Gainesville, FL, 32610, USA.
| | - Steven J Hughes
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Room 6116, Shands Hospital, 1600 SW Archer Rd, Gainesville, FL, 32610, USA.
| |
Collapse
|
223
|
Zambirinis CP, Levie E, Nguy S, Avanzi A, Barilla R, Xu Y, Seifert L, Daley D, Greco SH, Deutsch M, Jonnadula S, Torres-Hernandez A, Tippens D, Pushalkar S, Eisenthal A, Saxena D, Ahn J, Hajdu C, Engle DD, Tuveson D, Miller G. TLR9 ligation in pancreatic stellate cells promotes tumorigenesis. J Exp Med 2015; 212:2077-94. [PMID: 26481685 PMCID: PMC4647258 DOI: 10.1084/jem.20142162] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 09/15/2015] [Indexed: 12/15/2022] Open
Abstract
Zambirinis et al. show that TLR9 stimulation has a protumorigenic effect in pancreatic carcinoma by inducing pancreatic stellate cells to become fibrogenic and produce chemokines that stimulate epithelial cell proliferation. Activation of TLR9 results also in an immune suppressive tumor microenvironment via recruitment of regulatory T cells and induction of myeloid-derived suppressor cell proliferation. Modulation of Toll-like receptor (TLR) signaling can have protective or protumorigenic effects on oncogenesis depending on the cancer subtype and on specific inflammatory elements within the tumor milieu. We found that TLR9 is widely expressed early during the course of pancreatic transformation and that TLR9 ligands are ubiquitous within the tumor microenvironment. TLR9 ligation markedly accelerates oncogenesis, whereas TLR9 deletion is protective. We show that TLR9 activation has distinct effects on the epithelial, inflammatory, and fibrogenic cellular subsets in pancreatic carcinoma and plays a central role in cross talk between these compartments. Specifically, TLR9 activation can induce proinflammatory signaling in transformed epithelial cells, but does not elicit oncogene expression or cancer cell proliferation. Conversely, TLR9 ligation induces pancreatic stellate cells (PSCs) to become fibrogenic and secrete chemokines that promote epithelial cell proliferation. TLR9-activated PSCs mediate their protumorigenic effects on the epithelial compartment via CCL11. Additionally, TLR9 has immune-suppressive effects in the tumor microenvironment (TME) via induction of regulatory T cell recruitment and myeloid-derived suppressor cell proliferation. Collectively, our work shows that TLR9 has protumorigenic effects in pancreatic carcinoma which are distinct from its influence in extrapancreatic malignancies and from the mechanistic effects of other TLRs on pancreatic oncogenesis.
Collapse
Affiliation(s)
| | - Elliot Levie
- Department of Surgery, New York University School of Medicine, New York, NY 10016
| | - Susanna Nguy
- Department of Surgery, New York University School of Medicine, New York, NY 10016
| | - Antonina Avanzi
- Department of Surgery, New York University School of Medicine, New York, NY 10016
| | - Rocky Barilla
- Department of Surgery, New York University School of Medicine, New York, NY 10016
| | - Yijie Xu
- Department of Surgery, New York University School of Medicine, New York, NY 10016
| | - Lena Seifert
- Department of Surgery, New York University School of Medicine, New York, NY 10016
| | - Donnele Daley
- Department of Surgery, New York University School of Medicine, New York, NY 10016
| | - Stephanie H Greco
- Department of Surgery, New York University School of Medicine, New York, NY 10016
| | - Michael Deutsch
- Department of Surgery, New York University School of Medicine, New York, NY 10016
| | - Saikiran Jonnadula
- Department of Surgery, New York University School of Medicine, New York, NY 10016
| | | | - Daniel Tippens
- Department of Surgery, New York University School of Medicine, New York, NY 10016
| | | | - Andrew Eisenthal
- Department of Surgery, New York University School of Medicine, New York, NY 10016
| | - Deepak Saxena
- New York University College of Dentistry, New York, NY 10016
| | - Jiyoung Ahn
- Department of Population Health, New York University School of Medicine, New York, NY 10016
| | - Cristina Hajdu
- Department of Pathology, New York University School of Medicine, New York, NY 10016
| | | | - David Tuveson
- Cold Spring Harbor Laboratories, Cold Spring Harbor, NY 11724
| | - George Miller
- Department of Surgery, New York University School of Medicine, New York, NY 10016 Department of Cell Biology, New York University School of Medicine, New York, NY 10016
| |
Collapse
|
224
|
Gopinathan A, Morton JP, Jodrell DI, Sansom OJ. GEMMs as preclinical models for testing pancreatic cancer therapies. Dis Model Mech 2015; 8:1185-200. [PMID: 26438692 PMCID: PMC4610236 DOI: 10.1242/dmm.021055] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Pancreatic ductal adenocarcinoma is the most common form of pancreatic tumour, with a very limited survival rate and currently no available disease-modifying treatments. Despite recent advances in the production of genetically engineered mouse models (GEMMs), the development of new therapies for pancreatic cancer is still hampered by a lack of reliable and predictive preclinical animal models for this disease. Preclinical models are vitally important for assessing therapies in the first stages of the drug development pipeline, prior to their transition to the clinical arena. GEMMs carry mutations in genes that are associated with specific human diseases and they can thus accurately mimic the genetic, phenotypic and physiological aspects of human pathologies. Here, we discuss different GEMMs of human pancreatic cancer, with a focus on the Lox-Stop-Lox (LSL)-Kras(G12D); LSL-Trp53(R172H); Pdx1-cre (KPC) model, one of the most widely used preclinical models for this disease. We describe its application in preclinical research, highlighting its advantages and disadvantages, its potential for predicting clinical outcomes in humans and the factors that can affect such outcomes, and, finally, future developments that could advance the discovery of new therapies for pancreatic cancer.
Collapse
Affiliation(s)
- Aarthi Gopinathan
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | | | - Duncan I Jodrell
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - Owen J Sansom
- Cancer Research UK Beatson Institute, Glasgow, G61 1BD, UK
| |
Collapse
|
225
|
Abstract
OBJECTIVES In injury conditions, myofibroblasts are induced to lay down matrix proteins and support the repair process. In this study, we investigated the role of myofibroblasts, particularly stellate cells, in the growth and regeneration of pancreatic β cells. METHODS We used both in vitro and in vivo approaches to address whether stellate cells may promote the growth of β cells. RESULTS Our experiments demonstrated that activated stellate cells support the proliferation of β cells in vitro. In vivo, mesenchymals surrounding the pancreatic islets are activated (induced to proliferate) in the islet regeneration model of Pten null mice. These mesenchymals display markers of pancreatic stellate cells, such as desmin and to a lesser extent, smooth muscle actin α. We have shown previously that targeted β-cell deletion of Pten lead to a significant increase in total islet mass. This phenotype was accompanied by an increase in peri-islet mitotic activity, particularly in islets injured by streptozotocin, a β cell-specific toxin. CONCLUSIONS Together with the in vitro observations, our data, here, suggest that that these mesenchymal cells may support the regeneration of the islets. Identifying how the communication occurs may provide clinically relevant mechanism for inducing β-cell regeneration.
