51
|
Imai D, Yoshizumi T, Okano S, Itoh S, Ikegami T, Harada N, Aishima S, Oda Y, Maehara Y. IFN-γ Promotes Epithelial-Mesenchymal Transition and the Expression of PD-L1 in Pancreatic Cancer. J Surg Res 2019; 240:115-123. [PMID: 30927618 DOI: 10.1016/j.jss.2019.02.038] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 02/06/2019] [Accepted: 02/22/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Tumor immune reactions not only provide host defense but also accelerate tumor immune escape and phenotype switching. Here, we examined the association of programmed cell death ligand 1 (PD-L1) expression with epithelial-mesenchymal transition (EMT)-associated markers in pancreatic ductal adenocarcinoma (PDA) within the context of the tumor microenvironment. MATERIALS AND METHODS PDA samples from 36 patients were analyzed for PD-L1, vimentin, E-cadherin, and Snail expressions and for PDA cell and immune cell infiltration. PD-L1 expression and EMT in PDA cell lines under conditions of altering interferon gamma (IFN-γ) signals were also assessed. RESULTS Immunohistochemistry revealed a significant correlation between vimentin and PD-L1 expression, whereas double staining showed them to be simultaneously expressed by PDA cells. Positive vimentin expression was associated with the infiltration of a lower number of CD8+ T cells and a higher number of FoxP3+ cells and poor patient prognosis (P = 0.03). PDA tumor cells promoted PD-L1 expression and EMT under the presence of IFN-γ, which was inhibited by the signal transducer and activator of transcription (STAT)1 small interfering RNA. CONCLUSIONS Strong correlations were observed between PD-L1 expression, EMT, and the immunosuppressive tumor microenvironment. Targeting STAT1 combined with PD-1/PD-L1 immunotherapy may improve outcomes for patients with PDA.
Collapse
Affiliation(s)
- Daisuke Imai
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomoharu Yoshizumi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Shinji Okano
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shinji Itoh
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toru Ikegami
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Noboru Harada
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shinichi Aishima
- Department of Pathology and Microbiology, Saga Medical School Faculty of Medicine, Saga University, Saga, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Pathological Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshihiko Maehara
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
52
|
Awaji M, Singh RK. Cancer-Associated Fibroblasts' Functional Heterogeneity in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2019; 11:cancers11030290. [PMID: 30832219 PMCID: PMC6468677 DOI: 10.3390/cancers11030290] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 02/22/2019] [Accepted: 02/26/2019] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a leading cause of cancer-related deaths in the USA. Desmoplasia and inflammation are two major hallmarks of PDAC. Desmoplasia, composed of extracellular matrix (ECM), cancer-associated fibroblasts (CAFs), and infiltrating immune and endothelial cells, acts as a biophysical barrier to hinder chemotherapy and actively contributes to tumor progression and metastasis. CAFs represent a multifunctional subset of PDAC microenvironment and contribute to tumor initiation and progression through ECM deposition and remodeling, as well as the secretion of paracrine factors. Attempts to resolve desmoplasia by targeting CAFs can render an adverse outcome, which is likely due to CAFs heterogeneity. Recent reports describe subsets of CAFs that assume more secretory functions, in addition to the typical myofibroblast phenotype. Here, we review the literature and describe the relationship between CAFs and inflammation and the role of the secretory-CAFs in PDAC.
Collapse
Affiliation(s)
- Mohammad Awaji
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985845 UNMC, Omaha, NE 68198-5845, USA.
- Department of Pathology and Laboratory Medicine, King Fahad Specialist Hospital-Dammam, Dammam 31444, Saudi Arabia.
| | - Rakesh K Singh
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985845 UNMC, Omaha, NE 68198-5845, USA.
| |
Collapse
|
53
|
POLE Score: a comprehensive profiling of programmed death 1 ligand 1 expression in pancreatic ductal adenocarcinoma. Oncotarget 2019; 10:1572-1588. [PMID: 30899426 PMCID: PMC6422186 DOI: 10.18632/oncotarget.26705] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 01/19/2019] [Indexed: 01/19/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) being characterized by a pronounced stromal compartment is commonly diagnosed at an advanced stage limiting curative treatment options. Although therapeutical targeting of immune checkpoint regulators like programmed death 1 ligand 1 (PD-L1) represent a promising approach that substantially improved survival of several highly aggressive malignancies, convincing indicators for response prediction are still lacking for PDAC which might be attributed to the insufficient characterization of PD-L1 status. Therefore, we investigated PD-L1 expression by immunohistochemistry in a well characterized cohort of 59 PDAC and 18 peritumoral tissues. Despite the histopathological homogeneity within our cohort, tumor tissues exhibited a great heterogeneity regarding PD-L1 expression. Considering distinct PD-L1 expression patterns, we established the novel POLE Score that incorporates overall PD-L1 expression (P), cellular Origin of PD-L1 (O), PD-L1 level in tumor-associated Lymph follicles (L) and Enumerated local PD-L1 distribution (E). We show that tumoral PD-L1 expression is higher compared to peritumoral areas. Furthermore, POLE Score parameters correlated with overall survival, tumor grade, Ki67 status, local proximity of tumor cells and particular stroma composition. For the first time, we demonstrate that PD-L1 is mostly expressed by stroma and rarely by tumor cells in PDAC. Moreover, our in situ analyses on serial tissue sections and in vitro data suggest that PD-L1 is prominently expressed by tumor-associated macrophages. In conclusion, POLE Score represents a comprehensive characterization of PD-L1 expression in tumor and stroma compartment and might provide the basis for improved patient stratification in future clinical trials on PD-1/PD-L1 targeting therapies in PDAC.
Collapse
|
54
|
Satyananda V, Gupta R, Hari DM, Yeh J, Chen KT. Advances in Translational Research and Clinical Care in Pancreatic Cancer: Where Are We Headed? Gastroenterol Res Pract 2019; 2019:7690528. [PMID: 30863442 PMCID: PMC6378762 DOI: 10.1155/2019/7690528] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 01/08/2019] [Indexed: 02/06/2023] Open
Abstract
While significant advances have been made in the treatment of many different solid tumors, pancreatic cancer remains a glaring exception. Overall 5-year survival rates for pancreatic cancer remain in the single digits. While newer chemotherapy regimens such as FOLFIRINOX and nab-paclitaxel/gemcitabine have demonstrated modest improvement in survival benefit for metastatic disease and have improved the resectability rates of previously borderline or locally advanced tumors, clinically significant improvements from immunotherapy and targeted therapy remain to be demonstrated. Regardless, a wealth of basic science research in pancreatic cancer has been directed at understanding its aggressive biology and its resistance to therapy. We present a brief summary of key areas of laboratory research and its translation to clinical care.
Collapse
Affiliation(s)
- Vikas Satyananda
- Division of Surgical Oncology, Department of Surgery, Harbor-UCLA Medical Center, USA
| | - Rohan Gupta
- Division of Medical Oncology, Department of Medicine, Harbor-UCLA Medical Center, USA
| | - Danielle M. Hari
- Division of Surgical Oncology, Department of Surgery, Harbor-UCLA Medical Center, USA
| | - James Yeh
- Division of Medical Oncology, Department of Medicine, Harbor-UCLA Medical Center, USA
| | - Kathryn T. Chen
- Division of Surgical Oncology, Department of Surgery, Harbor-UCLA Medical Center, USA
| |
Collapse
|
55
|
Uzunparmak B, Sahin IH. Pancreatic cancer microenvironment: a current dilemma. Clin Transl Med 2019; 8:2. [PMID: 30645701 PMCID: PMC6333596 DOI: 10.1186/s40169-019-0221-1] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Accepted: 01/09/2019] [Indexed: 12/13/2022] Open
Abstract
Pancreatic cancer is one of the leading causes of cancer-related death in the United States and survival outcomes remain dismal despite significant advances in molecular diagnostics and therapeutics in clinical practice. The microenvironment of pancreatic cancer carries unique features with increased desmoplastic reaction and is infiltrated by regulatory T cells and myeloid-derived suppressor cells which negatively impact the effector immune cells. Current evidence suggests that stellate cell-induced hypovascular stroma may have direct effects on aggressive behavior of pancreatic cancer. Preclinical studies suggested improvement in drug delivery to cancer cells with stroma modifying agents. However these findings so far have not been confirmed in clinical trials. In this article, we elaborate current-state-of-the science of the pancreatic cancer microenvironment and its impact on molecular behavior of cancer cells, chemotherapy resistance and druggability of stroma elements in combination with other agents to enhance the efficacy of therapeutic approaches.