Collapse
|
226
|
Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma. Nat Genet 2015; 47:1168-78. [PMID: 26343385 PMCID: PMC4912058 DOI: 10.1038/ng.3398] [Citation(s) in RCA: 1418] [Impact Index Per Article: 141.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 08/17/2015] [Indexed: 12/11/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a lethal disease with a 5-year survival rate of 4%. A key hallmark of PDAC is extensive stromal involvement, which makes capturing precise tumor-specific molecular information difficult. Here we have overcome this problem by applying blind source separation to a diverse collection of PDAC gene expression microarray data, including data from primary tumor, metastatic and normal samples. By digitally separating tumor, stromal and normal gene expression, we have identified and validated two tumor subtypes, including a 'basal-like' subtype that has worse outcome and is molecularly similar to basal tumors in bladder and breast cancers. Furthermore, we define 'normal' and 'activated' stromal subtypes, which are independently prognostic. Our results provide new insights into the molecular composition of PDAC, which may be used to tailor therapies or provide decision support in a clinical setting where the choice and timing of therapies are critical.
Collapse
|
227
|
Neesse A, Algül H, Tuveson DA, Gress TM. Stromal biology and therapy in pancreatic cancer: a changing paradigm. Gut 2015; 64:1476-84. [PMID: 25994217 DOI: 10.1136/gutjnl-2015-309304] [Citation(s) in RCA: 405] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 03/20/2015] [Indexed: 12/12/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDA) exhibits one of the poorest prognosis of all solid tumours and poses an unsolved problem in cancer medicine. Despite the recent success of two combination chemotherapies for palliative patients, the modest survival benefits are often traded against significant side effects and a compromised quality of life. Although the molecular events underlying the initiation and progression of PDA have been intensively studied and are increasingly understood, the reasons for the poor therapeutic response are hardly apprehended. One leading hypothesis over the last few years has been that the pronounced tumour microenvironment in PDA not only promotes carcinogenesis and tumour progression but also mediates therapeutic resistance. To this end, targeting of various stromal components and pathways was considered a promising strategy to biochemically and biophysically enhance therapeutic response. However, none of the efforts have yet led to efficacious and approved therapies in patients. Additionally, recent data have shown that tumour-associated fibroblasts may restrain rather than promote tumour growth, reinforcing the need to critically revisit the complexity and complicity of the tumour-stroma with translational implications for future therapy and clinical trial design.
Collapse
Affiliation(s)
- Albrecht Neesse
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Centre Goettingen, Georg August University Goettingen, Goettingen, Germany
| | - Hana Algül
- II. Medizinische Klinik und Poliklinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - David A Tuveson
- Cold Spring Harbor Laboratory, Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York, USA
| | - Thomas M Gress
- Department of Gastroenterology, Endocrinology, Infectiology and Metabolism, Philipps-University, Marburg, Germany
| |
Collapse
|
228
|
Li T, Wang YN, Khokhlova TD, D'Andrea S, Starr F, Chen H, McCune JS, Risler LJ, Mashadi-Hossein A, Hingorani SR, Chang A, Hwang JH. Pulsed High-Intensity Focused Ultrasound Enhances Delivery of Doxorubicin in a Preclinical Model of Pancreatic Cancer. Cancer Res 2015. [PMID: 26216548 DOI: 10.1158/0008-5472.can-15-0296] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Pancreatic cancer is characterized by extensive stromal desmoplasia, which decreases blood perfusion and impedes chemotherapy delivery. Breaking the stromal barrier could both increase perfusion and permeabilize the tumor, enhancing chemotherapy penetration. Mechanical disruption of the stroma can be achieved using ultrasound-induced bubble activity-cavitation. Cavitation is also known to result in microstreaming and could have the added benefit of actively enhancing diffusion into the tumors. Here, we report the ability to enhance chemotherapeutic drug doxorubicin penetration using ultrasound-induced cavitation in a genetically engineered mouse model (KPC mouse) of pancreatic ductal adenocarcinoma. To induce localized inertial cavitation in pancreatic tumors, pulsed high-intensity focused ultrasound (pHIFU) was used either during or before doxorubicin administration to elucidate the mechanisms of enhanced drug delivery (active vs. passive drug diffusion). For both types, the pHIFU exposures that were associated with high cavitation activity resulted in disruption of the highly fibrotic stromal matrix and enhanced the normalized doxorubicin concentration by up to 4.5-fold compared with controls. Furthermore, normalized doxorubicin concentration was associated with the cavitation metrics (P < 0.01), indicating that high and sustained cavitation results in increased chemotherapy penetration. No significant difference between the outcomes of the two types, that is, doxorubicin infusion during or after pHIFU treatment, was observed, suggesting that passive diffusion into previously permeabilized tissue is the major mechanism for the increase in drug concentration. Together, the data indicate that pHIFU treatment of pancreatic tumors when resulting in high and sustained cavitation can efficiently enhance chemotherapy delivery to pancreatic tumors. .
Collapse
Affiliation(s)
- Tong Li
- Center for Industrial and Medical Ultrasound, Applied Physics Laboratory, University of Washington, Seattle, Washington
| | - Yak-Nam Wang
- Center for Industrial and Medical Ultrasound, Applied Physics Laboratory, University of Washington, Seattle, Washington
| | - Tatiana D Khokhlova
- Division of Gastroenterology, Department of Medicine, University of Washington, Seattle, Washington
| | - Samantha D'Andrea
- Division of Gastroenterology, Department of Medicine, University of Washington, Seattle, Washington
| | - Frank Starr
- Center for Industrial and Medical Ultrasound, Applied Physics Laboratory, University of Washington, Seattle, Washington
| | - Hong Chen
- Center for Industrial and Medical Ultrasound, Applied Physics Laboratory, University of Washington, Seattle, Washington
| | - Jeannine S McCune
- Department of Pharmacy, University of Washington, Seattle, Washington. Department of Pharmaceutics, University of Washington, Seattle, Washington
| | - Linda J Risler
- Department of Pharmacy, University of Washington, Seattle, Washington. Department of Pharmaceutics, University of Washington, Seattle, Washington
| | | | | | | | - Joo Ha Hwang
- Division of Gastroenterology, Department of Medicine, University of Washington, Seattle, Washington.
| |
Collapse
|
229
|
Neural Regulation of Pancreatic Cancer: A Novel Target for Intervention. Cancers (Basel) 2015; 7:1292-312. [PMID: 26193320 PMCID: PMC4586771 DOI: 10.3390/cancers7030838] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 07/07/2015] [Accepted: 07/13/2015] [Indexed: 12/16/2022] Open
Abstract
The tumor microenvironment is known to play a pivotal role in driving cancer progression and governing response to therapy. This is of significance in pancreatic cancer where the unique pancreatic tumor microenvironment, characterized by its pronounced desmoplasia and fibrosis, drives early stages of tumor progression and dissemination, and contributes to its associated low survival rates. Several molecular factors that regulate interactions between pancreatic tumors and their surrounding stroma are beginning to be identified. Yet broader physiological factors that influence these interactions remain unclear. Here, we discuss a series of preclinical and mechanistic studies that highlight the important role chronic stress plays as a physiological regulator of neural-tumor interactions in driving the progression of pancreatic cancer. These studies propose several approaches to target stress signaling via the β-adrenergic signaling pathway in order to slow pancreatic tumor growth and metastasis. They also provide evidence to support the use of β-blockers as a novel therapeutic intervention to complement current clinical strategies to improve cancer outcome in patients with pancreatic cancer.
Collapse
|
230
|
Tang D, Gao J, Wang S, Yuan Z, Ye N, Chong Y, Xu C, Jiang X, Li B, Yin W, Miao Y, Wang D, Jiang K. Apoptosis and anergy of T cell induced by pancreatic stellate cells-derived galectin-1 in pancreatic cancer. Tumour Biol 2015; 36:5617-26. [PMID: 25725585 DOI: 10.1007/s13277-015-3233-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 02/06/2015] [Indexed: 02/07/2023] Open
Abstract
Galectin-1, a β-galactoside-binding protein implicated in cancer cell immune privilege, was highly expressed in activated pancreatic stellate cells (PSCs). This study was designed to investigate the relationship between PSC-derived galectin-1 and tumor immunity in pancreatic cancer. Isolated PSCs were identified as normal pancreas cells (hNPSCs) or pancreatic cancer cells (hCaPSCs) by immunohistochemical staining for α-SMA and vimentin, and galectin-1 expression was evaluated by Western blotting and quantitative RT-PCR. Apoptosis, caspase activity, and cytokine production (IL-6, IL-10, TNF-β, and IFN-γ) of T cells co-cultured with PSCs were evaluated, and immunohistochemical staining of galectin-1 was correlated with CD3 and clinicopathological variables in 66 pancreatic cancer and 10 normal pancreatic tissue samples. hCaPSCs exhibited higher galectin-1 expression than did hNPSCs, and hCaPSCs induced higher levels of apoptosis in T cells following co-culture. hCaPSCs activated caspase-9 and caspase-3 in the mitochondrial apoptotic pathway and stimulated secretion of Th2 cytokines (IL-6 and IL-10) but decreased secretion of Th1 cytokines (TNF-β and IFN-γ), compared with hNPSCs. Immunohistochemical staining indicated that galectin-1 and CD3 were more highly expressed in pancreatic cancer tissue. Galectin-1 expression was highest in poorly differentiated pancreatic cancer cells and lowest in well-differentiated pancreatic cancer cells and was associated with tumor size, lymph node metastasis, differentiation, and UICC stage. However, CD3 expression showed the opposite trend and was highest in well-differentiated pancreatic cancer cells and was associated with tumor differentiation and UICC stage. High expression of galectin-1 was associated with short survival, as was low expression of CD3. hCaPSC-derived galectin-1 enhanced apoptosis and anergy of T cells in pancreatic cancer, which contributes to the immune escape of pancreatic cancer cells.
Collapse
Affiliation(s)
- Dong Tang
- Department of General Surgery, Clinical Medical College, Yangzhou University (Subei People's Hospital of Jiangsu Province), Yangzhou, 225001, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
231
|
Pathophysiological role of microRNA-29 in pancreatic cancer stroma. Sci Rep 2015; 5:11450. [PMID: 26095125 PMCID: PMC4476113 DOI: 10.1038/srep11450] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 05/26/2015] [Indexed: 12/24/2022] Open
Abstract
Dense fibrotic stroma associated with pancreatic ductal adenocarcinoma (PDAC) is a major obstacle for drug delivery to the tumor bed and plays a crucial role in pancreatic cancer progression. Current, anti-stromal therapies have failed to improve tumor response to chemotherapy and patient survival. Furthermore, recent studies show that stroma impedes tumor progression, and its complete ablation accelerates PDAC progression. In an effort to understand the molecular mechanisms associated with tumor-stromal interactions, using in vitro and in vivo models and PDAC patient biopsies, we show that the loss of miR-29 is a common phenomenon of activated pancreatic stellate cells (PSCs)/fibroblasts, the major stromal cells responsible for fibrotic stromal reaction. Loss of miR-29 is correlated with a significant increase in extracellular matrix (ECM) deposition, a major component in PDAC stroma. Our in vitro miR-29 gain/loss-of-function studies document the role of miR-29 in PSC-mediated ECM stromal protein accumulation. Overexpression of miR-29 in activated stellate cells reduced stromal deposition, cancer cell viability, and cancer growth in co-culture. Furthermore, the loss of miR-29 in TGF-β1 activated PSCs is SMAD3 dependent. These results provide insights into the mechanistic role of miR-29 in PDAC stroma and its potential use as a therapeutic agent to target PDAC.
Collapse
|
232
|
Kiss K, Baghy K, Spisák S, Szanyi S, Tulassay Z, Zalatnai A, Löhr JM, Jesenofsky R, Kovalszky I, Firneisz G. Chronic hyperglycemia induces trans-differentiation of human pancreatic stellate cells and enhances the malignant molecular communication with human pancreatic cancer cells. PLoS One 2015; 10:e0128059. [PMID: 26010611 PMCID: PMC4444240 DOI: 10.1371/journal.pone.0128059] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 04/23/2015] [Indexed: 12/12/2022] Open
Abstract
Background Diabetes mellitus is linked to pancreatic cancer. We hypothesized a role for pancreatic stellate cells (PSC) in the hyperglycemia induced deterioration of pancreatic cancer and therefore studied two human cell lines (RLT-PSC, T3M4) in hyperglycemic environment. Methodology/Principal Findings The effect of chronic hyperglycemia (CHG) on PSCs was studied using mRNA expression array with real-time PCR validation and bioinformatic pathway analysis, and confirmatory protein studies. The stress fiber formation (IC: αSMA) indicated that PSCs tend to transdifferentiate to a myofibroblast-like state after exposure to CHG. The phosphorylation of p38 and ERK1/2 was increased with a consecutive upregulation of CDC25, SP1, cFOS and p21, and with downregulation of PPARγ after PSCs were exposed to chronic hyperglycemia. CXCL12 levels increased significantly in PSC supernatant after CHG exposure independently from TGF-β1 treatment (3.09-fold with a 2.73-fold without TGF-β1, p<0.05). The upregualtion of the SP1 transcription factor in PSCs after CHG exposure may be implicated in the increased CXCL12 and IGFBP2 production. In cancer cells, hyperglycemia induced an increased expression of CXCR4, a CXCL12 receptor that was also induced by PSC’s conditioned medium. The receptor-ligand interaction increased the phosphorylation of ERK1/2 and p38 resulting in activation of MAP kinase pathway, one of the most powerful stimuli for cell proliferation. Certainly, conditioned medium of PSC increased pancreatic cancer cell proliferation and this effect could be partially inhibited by a CXCR4 inhibitor. As the PSC conditioned medium (normal glucose concentration) increased the ERK1/2 and p38 phosphorylation, we concluded that PSCs produce other factor(s) that influence(s) pancreatic cancer behaviour. Conclusions Hyperglycemia induces increased CXCL12 production by the PSCs, and its receptor, CXCR4 on cancer cells. The ligand-receptor interaction activates MAP kinase signaling that causes increased cancer cell proliferation and migration.
Collapse
Affiliation(s)
- Katalin Kiss
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Kornélia Baghy
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Sándor Spisák
- Children's Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Szilárd Szanyi
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany
- School of Ph.D. Studies, Semmelweis University, Budapest, Hungary
| | - Zsolt Tulassay
- 2nd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Attila Zalatnai
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - J.-Matthias Löhr
- Karolinska Institutet, Gastrocentrum, Karolinska University Hospital, Stockholm, Sweden
| | - Ralf Jesenofsky
- University of Heidelberg, Medical Campus Mannheim, Dept. of Medicine II, Mannheim, Germany
| | - Ilona Kovalszky
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Gábor Firneisz
- 2nd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
- * E-mail:
| |
Collapse
|
233
|
Moir JAG, Mann J, White SA. The role of pancreatic stellate cells in pancreatic cancer. Surg Oncol 2015; 24:232-8. [PMID: 26080604 DOI: 10.1016/j.suronc.2015.05.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 05/11/2015] [Indexed: 01/05/2023]
Abstract
BACKGROUND The prognosis of pancreatic cancer remains desperately poor, with little progress made over the past 30 years despite the development of new combination chemotherapy regimens. Stromal activity is especially prominent in the tissue surrounding pancreatic tumours, and has a profound influence in dictating tumour development and dissemination. Pancreatic stellate cells (PaSCs) have a key role in this tumour microenvironment, and have been the subject of much research in the past decade. This review examines the relationship between PaSCs and cancer cells. METHODS A comprehensive literature search was performed of multiple databases up to March 2014, including Medline, Pubmed and Google Scholar. RESULTS A complex bidirectional interplay exists between PaSCs and cancer cells, resulting in a perpetuating loop of increased activity and an overriding pro-tumorigenic effect. This involves a number of signalling pathways that also impacts on other stromal components and vasculature, contributing to chemoresistance. The Reverse Warburg Effect is also introduced as a novel concept in tumour stroma. CONCLUSION This review highlights the pancreatic tumour microenvironment, and in particular PaSCs, as an ideal target for therapeutics. There are a number of cellular processes involving PaSCs which could hold the key to more effectively treating pancreatic cancer. The feasibility of targeting these pathways warrant further in depth investigation, with the aim of reducing the aggressiveness of pancreatic cancer and improving chemodelivery.