Collapse
Affiliation(s)
| | - Ibrahim Halil Sahin
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, USA.
| |
Collapse
|
56
|
Young K, Hughes DJ, Cunningham D, Starling N. Immunotherapy and pancreatic cancer: unique challenges and potential opportunities. Ther Adv Med Oncol 2018; 10:1758835918816281. [PMID: 30574212 PMCID: PMC6299311 DOI: 10.1177/1758835918816281] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 10/31/2018] [Indexed: 12/13/2022] Open
Abstract
Despite decades of research, pancreatic ductal adenocarcinoma (PDAC) continues to have the worst 5-year survival of any malignancy. With 338,000 new cases diagnosed and over 300,000 deaths per year globally there is an urgent unmet need to improve the therapeutic options available. Novel immunotherapies have shown promising results across multiple solid tumours, in a number of cases surpassing chemotherapy as a first-line therapeutic option. However, to date, trials of single-agent immunotherapies in PDAC have been disappointing and PDAC has been labelled as a nonimmunogenic cancer. This lack of response may in part be attributed to PDAC’s unique tumour microenvironment (TME), consisting of a dense fibrotic stroma and a scarcity of tumour infiltrating lymphocytes. However, as our understanding of the PDAC TME evolves, it is becoming apparent that the problem is not simply the immune system failing to recognize the cancer. There is a highly complex interplay between stromal signals, the immune system and tumour cells, at times possibly restraining tumour growth and at others supporting growth and metastasis. Understanding this complexity will enable the development of rational combinations with immunotherapy, priming the TME to offer immunotherapy the best chance of success. This review seeks to describe the unique challenges of the PDAC TME, the potential opportunities it may afford and the trials in progress capitalizing on recent insights in this area.
Collapse
Affiliation(s)
- Kate Young
- The Royal Marsden NHS Foundation Trust, Royal Marsden Hospital, London, UK
| | - Daniel J Hughes
- The Royal Marsden NHS Foundation Trust, Royal Marsden Hospital, London, UK
| | - David Cunningham
- The Royal Marsden NHS Foundation Trust, Royal Marsden Hospital, London, UK
| | - Naureen Starling
- Consultant Medical Oncologist, The Royal Marsden NHS Foundation Trust, 203 Fulham Road, London SW3 6JJ, UK
| |
Collapse
|
57
|
Ormanns S. [Personalized cancer medicine : Biomarkers for molecular therapy stratification in pancreatic carcinoma]. DER PATHOLOGE 2018; 39:221-224. [PMID: 30361776 DOI: 10.1007/s00292-018-0539-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Ductal adenocarcinoma of the pancreas has a very poor prognosis and a rising incidence. Even after curative intent resection, which is possible in a minority of patients, most patients relapse, whereas the majority is diagnosed with inoperable or metastatic disease. That's why palliative systemic chemotherapy is the current therapeutic mainstay. Biomarker-based tumor characterization could identify potential therapy targets and enable a personalized cancer medicine. Although potentially targetable alterations occur at very low frequencies, the possible impact on patient outcome can be significant. This article summarizes some of the contributions to these aspects and gives an outlook on their clinical meaning.
Collapse
Affiliation(s)
- S Ormanns
- Pathologisches Institut, Medizinische Fakultät, Ludwig-Maximilians-Universität München, Thalkirchner Straße 36, 80337, München, Deutschland.
| |
Collapse
|
58
|
Yi G, Guo S, Liu W, Wang H, Liu R, Tsun A, Jin G, Li B. Identification and functional analysis of heterogeneous FOXP3 + Treg cell subpopulations in human pancreatic ductal adenocarcinoma. Sci Bull (Beijing) 2018; 63:972-981. [PMID: 36658893 DOI: 10.1016/j.scib.2018.05.028] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 05/01/2018] [Accepted: 05/17/2018] [Indexed: 01/21/2023]
Abstract
CD4+ CD25+ regulatory T (Treg) cells express the transcription factor FOXP3 and play an essential role in preventing autoimmunity. Abundant Treg cell accumulation in tumors and tumor draining lymph nodes (TDLNs) has been reported to correlate with both poor and favorable prognosis in various cancers, which suggests that Tregs may have multiple effects on antitumor immunity. However, the heterogeneity of tumor- and TDLN-infiltrating Treg cells remains unclear. Here we provide heterogeneity analysis of tumor infiltrating human CD4+ Treg cells and their matched adjacent tissues and TDLNs. We defined three different subpopulations of tumor- and TDLN-infiltrating Treg cells by Helios and CCR8 expression in pancreatic ductal adenocarcinoma (PDAC) and confirmed their functional heterogeneity. Helios+ CCR8+ Treg cells with potent suppressor function and limited IL-2 and IFN-γ secretion were identified in tumors and TDLNs. On the contrary, Helios- CCR8- Treg cells have impaired suppressive activity, and elevated expression of pro-inflammatory cytokines. More advanced grades of PDAC have predominantly Helios+ CCR8+ Treg cells and few Helios- CCR8- Treg cells both in tumors and TDLNs that suggests poor prognosis. These data could help further define the role of Treg cells and their functional role in tumors and TDLNs.
Collapse
Affiliation(s)
- Gang Yi
- Shanghai Key Laboratory of Bio-energy Crops, School of Life Science, Shanghai University, Shanghai 200444, China; Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; CAS Center for Excellence in Molecular Cell Science, Unit of Molecular Immunology, Institute Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences Medical School, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shiwei Guo
- Department of General Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Wenyu Liu
- Department of General Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Huan Wang
- Department of General Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Rendong Liu
- Department of General Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Andy Tsun
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; CAS Center for Excellence in Molecular Cell Science, Unit of Molecular Immunology, Institute Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences Medical School, Chinese Academy of Sciences, Shanghai 200031, China
| | - Gang Jin
- Department of General Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China.
| | - Bin Li
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
59
|
Neoptolemos JP, Kleeff J, Michl P, Costello E, Greenhalf W, Palmer DH. Therapeutic developments in pancreatic cancer: current and future perspectives. Nat Rev Gastroenterol Hepatol 2018; 15:333-348. [PMID: 29717230 DOI: 10.1038/s41575-018-0005-x] [Citation(s) in RCA: 785] [Impact Index Per Article: 112.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The overall 5-year survival for pancreatic cancer has changed little over the past few decades, and pancreatic cancer is predicted to be the second leading cause of cancer-related mortality in the next decade in Western countries. The past few years, however, have seen improvements in first-line and second-line palliative therapies and considerable progress in increasing survival with adjuvant treatment. The use of biomarkers to help define treatment and the potential of neoadjuvant therapies also offer opportunities to improve outcomes. This Review brings together information on achievements to date, what is working currently and where successes are likely to be achieved in the future. Furthermore, we address the questions of how we should approach the development of pancreatic cancer treatments, including those for patients with metastatic, locally advanced and borderline resectable pancreatic cancer, as well as for patients with resected tumours. In addition to embracing newer strategies comprising genomics, stromal therapies and immunotherapies, conventional approaches using chemotherapy and radiotherapy still offer considerable prospects for greater traction and synergy with evolving concepts.
Collapse
Affiliation(s)
- John P Neoptolemos
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany.
| | - Jörg Kleeff
- Department of Visceral, Vascular and Endocrine Surgery, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany. .,Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, UK.
| | - Patrick Michl
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Eithne Costello
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - William Greenhalf
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Daniel H Palmer
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| |
Collapse
|
60
|
Liu L, Zhi Q, Shen M, Gong FR, Zhou BP, Lian L, Shen B, Chen K, Duan W, Wu MY, Tao M, Li W. FH535, a β-catenin pathway inhibitor, represses pancreatic cancer xenograft growth and angiogenesis. Oncotarget 2018; 7:47145-47162. [PMID: 27323403 PMCID: PMC5216931 DOI: 10.18632/oncotarget.9975] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 05/17/2016] [Indexed: 12/30/2022] Open
Abstract
The WNT/β-catenin pathway plays an important role in pancreatic cancer carcinogenesis. We evaluated the correlation between aberrant β-catenin pathway activation and the prognosis pancreatic cancer, and the potential of applying the β-catenin pathway inhibitor FH535 to pancreatic cancer treatment. Meta-analysis and immunohistochemistry showed that abnormal β-catenin pathway activation was associated with unfavorable outcome. FH535 repressed pancreatic cancer xenograft growth in vivo. Gene Ontology (GO) analysis of microarray data indicated that target genes responding to FH535 participated in stemness maintenance. Real-time PCR and flow cytometry confirmed that FH535 downregulated CD24 and CD44, pancreatic cancer stem cell (CSC) markers, suggesting FH535 impairs pancreatic CSC stemness. GO analysis of β-catenin chromatin immunoprecipitation sequencing data identified angiogenesis-related gene regulation. Immunohistochemistry showed that higher microvessel density correlated with elevated nuclear β-catenin expression and unfavorable outcome. FH535 repressed the secretion of the proangiogenic cytokines vascular endothelial growth factor (VEGF), interleukin (IL)-6, IL-8, and tumor necrosis factor-α, and also inhibited angiogenesis in vitro and in vivo. Protein and mRNA microarrays revealed that FH535 downregulated the proangiogenic genes ANGPT2, VEGFR3, IFN-γ, PLAUR, THPO, TIMP1, and VEGF. FH535 not only represses pancreatic CSC stemness in vitro, but also remodels the tumor microenvironment by repressing angiogenesis, warranting further clinical investigation.