Collapse
Affiliation(s)
- John A G Moir
- Freeman Hospital, Department of HPB and Transplant Surgery, Newcastle upon Tyne, United Kingdom; Institute of Cellular Medicine, Fibrosis Lab, Newcastle upon Tyne, United Kingdom.
| | - Jelena Mann
- Institute of Cellular Medicine, Fibrosis Lab, Newcastle upon Tyne, United Kingdom
| | - Steve A White
- Freeman Hospital, Department of HPB and Transplant Surgery, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
234
|
Sherman MH, Yu RT, Engle DD, Ding N, Atkins AR, Tiriac H, Collisson EA, Connor F, Van Dyke T, Kozlov S, Martin P, Tseng TW, Dawson DW, Donahue TR, Masamune A, Shimosegawa T, Apte MV, Wilson JS, Ng B, Lau SL, Gunton JE, Wahl GM, Hunter T, Drebin JA, O'Dwyer PJ, Liddle C, Tuveson DA, Downes M, Evans RM. Vitamin D receptor-mediated stromal reprogramming suppresses pancreatitis and enhances pancreatic cancer therapy. Cell 2015; 159:80-93. [PMID: 25259922 DOI: 10.1016/j.cell.2014.08.007] [Citation(s) in RCA: 856] [Impact Index Per Article: 85.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 07/01/2014] [Accepted: 07/31/2014] [Indexed: 12/14/2022]
Abstract
The poor clinical outcome in pancreatic ductal adenocarcinoma (PDA) is attributed to intrinsic chemoresistance and a growth-permissive tumor microenvironment. Conversion of quiescent to activated pancreatic stellate cells (PSCs) drives the severe stromal reaction that characterizes PDA. Here, we reveal that the vitamin D receptor (VDR) is expressed in stroma from human pancreatic tumors and that treatment with the VDR ligand calcipotriol markedly reduced markers of inflammation and fibrosis in pancreatitis and human tumor stroma. We show that VDR acts as a master transcriptional regulator of PSCs to reprise the quiescent state, resulting in induced stromal remodeling, increased intratumoral gemcitabine, reduced tumor volume, and a 57% increase in survival compared to chemotherapy alone. This work describes a molecular strategy through which transcriptional reprogramming of tumor stroma enables chemotherapeutic response and suggests vitamin D priming as an adjunct in PDA therapy. PAPERFLICK:
Collapse
Affiliation(s)
- Mara H Sherman
- Gene Expression Laboratory, Salk Institute, La Jolla, CA 92037, USA
| | - Ruth T Yu
- Gene Expression Laboratory, Salk Institute, La Jolla, CA 92037, USA
| | | | - Ning Ding
- Gene Expression Laboratory, Salk Institute, La Jolla, CA 92037, USA
| | - Annette R Atkins
- Gene Expression Laboratory, Salk Institute, La Jolla, CA 92037, USA
| | - Herve Tiriac
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Eric A Collisson
- Department of Medicine/Hematology and Oncology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Frances Connor
- Cancer Research UK Cambridge Research Institute, The Li Ka Shing Centre, Robinson Way, Cambridge CB2 ORE, UK
| | - Terry Van Dyke
- Center for Advanced Preclinical Research, NCI-Frederick, Frederick, MD 21702, USA
| | - Serguei Kozlov
- Center for Advanced Preclinical Research, Leidos Biomed, Inc. Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Philip Martin
- Center for Advanced Preclinical Research, Leidos Biomed, Inc. Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Tiffany W Tseng
- Gene Expression Laboratory, Salk Institute, La Jolla, CA 92037, USA
| | - David W Dawson
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Timothy R Donahue
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Atsushi Masamune
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai Miyagi, 980-8574, Japan
| | - Tooru Shimosegawa
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai Miyagi, 980-8574, Japan
| | - Minoti V Apte
- Pancreatic Research Group, Faculty of Medicine, South Western Sydney Clinical School, University of New South Wales, Sydney, NSW 2052, Australia
| | - Jeremy S Wilson
- Pancreatic Research Group, Faculty of Medicine, South Western Sydney Clinical School, University of New South Wales, Sydney, NSW 2052, Australia
| | - Beverly Ng
- Diabetes and Transcription Factors Group, Garvan Institute of Medical Research (GIMR), Sydney, NSW 2010, Australia; St Vincent's Clinical School, University of New South Wales, Sydney, NSW 2052, Australia
| | - Sue Lynn Lau
- Diabetes and Transcription Factors Group, Garvan Institute of Medical Research (GIMR), Sydney, NSW 2010, Australia; Faculty of Medicine, University of Sydney, Sydney, NSW 2052, Australia; Department of Diabetes and Endocrinology, Westmead Hospital, Sydney, NSW 2145, Australia
| | - Jenny E Gunton
- Diabetes and Transcription Factors Group, Garvan Institute of Medical Research (GIMR), Sydney, NSW 2010, Australia; St Vincent's Clinical School, University of New South Wales, Sydney, NSW 2052, Australia; Faculty of Medicine, University of Sydney, Sydney, NSW 2052, Australia; Department of Diabetes and Endocrinology, Westmead Hospital, Sydney, NSW 2145, Australia
| | - Geoffrey M Wahl
- Gene Expression Laboratory, Salk Institute, La Jolla, CA 92037, USA
| | - Tony Hunter
- Molecular and Cell Biology Laboratory, Salk Institute, La Jolla, CA 92037, USA
| | - Jeffrey A Drebin
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Peter J O'Dwyer
- Abramson Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Christopher Liddle
- The Storr Liver Unit, Westmead Millennium Institute and University of Sydney, Westmead Hospital, Westmead, NSW 2145, Australia
| | - David A Tuveson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Michael Downes
- Gene Expression Laboratory, Salk Institute, La Jolla, CA 92037, USA.
| | - Ronald M Evans
- Gene Expression Laboratory, Salk Institute, La Jolla, CA 92037, USA; Howard Hughes Medical Institute, Salk Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
235
|
Susanto JM, Colvin EK, Pinese M, Chang DK, Pajic M, Mawson A, Caldon CE, Musgrove EA, Henshall SM, Sutherland RL, Biankin AV, Scarlett CJ. The epigenetic agents suberoylanilide hydroxamic acid and 5‑AZA‑2' deoxycytidine decrease cell proliferation, induce cell death and delay the growth of MiaPaCa2 pancreatic cancer cells in vivo. Int J Oncol 2015; 46:2223-30. [PMID: 25695794 DOI: 10.3892/ijo.2015.2894] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 09/07/2014] [Indexed: 11/06/2022] Open
Abstract
Despite incremental advances in the diagnosis and treatment for pancreatic cancer (PC), the 5‑year survival rate remains <5%. Novel therapies to increase survival and quality of life for PC patients are desperately needed. Epigenetic thera-peutic agents such as histone deacetylase inhibitors (HDACi) and DNA methyltransferase inhibitors (DNMTi) have demonstrated therapeutic benefits in human cancer. We assessed the efficacy of these epigenetic therapeutic agents as potential therapies for PC using in vitro and in vivo models. Treatment with HDACi [suberoylanilide hydroxamic acid (SAHA)] and DNMTi [5‑AZA‑2' deoxycytidine (5‑AZA‑dc)] decreased cell proliferation in MiaPaCa2 cells, and SAHA treatment, with or without 5‑AZA‑dc, resulted in higher cell death and lower DNA synthesis compared to 5‑AZA‑dc alone and controls (DMSO). Further, combination treatment with SAHA and 5‑AZA‑dc significantly increased expression of p21WAF1, leading to G1 arrest. Treatment with epigenetic agents delayed tumour growth in vivo, but did not decrease growth of established pancreatic tumours. In conclusion, these data demonstrate a potential role for epigenetic modifier drugs for the management of PC, specifically in the chemoprevention of PC, in combination with other chemotherapeutic agents.