Collapse
Affiliation(s)
- Lu Liu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qiaoming Zhi
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Meng Shen
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Fei-Ran Gong
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Binhua P Zhou
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA.,Departments of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Lian Lian
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Oncology, Suzhou Xiangcheng People's Hospital, Suzhou, China.,Department of Pathology, Suzhou Xiangcheng People's Hospital, Suzhou, China
| | - Bairong Shen
- Center for Systems Biology, Soochow University, Suzhou, China
| | - Kai Chen
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Weiming Duan
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Meng-Yao Wu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Min Tao
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China.,PREMED Key Laboratory for Precision Medicine, Soochow University, Suzhou, China.,Jiangsu Institute of Clinical Immunology, Suzhou, China.,Institute of Medical Biotechnology, Soochow University, Suzhou, China
| | - Wei Li
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Markey Cancer Center, University of Kentucky, Lexington, KY, USA.,Center for Systems Biology, Soochow University, Suzhou, China.,PREMED Key Laboratory for Precision Medicine, Soochow University, Suzhou, China.,Jiangsu Institute of Clinical Immunology, Suzhou, China
| |
Collapse
|
61
|
Contemporary Management of Localized Resectable Pancreatic Cancer. Cancers (Basel) 2018; 10:cancers10010024. [PMID: 29361690 PMCID: PMC5789374 DOI: 10.3390/cancers10010024] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 01/16/2018] [Accepted: 01/18/2018] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is the third most common cause of cancer deaths in the United States. Surgical resection with negative margins still constitutes the cornerstone of potentially curative therapy, but is possible only in 15–20% of patients at the time of initial diagnosis. Accumulating evidence suggests that the neoadjuvant approach may improve R0 resection rate in localized resectable and borderline resectable diseases, and potentially downstage locally advanced disease to achieve surgical resection, though the impact on survival is to be determined. Despite advancements in the last decade in developing effective combinational chemo-radio therapeutic options, preoperative treatment strategies, and better peri-operative care, pancreatic cancer continues to carry a dismal prognosis in the majority. Prodigious efforts are currently being made in optimizing the neoadjuvant therapy with a better toxicity profile, developing novel agents, imaging techniques, and identification of biomarkers for the disease. Advancement in our understanding of the tumor microenvironment and molecular pathology is urgently needed to facilitate the development of novel targeted and immunotherapies for this setting. In this review, we detail the current literature on contemporary management of resectable, borderline resectable and locally advanced pancreatic cancer with a focus on future directions in the field.
Collapse
|
62
|
Schmitz-Winnenthal FH, Hohmann N, Schmidt T, Podola L, Friedrich T, Lubenau H, Springer M, Wieckowski S, Breiner KM, Mikus G, Büchler MW, Keller AV, Koc R, Springfeld C, Knebel P, Bucur M, Grenacher L, Haefeli WE, Beckhove P. A phase 1 trial extension to assess immunologic efficacy and safety of prime-boost vaccination with VXM01, an oral T cell vaccine against VEGFR2, in patients with advanced pancreatic cancer. Oncoimmunology 2018; 7:e1303584. [PMID: 29632710 DOI: 10.1080/2162402x.2017.1303584] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 02/24/2017] [Accepted: 03/01/2017] [Indexed: 12/17/2022] Open
Abstract
VXM01 is a first-in-kind orally applied tumor vaccine based on live attenuated Salmonella typhi carrying an expression plasmid encoding VEGFR2, an antigen expressed on tumor vasculature and a stable and accessible target for anti-angiogenic intervention. A recent randomized, placebo-controlled, phase I dose-escalation trial in advanced pancreatic cancer patients demonstrated safety, immunogenicity and transient, T-cell response-related anti-angiogenic activity of four priming vaccinations applied within one week. We here evaluated whether monthly boost vaccinations are safe and can sustain increased frequencies of vaccine-specific T cells. Patients with advanced pancreatic cancer were randomly assigned at a ratio of 2:1 to priming with VXM01 followed by up to six monthly boost vaccinations, or placebo treatment. Vaccinations were applied orally at two alternative doses of either 106 colony-forming units (CFU) or 107 CFU, and concomitant treatment with standard-of-care gemcitabine during the priming phase, and any treatment thereafter, was allowed in the study. Immunomonitoring involved interferon-gamma (IFNγ) ELIspot analysis with long overlapping peptides spanning the entire VEGFR2 sequence. A total of 26 patients were treated. Treatment-related adverse events preferentially associated with VXM01 were decreases in lymphocyte numbers in the blood, increased frequencies of neutrophils and diarrhea. Eight out of 16 patients who received at least one boosting vaccination responded with pronounced, i.e. at least 3-fold, increase in VEGFR2-specific T cell response over baseline levels. In the VXM01 vaccination group, VEGFR2-specific T cells peaked preferentially during the boosting phase with an average 4-fold increase over baseline levels. In conclusion, prime/boost vaccination with VXM01 was safe and immunogenic and increased vaccine specific T cell responses compared with placebo treatment.
Collapse
Affiliation(s)
| | - Nicolas Hohmann
- Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Thomas Schmidt
- Department of General, Abdominal and Transplant Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Lilli Podola
- Regensburg Center for Interventional Immunology (RCI), University Hospital Regensburg, Regensburg, Germany.,Medical Oncology, National Center for Tumor Diseases, Heidelberg, Germany
| | - Tobias Friedrich
- Diagnostic and Interventional Radiology, Heidelberg University Hospital, Heidelberg, Germany
| | | | | | | | | | - Gerd Mikus
- Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Markus W Büchler
- Department of General, Abdominal and Transplant Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Ruhan Koc
- Department of General, Abdominal and Transplant Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Phillip Knebel
- Department of General, Abdominal and Transplant Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Mariana Bucur
- Regensburg Center for Interventional Immunology (RCI), University Hospital Regensburg, Regensburg, Germany
| | - Lars Grenacher
- Diagnostic Munich, Diagnostic Prevention and Imaging Center, Munich, Germany
| | - Walter E Haefeli
- Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Philipp Beckhove
- Regensburg Center for Interventional Immunology (RCI), University Hospital Regensburg, Regensburg, Germany.,Medical Oncology, National Center for Tumor Diseases, Heidelberg, Germany
| |
Collapse
|
63
|
Expression of the bitter receptor T2R38 in pancreatic cancer: localization in lipid droplets and activation by a bacteria-derived quorum-sensing molecule. Oncotarget 2017; 7:12623-32. [PMID: 26862855 PMCID: PMC4914309 DOI: 10.18632/oncotarget.7206] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Accepted: 01/24/2016] [Indexed: 01/26/2023] Open
Abstract
T2R38 belongs to the family of bitter receptors and was initially detected in cells of the oral cavity. We now describe expression of T2R38 in tumor cells in patients with pancreatic cancer and in tumor-derived cell lines. T2R38 is localized predominantly intracellular in association with lipid droplets, particularly with the lipid droplet membrane. The receptor can be activated by the bona fide ligand for T2R38, phenylthiourea (PTU), and by N-acetyl-dodecanoyl homoserine (AHL-12), a quorum sensing molecule of Pseudomonas aeruginosa, the latter is the only known natural ligand for T2R38. In response to PTU or AHL-12, key transcription factors are activated including phosphorylation of the MAP kinases p38 and ERK1/2, and upregulation of NFATc1. Moreover, we found increased expression of the multi-drug resistance protein 1 (also known as ABCB1), a transmembrane transporter molecule, participating in shuttling of a plethora of drugs, such as chemotherapeutics or antibiotics. In conclusion, our data indicate a new, additional function of the taste receptor T2R38 beyond sensing ‘bitter’. Moreover, because T2R38 can be stimulated by a bacteria-derived signaling molecule the receptor could link microbiota and cancer.
Collapse
|
64
|
Zhan B, Wen S, Lu J, Shen G, Lin X, Feng J, Huang H. Identification and causes of metabonomic difference between orthotopic and subcutaneous xenograft of pancreatic cancer. Oncotarget 2017; 8:61264-61281. [PMID: 28977862 PMCID: PMC5617422 DOI: 10.18632/oncotarget.18057] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 04/24/2017] [Indexed: 01/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal tumors. However, the methodological differences between orthotopic and subcutaneous xenograft (OX and SX) models will cause confusion in understanding its pathological mechanism and clinical relevance. In this study, SX and OX models were established by implanting Panc-1 and BxPC-3 cell strains under skin and on the pancreas of mice, respectively. The tumor tissue and serum samples were collected for1H NMR spectroscopy followed by univariate and multivariate statistical analyses. As results, no obvious metabonomic difference was demonstrated in serum between the two models, however, the model- and cell strain-specific metabonomic differences were observed in tumor tissues. According to the KEGG analysis, ABC transporters, glycerophospholipid metabolism, purine metabolism and central carbon metabolism were identified to be the most significant components involved in metabonomic differences. Considering the methodological discrepancy in SX and OX models, such differences should be contributed to tumor microenvironment. In general, SX are not equivalent to OX models at molecular level. Subcutaneous transplantation displayed its inherent limitations though it offered a simple, inexpensive, reproducible and quantifiable advantage. And orthotopic transplantation may be favorable to simulate PDAC in patients due to its similar pathogenesis to human pancreatic cancer.
Collapse
Affiliation(s)
- Bohan Zhan
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen 361005, China
| | - Shi Wen
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Jie Lu
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen 361005, China
| | - Guiping Shen
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen 361005, China
| | - Xianchao Lin
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Jianghua Feng
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen 361005, China
| | - Heguang Huang
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
| |
Collapse
|
65
|
Matera R, Saif MW. New therapeutic directions for advanced pancreatic cancer: cell cycle inhibitors, stromal modifiers and conjugated therapies. Expert Opin Emerg Drugs 2017; 22:223-233. [PMID: 28783977 DOI: 10.1080/14728214.2017.1362388] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Pancreatic adenocarcinoma is a devastating malignancy with an extremely poor prognosis. These tumors progress rapidly and somewhat silently with few specific symptoms and are relatively resistant to chemotherapeutic agents. Many agents, including cell cycle inhibitors, are under development for the treatment of this cancer for which there are disappointingly few treatment options. Areas covered: Here we outline the existing approved treatments for advanced pancreatic disease and discuss a range of novel therapies currently under development including cell cycle inhibitors, stromal modifiers and conjugated therapies. We also describe the current state of the pancreatic cancer therapeutics market both past and future. Expert opinion: Despite the recent explosion of novel therapies with an array of unique targets, the core treatment of pancreatic cancer still with traditional cytotoxic agents with a few exceptions. However, as these novel treatments move through the pipeline, we are hopeful that there will soon be a number of effective options for patients with advanced pancreatic cancer.
Collapse
Affiliation(s)
- Robert Matera
- a Department of Hematology and Oncology , Tufts University School of Medicine , Boston , MA , USA
| | - Muhammad Wasif Saif
- a Department of Hematology and Oncology , Tufts University School of Medicine , Boston , MA , USA
| |
Collapse
|
66
|
Zeleniak AE, Huang W, Brinkman MK, Fishel ML, Hill R. Loss of MTSS1 results in increased metastatic potential in pancreatic cancer. Oncotarget 2017; 8:16473-16487. [PMID: 28146435 PMCID: PMC5369978 DOI: 10.18632/oncotarget.14869] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 01/19/2017] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a 5-year survival rate of 7%. This dismal prognosis is largely due to the inability to diagnose the disease before metastasis occurs. Tumor cell dissemination occurs early in PDAC. While it is known that inflammation facilitates this process, the underlying mechanisms responsible for this progression have not been fully characterized. Here, we functionally test the role of metastasis suppressor 1 (MTSS1) in PDAC. Despite evidence showing that MTSS1 could be important for regulating metastasis in many different cancers, its function in PDAC has not been studied. Here, we show that loss of MTSS1 leads to increased invasion and migration in PDAC cell lines. Moreover, PDAC cells treated with cancer-associated fibroblast-conditioned media also have increased metastatic potential, which is augmented by loss of MTSS1. Finally, overexpression of MTSS1 in PDAC cell lines leads to a loss of migratory potential in vitro and an increase in overall survival in vivo. Collectively, our data provide insight into an important role for MTSS1 in suppressing tumor cell invasion and migration driven by the tumor microenvironment and suggest that therapeutic strategies aimed at increasing MTSS1 levels may effectively slow the development of metastatic lesions, increasing survival of patients with PDAC.
Collapse
Affiliation(s)
- Ann E Zeleniak
- Integrated Biomedical Sciences Program, University of Notre Dame, South Bend, Indiana, USA.,Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana, USA
| | - Wei Huang
- Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana, USA.,Department of Biological Sciences, University of Notre Dame, South Bend, Indiana, USA
| | - Mary K Brinkman
- Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana, USA.,Department of Biological Sciences, University of Notre Dame, South Bend, Indiana, USA
| | - Melissa L Fishel
- Indiana University School of Medicine, Department of Pharmacology and Toxicology, Indianapolis, Indiana, USA.,Indiana University School of Medicine, Department of Pediatrics, Wells Center for Pediatric Research, Indianapolis, Indiana, USA.,Pancreatic Cancer Signature Center, Indianapolis, Indiana, USA
| | - Reginald Hill
- Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana, USA.,Department of Biological Sciences, University of Notre Dame, South Bend, Indiana, USA
| |
Collapse
|
67
|
Pancreatic adenocarcinoma response to chemotherapy enhanced with non-invasive radio frequency evaluated via an integrated experimental/computational approach. Sci Rep 2017; 7:3437. [PMID: 28611425 PMCID: PMC5469743 DOI: 10.1038/s41598-017-03040-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 04/21/2017] [Indexed: 12/13/2022] Open
Abstract
Although chemotherapy combined with radiofrequency exposure has shown promise in cancer treatment by coupling drug cytotoxicity with thermal ablation or thermally-induced cytotoxicity, limited access of the drug to tumor loci in hypo-vascularized lesions has hampered clinical application. We recently showed that high-intensity short-wave capacitively coupled radiofrequency (RF) electric-fields may reach inaccessible targets in vivo. This non-invasive RF combined with gemcitabine (Gem) chemotherapy enhanced drug uptake and effect in pancreatic adenocarcinoma (PDAC), notorious for having poor response and limited therapeutic options, but without inducing thermal injury. We hypothesize that the enhanced cytotoxicity derives from RF-facilitated drug transport in the tumor microenvironment. We propose an integrated experimental/computational approach to evaluate chemotherapeutic response combined with RF-induced phenotypic changes in tissue with impaired transport. Results show that RF facilitates diffusive transport in 3D cell cultures representing hypo-vascularized lesions, enhancing drug uptake and effect. Computational modeling evaluates drug vascular extravasation and diffusive transport as key RF-modulated parameters, with transport being dominant. Assessment of hypothetical schedules following current clinical protocol for Stage-IV PDAC suggests that unresponsive lesions may be growth-restrained when exposed to Gem plus RF. Comparison of these projections to experiments in vivo indicates that synergy may result from RF-induced cell phenotypic changes enhancing drug transport and cytotoxicity, thus providing a potential baseline for clinically-focused evaluation.
Collapse
|
68
|
Imai D, Yoshizumi T, Okano S, Uchiyama H, Ikegami T, Harimoto N, Itoh S, Soejima Y, Aishima S, Oda Y, Maehara Y. The prognostic impact of programmed cell death ligand 1 and human leukocyte antigen class I in pancreatic cancer. Cancer Med 2017; 6:1614-1626. [PMID: 28602029 PMCID: PMC5504334 DOI: 10.1002/cam4.1087] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 04/02/2017] [Accepted: 04/04/2017] [Indexed: 12/20/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) is associated with an immunosuppressive tumor‐microenvironment (TME) that supports the growth of tumors and mediates tumors enabling evasion of the immune system. Expression of programmed cell death ligand 1 (PD‐L1) and loss of human leukocyte antigen (HLA) class I on tumor cells are methods by which tumors escape immunosurveillance. We examined immune cell infiltration, the expression of PD‐L1 and HLA class I by PDA cells, and the correlation between these immunological factors and clinical prognosis. PDA samples from 36 patients were analyzed for HLA class I, HLA‐DR, PD‐L1, PD‐1, CD4, CD8, CD56, CD68, and FoxP3 expression by immunohistochemistry. The correlations between the expression of HLA class I, HLA‐DR, PD‐L1 or PD‐1 and the pattern of tumor infiltrating immune cells or the patients’ prognosis were assessed. PD‐L1 expression correlated with tumor infiltration by CD68+ and FoxP3+ cells. Low HLA class I expression was an only risk factor for poor survival. PD‐L1 negative and HLA class I high‐expressing PDA was significantly associated with higher numbers of infiltrating CD8+ T cells in the TME, and a better prognosis. Evaluation of both PD‐L1 and HLA class I expression by PDA may be a good predictor of prognosis for patients. HLA class I expression by tumor cells should be evaluated when selecting PDA patients who may be eligible for treatment with PD‐1/PD‐L1 immune checkpoint blockade therapies.