Collapse
Affiliation(s)
- Johana M Susanto
- Cancer Research Program, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Emily K Colvin
- Cancer Research Program, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Mark Pinese
- Cancer Research Program, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - David K Chang
- Cancer Research Program, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Marina Pajic
- Cancer Research Program, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Amanda Mawson
- Cancer Research Program, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - C Elizabeth Caldon
- Cancer Research Program, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Elizabeth A Musgrove
- Cancer Research Program, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Susan M Henshall
- Cancer Research Program, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Robert L Sutherland
- Cancer Research Program, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Andrew V Biankin
- Cancer Research Program, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Christopher J Scarlett
- Cancer Research Program, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| |
Collapse
|
236
|
The chemokine receptors CXCR4/CXCR7 and their primary heterodimeric ligands CXCL12 and CXCL12/high mobility group box 1 in pancreatic cancer growth and development: finding flow. Pancreas 2015; 44:528-34. [PMID: 25872129 DOI: 10.1097/mpa.0000000000000298] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Novel therapies need to be developed for patients with pancreatic cancer because of the poor outcomes of current regimens. Pancreatic cancer cells respond to the C-X-C chemokine receptor type 4 (CXCR4)/C-X-C chemokine receptor type 7 (CXCR7)/C-X-C motif chemokine 12 (CXCL12)/high-mobility group box 1 signaling axis and this axis presents a novel target for therapy. C-X-C motif chemokine 12 stimulates CXCR4/CXCR7-bearing cells in a paracrine manner. C-X-C chemokine receptor type 4 and CXCR7 are transmembrane G protein-coupled receptors that, upon interaction with ligand CXCL12, activate downstream protein kinases that promote a more aggressive behavior. C-X-C chemokine receptor type 4 is expressed on most pancreatic cancer cells, whereas CXCR7 is primarily expressed on tumor-associated endothelium. High-mobility group box 1 promotes the CXCR4 and CXCL12 interaction, promoting angiogenesis and lymphangiogenesis. Hypoxia-inducible factor 1 is a potent stimulator of CXCR4 and CXCL12 expression, promoting more aggressive behavior. This receptor/ligand interaction can be disrupted by CXCR4 antagonists available and in clinical use to harvest bone marrow stem cells. Novel imaging strategies are now being developed at several centers to evaluate response to therapy and identify early recurrence. Thus, the CXCR4/CXCR7/CXCL12 interaction plays a critical role in cancer cell progression, proliferation, invasion, as well as metastasis and is a suitable target for therapy, imaging, as well as development of novel diagnostics.
Collapse
|
237
|
A multistep high-content screening approach to identify novel functionally relevant target genes in pancreatic cancer. PLoS One 2015; 10:e0122946. [PMID: 25849100 PMCID: PMC4388713 DOI: 10.1371/journal.pone.0122946] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 12/30/2014] [Indexed: 01/05/2023] Open
Abstract
In order to foster the systematic identification of novel genes with important functional roles in pancreatic cancer, we have devised a multi-stage screening strategy to provide a rational basis for the selection of highly relevant novel candidate genes based on the results of functional high-content analyses. The workflow comprised three consecutive stages: 1) serial gene expression profiling analyses of primary human pancreatic tissues as well as a number of in vivo and in vitro models of tumor-relevant characteristics in order to identify genes with conspicuous expression patterns; 2) use of ‘reverse transfection array’ technology for large-scale parallelized functional analyses of potential candidate genes in cell-based assays; and 3) selection of individual candidate genes for further in-depth examination of their cellular roles. A total of 14 genes, among them 8 from “druggable” gene families, were classified as high priority candidates for individual functional characterization. As an example to demonstrate the validity of the approach, comprehensive functional data on candidate gene ADRBK1/GRK2, which has previously not been implicated in pancreatic cancer, is presented.
Collapse
|
238
|
Liu Y, Du L. Role of pancreatic stellate cells and periostin in pancreatic cancer progression. Tumour Biol 2015; 36:3171-7. [PMID: 25840689 DOI: 10.1007/s13277-015-3386-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 03/24/2015] [Indexed: 12/30/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive and one of the five most lethal malignancies characterized by prominent desmoplastic reaction. Accumulating evidences indicate that tumor desmoplasia plays a pivotal role in PDAC progression, and it has been largely ignored until recent times. It has now been unequivocally shown that pancreatic stellate cells (PSCs) are the principal effector cells responsible for stroma production. Periostin, also known as osteoblast-specific factor 2, is a secretory protein and originally identified as an osteoblast-specific factor that expressed in periosteum. Periostin is exclusively produced by activated PSCs, and periostin overexpression presents in various malignant tumors and closely relates with disease progression. In addition, periostin has been suggested to stimulate pancreatic cancer cells proliferation and enhance their resistance to serum starvation and hypoxia. Therefore, the interplay between cancer cells and stromal cells plays a vital role in PDAC development. However, the function of periostin in pancreatic cancer development is controversial. This review summarizes existing knowledge about the role of PSCs in cancer stroma production, the interaction between PSCs and pancreatic cancer cells, tumor angiogenesis, and hypoxic microenvironment, with particular focus on the expression and function as well as signaling pathways of periostin in PDAC cells and PSCs.
Collapse
Affiliation(s)
- Yang Liu
- Department of Ultrasound, Shanghai First People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200080, People's Republic of China
| | | |
Collapse
|
239
|
Sharbeen G, McCarroll J, Goldstein D, Phillips PA. Exploiting base excision repair to improve therapeutic approaches for pancreatic cancer. Front Nutr 2015; 2:10. [PMID: 25988138 PMCID: PMC4428371 DOI: 10.3389/fnut.2015.00010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 03/10/2015] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) is a highly chemoresistant and metastatic disease with a dismal 5-year survival rate of 6%. More effective therapeutic targets and approaches are urgently needed to tackle this devastating disease. The base excision repair (BER) pathway has been identified as a predictor of therapeutic response, prognostic factor, and therapeutic target in a variety of cancers. This review will discuss our current understanding of BER in PDA and its potential to improve PDA treatment.