Collapse
Affiliation(s)
- Daisuke Imai
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomoharu Yoshizumi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shinji Okano
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of General surgery, Digestive Disease and Surgery institute, Cleveland Clinic, Cleveland, USA
| | - Hideaki Uchiyama
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toru Ikegami
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Norifumi Harimoto
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shinji Itoh
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuji Soejima
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shinichi Aishima
- Department of Pathology and Microbiology, Saga Medical School, Faculty of Medicine, Saga University, Saga, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Pathological sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshihiko Maehara
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
69
|
A Preliminary Report: Radical Surgery and Stem Cell Transplantation for the Treatment of Patients With Pancreatic Cancer. J Immunother 2017; 40:132-139. [PMID: 28338506 DOI: 10.1097/cji.0000000000000164] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
We examined the immunologic effects of allogeneic hematopoietic stem cell transplantation (HSCT) in the treatment of pancreatic ductal adenocarcinoma, a deadly disease with a median survival of 24 months for resected tumors and a 5-year survival rate of 6%. After adjuvant chemotherapy, 2 patients with resected pancreatic ductal adenocarcinoma underwent HSCT with HLA-identical sibling donors. Comparable patients who underwent radical surgery, but did not have a donor, served as controls (n=6). Both patients developed humoral and cellular (ie, HLA-A*01:01-restricted) immune responses directed against 2 novel tumor-associated antigens (TAAs), INO80E and UCLH3 after HSCT. Both TAAs were highly expressed in the original tumor tissue suggesting that HSCT promoted a clinically relevant, long-lasting cellular immune response. In contrast to untreated controls, who succumbed to progressive disease, both patients are tumor-free 9 years after diagnosis. Radical surgery combined with HSCT may cure pancreatic adenocarcinoma and change the cellular immune repertoire capable of responding to clinically and biologically relevant TAAs.
Collapse
|
70
|
Knudsen ES, Vail P, Balaji U, Ngo H, Botros IW, Makarov V, Riaz N, Balachandran V, Leach S, Thompson DM, Chan TA, Witkiewicz AK. Stratification of Pancreatic Ductal Adenocarcinoma: Combinatorial Genetic, Stromal, and Immunologic Markers. Clin Cancer Res 2017; 23:4429-4440. [PMID: 28348045 DOI: 10.1158/1078-0432.ccr-17-0162] [Citation(s) in RCA: 136] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 02/23/2017] [Accepted: 03/14/2017] [Indexed: 02/06/2023]
Abstract
Purpose: Pancreatic ductal adenocarcinoma (PDAC) is associated with an immunosuppressive milieu that supports immune system evasion and disease progression. Here, we interrogated genetic, stromal, and immunologic features of PDAC to delineate impact on prognosis and means to more effectively employ immunotherapy.Experimental Design: A cohort of 109 PDAC cases annotated for overall survival was utilized as a primary discovery cohort. Gene expression analysis defined immunologic subtypes of PDAC that were confirmed in the Cancer Genome Atlas dataset. Stromal and metabolic characteristics of PDAC cases were evaluated by histologic analysis and immunostaining. Enumeration of lymphocytes, as well as staining for CD8, FOXP3, CD68, CD163, PDL1, and CTLA4 characterized immune infiltrate. Neoantigens were determined by analysis of whole-exome sequencing data. Random-forest clustering was employed to define multimarker subtypes, with univariate and multivariate analyses interrogating prognostic significance.Results: PDAC cases exhibited distinct stromal phenotypes that were associated with prognosis, glycolytic and hypoxic biomarkers, and immune infiltrate composition. Immune infiltrate was diverse among PDAC cases and enrichment for M2 macrophages and select immune checkpoints regulators were specifically associated with survival. Composite analysis with neoantigen burden, immunologic, and stromal features defined novel subtypes of PDAC that could have bearing on sensitivity to immunologic therapy approaches. In addition, a subtype with low levels of neoantigens and minimal lymphocyte infiltrate was associated with improved overall survival.Conclusions: The mutational burden of PDAC is associated with distinct immunosuppressive mechanisms that are conditioned by the tumor stromal environment. The defined subtypes have significance for utilizing immunotherapy in the treatment of PDAC. Clin Cancer Res; 23(15); 4429-40. ©2017 AACR.
Collapse
Affiliation(s)
- Erik S Knudsen
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas.,University of Arizona Cancer Center, University of Arizona, Tucson, Arizona.,Department of Medicine, University of Arizona, Tucson, Arizona
| | - Paris Vail
- Department of Medicine, University of Arizona, Tucson, Arizona
| | - Uthra Balaji
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Hoai Ngo
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas
| | | | - Vladimir Makarov
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.,Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nadeem Riaz
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.,Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York.,Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vinod Balachandran
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Steven Leach
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Timothy A Chan
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.,Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York.,Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Agnieszka K Witkiewicz
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas. .,University of Arizona Cancer Center, University of Arizona, Tucson, Arizona.,Department of Medicine, University of Arizona, Tucson, Arizona.,Department of Pathology, University of Arizona, Tucson, Arizona
| |
Collapse
|
71
|
Kipps E, Young K, Starling N. Liposomal irinotecan in gemcitabine-refractory metastatic pancreatic cancer: efficacy, safety and place in therapy. Ther Adv Med Oncol 2017; 9:159-170. [PMID: 28344661 PMCID: PMC5349428 DOI: 10.1177/1758834016688816] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal disease. The majority of patients are diagnosed with locally advanced or metastatic disease with a prognosis of short months. Therapeutic options are limited and until recently, there was no standard second-line chemotherapy option. Liposomal constructs have been engineered to encapsulate chemotherapy thereby preventing premature metabolism, improving distribution and minimizing toxicity. Favourable preclinical data on liposomal irinotecan and early phase trials, led to a recently published phase III trial of liposomal irinotecan in combination with fluorouracil and folinic acid in patients with metastatic PDAC, who progressed after gemcitabine-based chemotherapy. As a direct result, the United States Food and Drug Administration (FDA) and European Medicines Agency (EMA) have approved the use of liposomal irinotecan in this setting. However, first-line treatment options for this disease now include the combination regimen, FOLFIRINOX, in patients with good performance status, and the role of second-line combination treatment with liposomal irinotecan in this setting is unclear. Recent advances have changed the therapeutic landscape, as clinicians are now able to choose a sequential approach to treatment tailored to the individual patient characteristics. This article reviews current treatment options for metastatic PDAC and focuses on the efficacy, safety and place in therapy of liposomal irinotecan.
Collapse
Affiliation(s)
- Emma Kipps
- Royal Marsden NHS Foundation Trust Ringgold standard institution - Department of Gastrointestinal Oncology, London, UK
| | - Kate Young
- Royal Marsden NHS Foundation Trust Ringgold standard institution - Department of Gastrointestinal Oncology, London, UK
| | - Naureen Starling
- Royal Marsden NHS Foundation Trust Ringgold standard institution - Department of Gastrointestinal Oncology, London, UK
| |
Collapse
|
72
|
Smigiel JM, Parameswaran N, Jackson MW. Potent EMT and CSC Phenotypes Are Induced By Oncostatin-M in Pancreatic Cancer. Mol Cancer Res 2017; 15:478-488. [PMID: 28053127 DOI: 10.1158/1541-7786.mcr-16-0337] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 11/21/2016] [Accepted: 11/30/2016] [Indexed: 12/19/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is referred to as a silent killer due to the lack of clear symptoms, a lack of early detection methods, and a high frequency of metastasis at diagnosis. In addition, pancreatic cancer is remarkably resistant to chemotherapy, and clinical treatment options remain limited. The tumor microenvironment (TME) and associated factors are important determinants of metastatic capacity and drug resistance. Here, oncostatin M (OSM), an IL6 cytokine family member, was identified as an important driver of mesenchymal and cancer stem cell (CSC) phenotypes. Furthermore, the generation of cells that harbor mesenchymal/CSC properties following OSM exposure resulted in enhanced tumorigenicity, increased metastasis, and resistance to gemcitabine. OSM induced the expression of ZEB1, Snail (SNAI1), and OSM receptor (OSMR), engaging a positive feedback loop to potentiate the mesenchymal/CSC program. Suppression of JAK1/2 by ruxolitinib prevented STAT3-mediated transcription of ZEB1, SNAI1 and OSMR, as well as the emergence of a mesenchymal/CSC phenotype. Likewise, ZEB1 silencing, by shRNA-mediated knockdown, in OSM-driven mesenchymal/CSC reverted the phenotype back to an epithelial/non-CSC state. Importantly, the generation of cells with mesenchymal/CSC properties was unique to OSM, and not observed following IL6 exposure, implicating OSMR and downstream effector signaling as a distinct target in PDAC. Overall, these data demonstrate the capacity of OSM to regulate an epithelial-mesenchymal transition (EMT)/CSC plasticity program that promotes tumorigenic properties.Implications: Therapeutic targeting the OSM/OSMR axis within the TME may prevent or reverse the aggressive mesenchymal and CSC phenotypes associated with poor outcomes in patients with PDAC. Mol Cancer Res; 15(4); 478-88. ©2017 AACR.
Collapse
Affiliation(s)
- Jacob M Smigiel
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | | | - Mark W Jackson
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio. .,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
73
|
Smigiel JM, Parameswaran N, Jackson MW, Gold DV, Ananthanarayan V, Munshi HG, Mengele K, Schmitt M, Höfler H, Aubele M. Potent EMT and CSC Phenotypes Are Induced By Oncostatin-M in Pancreatic Cancer. Mol Cancer Res 2017. [PMID: 28053127 DOI: 10.1158/1541-7786] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is referred to as a silent killer due to the lack of clear symptoms, a lack of early detection methods, and a high frequency of metastasis at diagnosis. In addition, pancreatic cancer is remarkably resistant to chemotherapy, and clinical treatment options remain limited. The tumor microenvironment (TME) and associated factors are important determinants of metastatic capacity and drug resistance. Here, oncostatin M (OSM), an IL6 cytokine family member, was identified as an important driver of mesenchymal and cancer stem cell (CSC) phenotypes. Furthermore, the generation of cells that harbor mesenchymal/CSC properties following OSM exposure resulted in enhanced tumorigenicity, increased metastasis, and resistance to gemcitabine. OSM induced the expression of ZEB1, Snail (SNAI1), and OSM receptor (OSMR), engaging a positive feedback loop to potentiate the mesenchymal/CSC program. Suppression of JAK1/2 by ruxolitinib prevented STAT3-mediated transcription of ZEB1, SNAI1 and OSMR, as well as the emergence of a mesenchymal/CSC phenotype. Likewise, ZEB1 silencing, by shRNA-mediated knockdown, in OSM-driven mesenchymal/CSC reverted the phenotype back to an epithelial/non-CSC state. Importantly, the generation of cells with mesenchymal/CSC properties was unique to OSM, and not observed following IL6 exposure, implicating OSMR and downstream effector signaling as a distinct target in PDAC. Overall, these data demonstrate the capacity of OSM to regulate an epithelial-mesenchymal transition (EMT)/CSC plasticity program that promotes tumorigenic properties.Implications: Therapeutic targeting the OSM/OSMR axis within the TME may prevent or reverse the aggressive mesenchymal and CSC phenotypes associated with poor outcomes in patients with PDAC. Mol Cancer Res; 15(4); 478-88. ©2017 AACR.
Collapse
Affiliation(s)
- Jacob M Smigiel
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | | | - Mark W Jackson
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio. .,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | | | | | | | | | | | | | | |
Collapse
|
74
|
Jorand R, Biswas S, Wakefield DL, Tobin SJ, Golfetto O, Hilton K, Ko M, Ramos JW, Small AR, Chu P, Singh G, Jovanovic-Talisman T. Molecular signatures of mu opioid receptor and somatostatin receptor 2 in pancreatic cancer. Mol Biol Cell 2016; 27:3659-3672. [PMID: 27682590 PMCID: PMC5221597 DOI: 10.1091/mbc.e16-06-0427] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 09/20/2016] [Indexed: 12/21/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), a particularly aggressive malignancy, has been linked to atypical levels, certain mutations, and aberrant signaling of G-protein-coupled receptors (GPCRs). GPCRs have been challenging to target in cancer because they organize into complex networks in tumor cells. To dissect such networks with nanometer-scale precision, here we combine traditional biochemical approaches with superresolution microscopy methods. A novel interaction specific to PDAC is identified between mu opioid receptor (MOR) and somatostatin receptor 2 (SSTR2). Although MOR and SSTR2 did not colocalize in healthy pancreatic cells or matching healthy patient tissues, the pair did significantly colocalize in pancreatic cancer cells, multicellular tumor spheroids, and cancerous patient tissues. Moreover, this association in pancreatic cancer cells correlated with functional cross-talk and increased metastatic potential of cells. Coactivation of MOR and SSTR2 in PDAC cells led to increased expression of mesenchymal markers and decreased expression of an epithelial marker. Together these results suggest that the MOR-SSTR2 heteromer may constitute a novel therapeutic target for PDAC.
Collapse
Affiliation(s)
- Raphael Jorand
- Department of Molecular Medicine, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010
| | - Sunetra Biswas
- Department of Molecular Medicine, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010
| | - Devin L Wakefield
- Department of Molecular Medicine, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010
| | - Steven J Tobin
- Department of Molecular Medicine, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010
| | - Ottavia Golfetto
- Department of Molecular Medicine, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010
| | - Kelsey Hilton
- Department of Molecular Medicine, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010
| | - Michelle Ko
- Department of Molecular Medicine, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010
| | - Joe W Ramos
- Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, HI 96813
| | - Alexander R Small
- Department of Physics and Astronomy, California State Polytechnic University, Pomona, CA 91768
| | - Peiguo Chu
- Department of Pathology, City of Hope Comprehensive Cancer Center, Duarte, CA 91010
| | - Gagandeep Singh
- Division of Surgical Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA 91010
| | - Tijana Jovanovic-Talisman
- Department of Molecular Medicine, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010
| |
Collapse
|
75
|
Richards KE, Zeleniak AE, Fishel ML, Wu J, Littlepage LE, Hill R. Cancer-associated fibroblast exosomes regulate survival and proliferation of pancreatic cancer cells. Oncogene 2016; 36:1770-1778. [PMID: 27669441 PMCID: PMC5366272 DOI: 10.1038/onc.2016.353] [Citation(s) in RCA: 579] [Impact Index Per Article: 64.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 07/30/2016] [Accepted: 08/15/2016] [Indexed: 02/07/2023]
Abstract
Cancer associated fibroblasts (CAFs) comprise the majority of the tumor bulk of pancreatic adenocarcinomas (PDACs). Current efforts to eradicate these tumors focus predominantly on targeting the proliferation of rapidly growing cancer epithelial cells. We know that this is largely ineffective with resistance arising in most tumors following exposure to chemotherapy. Despite the long-standing recognition of the prominence of CAFs in PDAC, the effect of chemotherapy on CAFs and how they may contribute to drug resistance in neighboring cancer cells is not well characterized. Here we show that CAFs exposed to chemotherapy play an active role in regulating the survival and proliferation of cancer cells. We found that CAFs are intrinsically resistant to gemcitabine, the chemotherapeutic standard of care for PDAC. Further, CAFs exposed to gemcitabine significantly increase the release of extracellular vesicles called exosomes. These exosomes increased chemoresistance-inducing factor, Snail, in recipient epithelial cells and promote proliferation and drug resistance. Finally, treatment of gemcitabine-exposed CAFs with an inhibitor of exosome release, GW4869, significantly reduces survival in co-cultured epithelial cells, signifying an important role of CAF exosomes in chemotherapeutic drug resistance. Collectively, these findings show the potential for exosome inhibitors as treatment options alongside chemotherapy for overcoming PDAC chemoresistance.