Collapse
Affiliation(s)
- George Sharbeen
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, Prince of Wales Clinical School, UNSW Australia , Sydney, NSW , Australia
| | - Joshua McCarroll
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Australia , Sydney, NSW , Australia
| | - David Goldstein
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, Prince of Wales Clinical School, UNSW Australia , Sydney, NSW , Australia
| | - Phoebe A Phillips
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, Prince of Wales Clinical School, UNSW Australia , Sydney, NSW , Australia
| |
Collapse
|
240
|
Xu Y, Li H, Huang C, Zhao T, Zhang H, Zheng C, Ren H, Hao J. Wnt2 protein plays a role in the progression of pancreatic cancer promoted by pancreatic stellate cells. Med Oncol 2015; 32:97. [PMID: 25731618 DOI: 10.1007/s12032-015-0513-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Accepted: 02/13/2015] [Indexed: 12/20/2022]
Abstract
This study aimed to investigate the expression of Wnt2 protein in pancreatic cancer tissues and pancreatic stellate cells (PSCs), and determine its effect on the biological functions of pancreatic cancer cells. Immunohistochemistry was used to study the expression pattern of Wnt2 in pancreatic cancer tissues. The relationship between Wnt2 protein expression level and patient prognosis was analyzed. PSCs were isolated and cultured. The expression of Wnt2 in activated PSCs was investigated using Western blot and immunofluorescence. We also analyzed the effect of Wnt2 recombinant protein and stellate cell culture supernatant on the Wnt/β-catenin signaling pathway, as well as the effect of Wnt2 recombinant protein on the biological functions of pancreatic cancer cells. The expression of Wnt2 in interstitial cells of pancreatic cancer was correlated with the prognosis of pancreatic cancer. Wnt2 protein was expressed in activated PSCs. Both stellate cell culture supernatant and Wnt2 recombinant protein could activate the classic Wnt/β-catenin signaling pathway. Wnt2 protein enhanced the migration, invasion, and metastasis of pancreatic cancer cells. These results suggested that Wnt2 protein secreted by PSCs promoted the progression of pancreatic cancer by activating the classic Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Yong Xu
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | | | | | | | | | | | | | | |
Collapse
|
241
|
Wu HH, Hwang-Verslues WW, Lee WH, Huang CK, Wei PC, Chen CL, Shew JY, Lee EYHP, Jeng YM, Tien YW, Ma C, Lee WH. Targeting IL-17B-IL-17RB signaling with an anti-IL-17RB antibody blocks pancreatic cancer metastasis by silencing multiple chemokines. ACTA ACUST UNITED AC 2015; 212:333-49. [PMID: 25732306 PMCID: PMC4354366 DOI: 10.1084/jem.20141702] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pancreatic cancer has an extremely high mortality rate due to its aggressive metastatic nature. Resolving the underlying mechanisms will be crucial for treatment. Here, we found that overexpression of IL-17B receptor (IL-17RB) strongly correlated with postoperative metastasis and inversely correlated with progression-free survival in pancreatic cancer patients. Consistently, results from ex vivo experiments further validated that IL-17RB and its ligand, IL-17B, plays an essential role in pancreatic cancer metastasis and malignancy. Signals from IL-17B-IL-17RB activated CCL20/CXCL1/IL-8/TFF1 chemokine expressions via the ERK1/2 pathway to promote cancer cell invasion, macrophage and endothelial cell recruitment at primary sites, and cancer cell survival at distant organs. Treatment with a newly derived monoclonal antibody against IL-17RB blocked tumor metastasis and promoted survival in a mouse xenograft model. These findings not only illustrate a key mechanism underlying the highly aggressive characteristics of pancreatic cancer but also provide a practical approach to tackle this disease.
Collapse
Affiliation(s)
- Heng-Hsiung Wu
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | | | - Wen-Hsin Lee
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Chun-Kai Huang
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Pei-Chi Wei
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Chia-Lin Chen
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Jin-Yuh Shew
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Eva Y-H P Lee
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697
| | - Yung-Ming Jeng
- Department of Pathology and Department of Surgery, National Taiwan University Hospital, Taipei 10617, Taiwan
| | - Yu-Wen Tien
- Department of Pathology and Department of Surgery, National Taiwan University Hospital, Taipei 10617, Taiwan
| | - Che Ma
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Wen-Hwa Lee
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan Graduate Institute of Clinical Medicine, China Medical University, Taichung 40402, Taiwan
| |
Collapse
|
242
|
Cao F, Li J, Sun H, Liu S, Cui Y, Li F. HES 1 is essential for chemoresistance induced by stellate cells and is associated with poor prognosis in pancreatic cancer. Oncol Rep 2015; 33:1883-9. [PMID: 25672829 DOI: 10.3892/or.2015.3789] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 01/07/2015] [Indexed: 11/06/2022] Open
Abstract
The role of pancreatic stellate cells (PSCs) has been established in many studies. However, the potential mechanism for the chemoresistance induced by PSCs has not been fully elucidated. In the present study, human pancreatic cancer cell lines were directly or indirectly co-cultured with PSCs. The inhibition rate and IC50 values were assessed to determine the ability of chemoresistance. RT-PCR and western blot analysis were used to evaluate Hes 1 and Jagged 1 expression before and after co-culture with PSCs. To determine the relationship between Hes 1 expression and survival in pancreatic cancer patients, Kaplan-Meier survival analysis was performed. PSCs promoted the expression of Hes 1 in both PANC-1 and BxPC-3 cell lines and induced chemoresistance to gemcitabine. A Notch signaling pathway inhibitor (L1790) and Hes 1 siRNA reversed the chemoresistance induced by PSCs. In 72 resected pancreatic cancer patients, high Hes 1 expression was observed in 34 patients with shorter overall and progression-free survival times. In conclusion, Hes 1 is essential for chemoresistance induced by PSCs and is associated with poor prognosis in pancreatic cancer patients. Therapy targeting the Notch signaling pathway may reverse chemoresistance and improve survival in patients with pancreatic cancer.
Collapse
Affiliation(s)
- Feng Cao
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Jia Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Haichen Sun
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Shuang Liu
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Yeqing Cui
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Fei Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| |
Collapse
|
243
|
Zang G, Sandberg M, Carlsson PO, Welsh N, Jansson L, Barbu A. Activated pancreatic stellate cells can impair pancreatic islet function in mice. Ups J Med Sci 2015; 120:169-80. [PMID: 25854824 PMCID: PMC4526872 DOI: 10.3109/03009734.2015.1032453] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Pancreatic or islet fibrosis is often associated with activated pancreatic stellate cells (PSCs). PSCs are considered not only to promote fibrosis, but also to be associated with glucose intolerance in some diseases. We therefore evaluated morphological and functional relationships between islets and PSCs in the normal mouse pancreas and transplanted islets. METHODS Immunohistochemistry was used to map the presence of PSCs in the normal mouse pancreas and islets implanted under the renal capsule. We isolated and cultured mouse PSCs and characterized them morphologically by immunofluorescence staining. Furthermore, we measured their cytokine production and determined their effects on insulin release from simultaneously cultured islets. RESULTS PSCs were scattered throughout the pancreas, with occasional cells within the islets, particularly in the islet capsule. In islet transplants they were found mainly in the graft periphery. Cultured PSCs became functionally activated and produced several cytokines. Throughout the culture period they linearly increased their production of interleukin-6 and mammalian keratinocyte-derived chemokine. PSC cytokine production was not affected by acute hyperglycemia. Syngeneic islets co-cultured with PSCs for 24-48 h increased their insulin release and lowered their insulin content. However, short-term insulin release in batch-type incubations was unaffected after 48 h of co-culture. Increased islet cell caspase-3 activation and a decreased islet cell replication were consistently observed after co-culture for 2 or 7 days. CONCLUSION Activated PSCs may contribute to impaired islet endocrine function seen in exocrine pancreatitis and in islet fibrosis associated with some cases of type 2 diabetes.