Collapse
Affiliation(s)
- K E Richards
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA.,Department of Biological Sciences, Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, USA
| | - A E Zeleniak
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA.,Department of Biological Sciences, Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, USA.,Integrated Biomedical Sciences Program, University of Notre Dame, South Bend, IN, USA
| | - M L Fishel
- Department of Pediatrics, Indiana University School of Medicine, Indiana University, Indianapolis, IN, USA.,Department of Pharmacology, Indiana University, Indianapolis, IN, USA.,Indiana University Simon Cancer Center, Indianapolis, IN, USA.,Department of Toxicology, Indiana University, Indianapolis, IN, USA
| | - J Wu
- Department of Biological Sciences, Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, USA.,Department of Chemistry, University of Notre Dame, Notre Dame, IN, USA.,Department of Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - L E Littlepage
- Department of Biological Sciences, Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, USA.,Integrated Biomedical Sciences Program, University of Notre Dame, South Bend, IN, USA.,Indiana University Simon Cancer Center, Indianapolis, IN, USA.,Department of Chemistry, University of Notre Dame, Notre Dame, IN, USA.,Department of Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - R Hill
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA.,Department of Biological Sciences, Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, USA.,Integrated Biomedical Sciences Program, University of Notre Dame, South Bend, IN, USA.,Indiana University Simon Cancer Center, Indianapolis, IN, USA
| |
Collapse
|
76
|
Zaccagnino A, Pilarsky C, Tawfik D, Sebens S, Trauzold A, Novak I, Schwab A, Kalthoff H. In silico analysis of the transportome in human pancreatic ductal adenocarcinoma. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2016; 45:749-763. [PMID: 27652669 DOI: 10.1007/s00249-016-1171-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 07/18/2016] [Accepted: 08/30/2016] [Indexed: 12/14/2022]
Abstract
The altered expression and/or activity of ion channels and transporters (transportome) have been associated with malignant behavior of cancer cells and were proposed to be a hallmark of cancer. However, the impact of altered transportome in epithelial cancers, such as pancreatic ductal adenocarcinoma (PDAC), as well as its pathophysiological consequences, still remains unclear. Here, we report the in silico analysis of 840 transportome genes in PDAC patients' tissues. Our study was focused on the transportome changes and their correlation with functional and behavioral responses in PDAC tumor and stromal compartments. The dysregulated gene expression datasets were filtered using a cut-off of fold-change values ≤-2 or ≥2 (adjusted p value ≤0.05). The dysregulated transportome genes were clearly associated with impaired physiological secretory mechanisms and/or pH regulation, control of cell volume, and cell polarity. Additionally, some down-regulated transportome genes were found to be closely linked to epithelial cell differentiation. Furthermore, the observed decrease in genes coding for calcium and chloride transport might be a mechanism for evasion of apoptosis. In conclusion, the current work provides a comprehensive overview of the altered transportome expression and its association with predicted PDAC malignancy with special focus on the epithelial compartment.
Collapse
Affiliation(s)
- A Zaccagnino
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, UKSH, Campus Kiel, Arnold-Heller-Str. 3, 24105, Kiel, Germany.
| | - C Pilarsky
- Department of Surgery, University Clinic, Krankenhausstr. 12, 91054, Erlangen, Germany
| | - D Tawfik
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, UKSH, Campus Kiel, Arnold-Heller-Str. 3, 24105, Kiel, Germany
| | - S Sebens
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, UKSH, Campus Kiel, Arnold-Heller-Str. 3, 24105, Kiel, Germany
| | - A Trauzold
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, UKSH, Campus Kiel, Arnold-Heller-Str. 3, 24105, Kiel, Germany
| | - I Novak
- Section for Cell Biology and Physiology, Department of Biology, Faculty of Science, University of Copenhagen, Universitetsparken 13, 2100, Copenhagen, Denmark
| | - A Schwab
- Institute of Physiology II, University of Muenster, Robert-Koch-Str. 27 b, 48149, Muenster, Germany
| | - H Kalthoff
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, UKSH, Campus Kiel, Arnold-Heller-Str. 3, 24105, Kiel, Germany.
| |
Collapse
|
77
|
Hutcheson J, Balaji U, Porembka MR, Wachsmann MB, McCue PA, Knudsen ES, Witkiewicz AK. Immunologic and Metabolic Features of Pancreatic Ductal Adenocarcinoma Define Prognostic Subtypes of Disease. Clin Cancer Res 2016; 22:3606-17. [PMID: 26858311 PMCID: PMC4947442 DOI: 10.1158/1078-0432.ccr-15-1883] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 01/25/2016] [Indexed: 12/20/2022]
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDA) is associated with an immunosuppressive microenvironment that supports the growth of the malignancy as well as immune system evasion. Here we examine markers of immunosuppression in PDA within the context of the glycolytic tumor microenvironment, their interrelationship with tumor biology and association with overall survival. EXPERIMENTAL DESIGN We utilized tissue microarrays consisting of 223 PDA patients annotated for clinical stage, tumor size, lymph node involvement, and survival. Expression of CD163, FoxP3, PD-L1, and MCT4 was assessed by IHC and statistical associations were evaluated by univariate and multivariate analysis. Multimarker subtypes were defined by random forest analysis. Mechanistic interactions were evaluated using PDA cell lines and models for myeloid differentiation. RESULTS PDA exhibits discrete expression of CD163, FoxP3, and PD-L1 with modest individual significance. However, combined low expression of these markers was associated with improved prognosis (P = 0.02). PDA tumor cells altered macrophage phenotype and function, which supported enhanced invasiveness in cell-based models. Lactate efflux mediated by MCT4 was associated with, and required for, the selective conversion of myeloid cells. Correspondingly, MCT4 expression correlated with immune markers in PDA cases, and increased the significance of prognostic subtypes (P = 0.002). CONCLUSIONS There exists a complex interplay between PDA tumor cells and the host immune system wherein immunosuppression is associated with negative outcome. MCT4 expression, representative of the glycolytic state of PDA, contributes to the phenotypic conversion of myeloid cells. Thus, metabolic status of PDA tumors is an important determinant of the immunosuppressive environment. Clin Cancer Res; 22(14); 3606-17. ©2016 AACR.
Collapse
Affiliation(s)
- Jack Hutcheson
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, Texas
| | - Uthra Balaji
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, Texas
| | - Matthew R Porembka
- Simmons Cancer Center, UT Southwestern Medical Center, Dallas, Texas. Department of Surgery, UT Southwestern Medical Center, Dallas, Texas
| | - Megan B Wachsmann
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | - Peter A McCue
- Department of Pathology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Erik S Knudsen
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, Texas. Simmons Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| | - Agnieszka K Witkiewicz
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, Texas. Simmons Cancer Center, UT Southwestern Medical Center, Dallas, Texas. Department of Pathology, UT Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
78
|
Monitoring and functional characterization of the lymphocytic compartment in pancreatic ductal adenocarcinoma patients. Pancreatology 2016; 16:1069-1079. [PMID: 27424476 DOI: 10.1016/j.pan.2016.07.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 05/26/2016] [Accepted: 07/09/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND/OBJECTIVES Pancreatic ductal adenocarcinoma (PDAC) still has a poor prognosis and current treatments including immunotherapy often fail. This might be due to the pronounced immunosuppressive milieu impairing infiltration and function of immune effector cells. This study aimed at a comprehensive analysis of immune cells in PDAC patients by determining absolute and relative peripheral blood cell numbers of immune cell subsets along with their functional capacity. METHODS Whole blood cells or isolated peripheral blood mononuclear cells were characterized by flow cytometry. PDAC tissues were analyzed by immunohistochemistry. Anti-tumor activity of immune effector cells was determined by RTCA system. RESULTS Our data demonstrate that relative CD4+ memory- and regulatory T cell numbers were enhanced, whereas determination of absolute cell numbers revealed generally lower immune cell numbers in PDAC patients compared to healthy controls. γδ T cells accumulated at higher numbers compared to αβ T cells in the malignant ductal epithelium of PDAC tissues indicating that γδ T cells infiltrate into the tumor. Cytotoxicity against tumor cells of even small numbers of T- and NK cells could be induced by a bispecific antibody targeting CD3+ T cells to human epidermal growth factor receptor (HER)2 expressing PDAC cells or Trastuzumab. Importantly, a critical number of γδ T cells was required for significant tumor cell killing by a bispecific antibody engaging the γδ T cell receptor on γδ T cells and HER2 on tumor cells. CONCLUSION Monitoring immune cells along with the determination of their functional capacity provides a comprehensive assessment as a prerequisite for a personalized immunotherapeutic PDAC treatment.
Collapse
|
79
|
Lapin NA, Krzykawska-Serda M, Ware MJ, Curley SA, Corr SJ. Intravital microscopy for evaluating tumor perfusion of nanoparticles exposed to non-invasive radiofrequency electric fields. Cancer Nanotechnol 2016; 7:5. [PMID: 27429662 PMCID: PMC4927593 DOI: 10.1186/s12645-016-0016-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 05/10/2016] [Indexed: 12/26/2022] Open
Abstract
Poor biodistribution and accumulation of chemotherapeutics in tumors due to limitations on diffusive transport and high intra-tumoral pressures (Jain RK, Nat Med. 7(9):987–989, 2001) have prompted the investigation of adjunctive therapies to improve treatment outcomes. Hyperthermia has been widely applied in attempts to meet this need, but it is limited in its ability to reach tumors in deeply located body regions. High-intensity radiofrequency (RF) electric fields have the potential to overcome such barriers enhancing delivery and extravasation of chemotherapeutics. However, due to factors, including tumor heterogeneity and lack of kinetic information, there is insufficient understanding of time-resolved interaction between RF fields and tumor vasculature, drug molecules and nanoparticle (NP) vectors. Intravital microscopy (IVM) provides time-resolved high-definition images of specific tumor microenvironments, overcoming heterogeneity issues, and can be integrated with a portable RF device to enable detailed observation over time of the effects of the RF field on kinetics and biodistribution at the microvascular level. Herein, we provide a protocol describing the safe integration of IVM with a high-powered non-invasive RF field applied to 4T1 orthotopic breast tumors in live mice. Results show increased perfusion of NPs in microvasculature upon RF hyperthermia treatment and increased perfusion, release and spreading of injected reagents preferentially in irregular vessels during RF exposure.