Collapse
Affiliation(s)
- Guangxiang Zang
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Monica Sandberg
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Per-Ola Carlsson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Nils Welsh
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Leif Jansson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Andreea Barbu
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
244
|
Apte MV, Pirola RC, Wilson JS. Pancreatic Stellate Cells. STELLATE CELLS IN HEALTH AND DISEASE 2015:271-306. [DOI: 10.1016/b978-0-12-800134-9.00016-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
245
|
Matsuoka Y, Yoshida R, Nakayama H, Nagata M, Hirosue A, Tanaka T, Kawahara K, Nakagawa Y, Sakata J, Arita H, Hiraki A, Shinohara M. The tumour stromal features are associated with resistance to 5-FU-based chemoradiotherapy and a poor prognosis in patients with oral squamous cell carcinoma. APMIS 2014; 123:205-14. [PMID: 25639735 DOI: 10.1111/apm.12344] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 10/17/2014] [Indexed: 12/18/2022]
Abstract
It has been increasingly recognized that the tumour microenvironment is a critical factor involved in cancer progression. However, little is known about the clinical value of the stromal features in oral squamous cell carcinoma (OSCC). The purpose of this study was to determine the clinical significance of cancer-associated fibroblasts (CAFs) and tumour-associated macrophages (TAMs) in OSCC. OSCC specimens were obtained from 60 patients who underwent surgery following 5-fluorouracil-based chemoradiotherapy. Paraffin-embedded sections obtained from biopsy specimens were immunohistochemically analysed. The associations among CAFs, TAMs and various clinicopathological features were examined, and the effects of CAFs and TAMs on the prognosis were evaluated. In the group with a high level of CAFs, the incidence of advanced pT- and pN-stage cases was significantly higher than that in the group with the low level. A high TAMs tumour expression was significantly correlated with a poor response to preoperative chemoradiotherapy. A Kaplan-Meier analysis revealed that higher numbers of CAFs and TAMs were significantly correlated with a poor prognosis. These findings suggest that TAMs are a potential biomarker for predicting the clinical response to 5-FU-based chemoradiotherapy, and the expression status of the CAFs and TAMs may be useful for making treatment decisions to improve the survival of OSCC patients.
Collapse
Affiliation(s)
- Yuichiro Matsuoka
- Departments of Oral and Maxillofacial Surgery, Kumamoto University Graduate School of Life Sciences, Kumamoto, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
246
|
Zhu J, Thakolwiboon S, Liu X, Zhang M, Lubman DM. Overexpression of CD90 (Thy-1) in pancreatic adenocarcinoma present in the tumor microenvironment. PLoS One 2014; 9:e115507. [PMID: 25536077 PMCID: PMC4275230 DOI: 10.1371/journal.pone.0115507] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 11/24/2014] [Indexed: 01/15/2023] Open
Abstract
CD90 (Thy-1) plays important roles in oncogenesis and shows potential as a candidate marker for cancer stem cells (CSCs) in various malignancies. Herein, we investigated the expression of CD90 in pancreatic adenocarcinoma (PDAC), with a comparison to normal pancreas and non-malignant pancreatic disease, by immunohistochemical (IHC) analysis of tissue microarrays containing 183 clinical tissue specimens. Statistical analysis was performed to evaluate the correlation between CD90 expression and the major clinicopathological factors after adjustment of age and gender. The IHC data showed that CD90 was significantly overexpressed in PDAC and its metastatic cancers as compared to chronic pancreatitis and benign islet tumors, while it was negative in normal pancreas and 82.7% of adjacent normal pancreas tissues. The abundant CD90 expression was predominantly present in PDAC stroma, such as fibroblasts and vascular endothelial cells, which could serve as a promising marker to distinguish pancreatic adenocarcinoma from normal pancreas and non-malignant pancreatic diseases. Double immunostaining of CD90 with CD24, a CSC marker for PDAC, showed that there was little overlap between these two markers. However, CD90+ fibroblast cells were clustered around CD24+ malignant ducts, suggesting that CD90 may be involved in the tumor-stroma interactions and promote pancreatic cancer development. Furthermore, CD90 mostly overlapped with α-smooth muscle actin (αSMA, a marker of activated pancreatic stellate cells (PSCs)) in PDAC stroma, which demonstrated that CD90+ stromal cells consist largely of activated PSCs. Double immunostaining of CD90 and a vascular endothelial cell marker CD31 demonstrated that CD90 expression on vascular endothelial cells was significantly increased in PDACs as compared to normal pancreas and non-malignant pancreatic diseases. Our findings suggest that CD90 could serve as a promising marker for pancreatic adenocarcinoma where desmoplastic stroma plays an important role in tumor growth and angiogenesis.
Collapse
Affiliation(s)
- Jianhui Zhu
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan 48109, United States of America
| | - Smathorn Thakolwiboon
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan 48109, United States of America
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Xinhua Liu
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan 48109, United States of America
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Min Zhang
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, Michigan 48109, United States of America
| | - David M. Lubman
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan 48109, United States of America
| |
Collapse
|
247
|
Neuzillet C, de Gramont A, Tijeras-Raballand A, de Mestier L, Cros J, Faivre S, Raymond E. Perspectives of TGF-β inhibition in pancreatic and hepatocellular carcinomas. Oncotarget 2014; 5:78-94. [PMID: 24393789 PMCID: PMC3960190 DOI: 10.18632/oncotarget.1569] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Advanced pancreatic ductal adenocarcinoma (PDAC) and hepatocellular carcinoma (HCC) are non-curable diseases with a particularly poor prognosis. Over the last decade, research has increasingly focused on the microenvironment surrounding cancer cells, and its role in tumour development and progression. PDAC and HCC differ markedly regarding their pathological features: PDAC are typically stromal-predominant, desmoplastic, poorly vascularized tumours, whereas HCC are cellular and highly vascularized. Despite these very different settings, PDAC and HCC share transforming growth factor-β (TGF-β) as a common key-signalling mediator, involved in epithelial-to-mesenchymal transition, invasion, and stroma-tumour dialogue. Recently, novel drugs blocking the TGF-β pathway have entered clinical evaluation demonstrating activity in patients with advanced PDAC and HCC. TGF-β signalling is complex and mediates both pro- and anti-tumoural activities in cancer cells depending on their context, in space and time, and their microenvironment. In this review we provide a comprehensive overview of the role of the TGF-β pathway and its deregulation in PDAC and HCC development and progression at the cellular and microenvironment levels. We also summarize key preclinical and clinical data on the role of TGF-β as a target for therapeutic intervention in PDAC and HCC, and explore perspectives to optimize TGF-β inhibition therapy.
Collapse
Affiliation(s)
- Cindy Neuzillet
- INSERM U728 and U773 and Department of Medical Oncology, Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), 100 boulevard du Général Leclerc, Clichy, France
| | | | | | | | | | | | | |
Collapse
|
248
|
Farid IM, Hamza IM, El-Abd DM, Mohyi AM, AbdulLatif MMA, Aref AT, Hamza DM. Transforming growth factor-β1 gene expression in hepatocellular carcinoma: a preliminary report. Arab J Gastroenterol 2014; 15:142-7. [PMID: 25475758 DOI: 10.1016/j.ajg.2014.10.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 09/05/2014] [Accepted: 10/28/2014] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND STUDY AIMS The transforming growth factor (TGF)-β signalling pathway plays a dual role in hepatocarcinogenesis. It has been recognised for its role as a tumour suppressor as well as a tumour promoter depending on the cellular context. The aim of this study was to investigate the clinical significance of serum TGF-β1 level and TGF-β1 messenger RNA (mRNA) in the peripheral blood of liver cirrhosis and hepatocellular carcinoma (HCC) patients as noninvasive biomarkers in diagnosing HCC. PATIENTS AND METHODS Twenty patients were allocated to each of the liver cirrhosis and HCC groups, in addition to 20 healthy volunteers. TGF-β1 gene expression in peripheral blood was quantitated using real-time polymerase chain reaction (PCR), while serum TGF-β1 was analysed using enzyme-linked immunosorbent assay (ELISA). RESULTS TGF-β1 gene expression was significantly lower in HCC patients (median 0.401 (0.241-0.699) fold change) than in liver cirrhosis patients (median 0.595 (0.464-0.816)) (p=0.042) and normal controls (median 1.00 (0.706-1.426) fold change) (p=0.001). TGF-β1 gene expression showed significant positive correlation with serum TGF-β1 (r=0.272, p=0.036) and significant negative correlation with alpha-fetoprotein (AFP) (r=-0.528, p=0.001). Receiver operating characteristic (ROC) analysis was conducted for TGF-β1 gene expression in comparison with AFP. The area under the curve for TGF-β1 gene expression was 0.688 (95% CI=0.517-0.858) (p=0.042) and AFP was 0.869 (95% CI=0.761-0.976) (p=0.001). The sensitivity and specificity of TGF-β1 gene expression were 65% and 75%, respectively, at a cutoff value of 0.462 fold change. CONCLUSION TGF-β1 gene expression in the peripheral blood may be used as a molecular marker for the diagnosis of HCC. Additional studies on a large-scale population are necessary to gain greater insight into the impact of TGF-β1 gene expression in the pathogenesis of HCC.
Collapse
Affiliation(s)
- Ibtisam M Farid
- Clinical and Chemical Pathology Department, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Iman M Hamza
- Endemic Medicine Department, Faculty of Medicine, Cairo University, Giza, Egypt.
| | - Dina M El-Abd
- Clinical and Chemical Pathology Department, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Abeer M Mohyi
- Clinical and Chemical Pathology Department, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Mona M A AbdulLatif
- Clinical and Chemical Pathology Department, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Adel T Aref
- Oncology Department, Dubai Hospital, Dubai, United Arab Emirates
| | - Dina M Hamza
- Oncology Department, Dubai Hospital, Dubai, United Arab Emirates
| |
Collapse
|
249
|
Lunardi S, Jamieson NB, Lim SY, Griffiths KL, Carvalho-Gaspar M, Al-Assar O, Yameen S, Carter RC, McKay CJ, Spoletini G, D'Ugo S, Silva MA, Sansom OJ, Janssen KP, Muschel RJ, Brunner TB. IP-10/CXCL10 induction in human pancreatic cancer stroma influences lymphocytes recruitment and correlates with poor survival. Oncotarget 2014; 5:11064-11080. [PMID: 25415223 PMCID: PMC4294325 DOI: 10.18632/oncotarget.2519] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 09/24/2014] [Indexed: 01/04/2023] Open
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.
Collapse
Affiliation(s)
- Serena Lunardi
- Gray Institute for Radiation Oncology and Biology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Nigel B. Jamieson
- West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, United Kingdom
| | - Su Yin Lim
- Gray Institute for Radiation Oncology and Biology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | | | - Manuela Carvalho-Gaspar
- Gray Institute for Radiation Oncology and Biology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Osama Al-Assar
- Gray Institute for Radiation Oncology and Biology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Sabira Yameen
- Gray Institute for Radiation Oncology and Biology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Ross C. Carter
- West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, United Kingdom
| | - Colin J. McKay
- West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, United Kingdom
| | - Gabriele Spoletini
- Hepatobiliary and Pancreatic Surgery, Churchill Hospital, Oxford, United Kingdom
| | - Stefano D'Ugo
- Hepatobiliary and Pancreatic Surgery, Churchill Hospital, Oxford, United Kingdom
| | - Michael A. Silva
- Hepatobiliary and Pancreatic Surgery, Churchill Hospital, Oxford, United Kingdom
| | - Owen J. Sansom
- Beatson Institute of Cancer Research, Garscube Estate, Glasgow, United Kingdom
| | - Klaus-Peter Janssen
- Department of Surgery, Technische Universitaet Muenchen, 81675 Muenchen, Germany
| | - Ruth J. Muschel
- Gray Institute for Radiation Oncology and Biology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Thomas B. Brunner
- Gray Institute for Radiation Oncology and Biology, Department of Oncology, University of Oxford, Oxford, United Kingdom
- Department of Radiation Oncology, University Hospitals Freiburg, 79106 Freiburg, Germany
| |
Collapse
|
250
|
Gonzalez-Villasana V, Rodriguez-Aguayo C, Arumugam T, Cruz-Monserrate Z, Fuentes-Mattei E, Deng D, Hwang RF, Wang H, Ivan C, Garza RJ, Cohen E, Gao H, Armaiz-Pena GN, Del C Monroig-Bosque P, Philip B, Rashed MH, Aslan B, Erdogan MA, Gutierrez-Puente Y, Ozpolat B, Reuben JM, Sood AK, Logsdon C, Lopez-Berestein G. Bisphosphonates inhibit stellate cell activity and enhance antitumor effects of nanoparticle albumin-bound paclitaxel in pancreatic ductal adenocarcinoma. Mol Cancer Ther 2014; 13:2583-94. [PMID: 25193509 PMCID: PMC4221441 DOI: 10.1158/1535-7163.mct-14-0028] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Pancreatic stellate cells (PSC) have been recognized as the principal cells responsible for the production of fibrosis in pancreatic ductal adenocarcinoma (PDAC). Recently, PSCs have been noted to share characteristics with cells of monocyte-macrophage lineage (MML cells). Thus, we tested whether PSCs could be targeted with the nitrogen-containing bisphosphonates (NBP; pamidronate or zoledronic acid), which are potent MML cell inhibitors. In addition, we tested NBPs treatment combination with nanoparticle albumin-bound paclitaxel (nab-paclitaxel) to enhance antitumor activity. In vitro, we observed that PSCs possess α-naphthyl butyrate esterase (ANBE) enzyme activity, a specific marker of MML cells. Moreover, NBPs inhibited PSCs proliferation, activation, release of macrophage chemoattractant protein-1 (MCP-1), and type I collagen expression. NBPs also induced PSCs apoptosis and cell-cycle arrest in the G1 phase. In vivo, NBPs inactivated PSCs; reduced fibrosis; inhibited tumor volume, tumor weight, peritoneal dissemination, angiogenesis, and cell proliferation; and increased apoptosis in an orthotopic murine model of PDAC. These in vivo antitumor effects were enhanced when NBPs were combined with nab-paclitaxel but not gemcitabine. Our study suggests that targeting PSCs and tumor cells with NBPs in combination with nab-paclitaxel may be a novel therapeutic approach to PDAC.
Collapse
Affiliation(s)
- Vianey Gonzalez-Villasana
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cristian Rodriguez-Aguayo
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Thiruvengadam Arumugam
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zobeida Cruz-Monserrate
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Enrique Fuentes-Mattei
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Defeng Deng
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rosa F Hwang
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Huamin Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cristina Ivan
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Raul Joshua Garza
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Evan Cohen
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hui Gao
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Guillermo N Armaiz-Pena
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Paloma Del C Monroig-Bosque
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas. University of Puerto Rico School of Medicine, San Juan, Puerto Rico
| | - Bincy Philip
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mohammed H Rashed
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas. Al-Azhar University, Cairo, Egypt
| | - Burcu Aslan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mumin Alper Erdogan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Bulent Ozpolat
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - James M Reuben
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anil K Sood
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Center for RNAi and Non-coding RNA, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Craig Logsdon
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gabriel Lopez-Berestein
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas. Center for RNAi and Non-coding RNA, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|