Collapse
Affiliation(s)
- Norman A Lapin
- Department of Surgery, Division of Surgical Research, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
| | - Martyna Krzykawska-Serda
- Department of Surgery, Division of Surgical Research, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA ; Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, 30-387 Poland
| | - Matthew J Ware
- Department of Surgery, Division of Surgical Research, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
| | - Steven A Curley
- Department of Surgery, Division of Surgical Research, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA ; Department of Mechanical Engineering & Materials Science, Rice University, Houston, 77005 TX USA
| | - Stuart J Corr
- Department of Surgery, Division of Surgical Research, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA ; Department of Chemistry, Rice University, Houston, 77005 TX USA ; Department of Biomedical Engineering, University of Houston, Houston, 77204 TX USA
| |
Collapse
|
80
|
Integrinβ1 modulates tumour resistance to gemcitabine and serves as an independent prognostic factor in pancreatic adenocarcinomas. Tumour Biol 2016; 37:12315-12327. [PMID: 27289231 DOI: 10.1007/s13277-016-5061-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 05/01/2016] [Indexed: 01/21/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies because of its broad resistance to chemotherapy. Numerous evidence indicates that integrinβ1 is upregulated in some human cancers, and it is correlated with resistance to various therapies. However, the role of integrinβ1 in chemotherapy is not clear in pancreatic cancer. The present study evaluates the potential of integrinβ1 to predict chemoresistance and prognosis in patients and to modulate resistance to gemcitabine in PDAC cells. Primary drug-resistance (DR) cancer cells were isolated, and DR cells from MiaPaCa-2 and AsPC-1 parent cell lines (PCL) were selected. Integrinβ1 expression was determined using immunohistochemistry (IHC), quantitative real-time PCR (qRT-PCR) and Western blotting. Changes in drug response after knockdown of integrinβ1 via RNA interference (RNAi) were evaluated using the viability of cancer cells as colon formation, proliferation using Western blot of Ki-67 and apoptosis using cleaved caspase-3 immunofluorescence. qRT-PCR and Western blot also detected variations in the activities of cdc42 and AKT after integrinβ1 suppression. Patient survival and relative factors were assessed using Kaplan-Meier and Cox regression analyses. Integrinβ1 expression was upregulated in PDAC, which was significantly associated with intrinsic and acquired gemcitabine resistance and worse outcomes. The downregulation of integrinβ1 attenuated PDAC chemoresistance, and this attenuation partially correlated with reduced Cdc42 and AKT activity, which are target molecules of integrinβ1 in some human cancers. These findings identified integrinβ1 as a special marker of drug resistance and a serious prognosis, and they furthermore support the use of integrinβ1 as a novel potential therapeutic target to overcome chemotherapy resistance. The results also suggest a possible drug-resistant signalling pathway of integrinβ1 in PDAC.
Collapse
|
81
|
Mühlberg L, Kühnemuth B, Costello E, Shaw V, Sipos B, Huber M, Griesmann H, Krug S, Schober M, Gress TM, Michl P. miRNA dynamics in tumor-infiltrating myeloid cells modulating tumor progression in pancreatic cancer. Oncoimmunology 2016; 5:e1160181. [PMID: 27471627 DOI: 10.1080/2162402x.2016.1160181] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 02/09/2016] [Accepted: 02/25/2016] [Indexed: 12/14/2022] Open
Abstract
Myeloid cells including tumor-associated macrophages (TAM) and myeloid-derived suppressor cells (MDSC) are known as important mediators of tumor progression in solid tumors such as pancreatic cancer. Infiltrating myeloid cells have been identified not only in invasive tumors, but also in early pre-invasive pancreatic intraepithelial precursor lesions (PanIN). The functional dynamics of myeloid cells during carcinogenesis is largely unknown. We aimed to systematically elucidate phenotypic and transcriptional changes in infiltrating myeloid cells during carcinogenesis and tumor progression in a genetic mouse model of pancreatic cancer. Using murine pancreatic myeloid cells isolated from the genetic mouse model at different time points during carcinogenesis, we examined both established markers of macrophage polarization using RT-PCR and FACS as well as transcriptional changes focusing on miRNA profiling. Myeloid cells isolated during carcinogenesis showed a simultaneous increase of established markers of M1 and M2 polarization during carcinogenesis, indicating that phenotypic changes of myeloid cells during carcinogenesis do not follow the established M1/M2 classification. MiRNA profiling revealed distinct regulations of several miRNAs already present in myeloid cells infiltrating pre-invasive PanIN lesions. Among them miRNA-21 was significantly increased in myeloid cells surrounding both PanIN lesions and invasive cancers. Functionally, miRNA-21-5p and -3p altered expression of the immune-modulating cytokines CXCL-10 and CCL-3 respectively. Our data indicate that miRNAs are dynamically regulated in infiltrating myeloid cells during carcinogenesis and mediate their functional phenotype by facilitating an immune-suppressive tumor-promoting micro-milieu.
Collapse
Affiliation(s)
- Leonie Mühlberg
- Department of Gastroenterology and Endocrinology, University Hospital, Philipps-University , Marburg, Germany
| | - Benjamin Kühnemuth
- Department of Gastroenterology and Endocrinology, University Hospital, Philipps-University , Marburg, Germany
| | - Eithne Costello
- Department of Surgery, University of Liverpool , Liverpool, UK
| | - Victoria Shaw
- Department of Surgery, University of Liverpool , Liverpool, UK
| | - Bence Sipos
- Institute of Pathology, University of Tuebingen , Tuebingen, Germany
| | - Magdalena Huber
- Institute of Microbiology, University Hospital, Philipps-University , Marburg, Germany
| | - Heidi Griesmann
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg , Halle, Germany
| | - Sebastian Krug
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg , Halle, Germany
| | - Marvin Schober
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg , Halle, Germany
| | - Thomas M Gress
- Department of Gastroenterology and Endocrinology, University Hospital, Philipps-University , Marburg, Germany
| | - Patrick Michl
- Department of Gastroenterology and Endocrinology, University Hospital, Philipps-University, Marburg, Germany; Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
82
|
Kleeff J, Korc M, Apte M, La Vecchia C, Johnson CD, Biankin AV, Neale RE, Tempero M, Tuveson DA, Hruban RH, Neoptolemos JP. Pancreatic cancer. Nat Rev Dis Primers 2016; 2:16022. [PMID: 27158978 DOI: 10.1038/nrdp.2016.22] [Citation(s) in RCA: 1304] [Impact Index Per Article: 144.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Pancreatic cancer is a major cause of cancer-associated mortality, with a dismal overall prognosis that has remained virtually unchanged for many decades. Currently, prevention or early diagnosis at a curable stage is exceedingly difficult; patients rarely exhibit symptoms and tumours do not display sensitive and specific markers to aid detection. Pancreatic cancers also have few prevalent genetic mutations; the most commonly mutated genes are KRAS, CDKN2A (encoding p16), TP53 and SMAD4 - none of which are currently druggable. Indeed, therapeutic options are limited and progress in drug development is impeded because most pancreatic cancers are complex at the genomic, epigenetic and metabolic levels, with multiple activated pathways and crosstalk evident. Furthermore, the multilayered interplay between neoplastic and stromal cells in the tumour microenvironment challenges medical treatment. Fewer than 20% of patients have surgically resectable disease; however, neoadjuvant therapies might shift tumours towards resectability. Although newer drug combinations and multimodal regimens in this setting, as well as the adjuvant setting, appreciably extend survival, ∼80% of patients will relapse after surgery and ultimately die of their disease. Thus, consideration of quality of life and overall survival is important. In this Primer, we summarize the current understanding of the salient pathophysiological, molecular, translational and clinical aspects of this disease. In addition, we present an outline of potential future directions for pancreatic cancer research and patient management.
Collapse
Affiliation(s)
- Jorg Kleeff
- NIHR Pancreas Biomedical Research Unit, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Royal Liverpool and Broadgreen University Hospitals NHS Trust, Duncan Building, Daulby Street, Liverpool L69 3GA, UK
- Department of General, Visceral and Pediatric Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Murray Korc
- Departments of Medicine, and Biochemistry and Molecular Biology, Indiana University School of Medicine, the Melvin and Bren Simon Cancer Center, and the Pancreatic Cancer Signature Center, Indianapolis, Indiana, USA
| | - Minoti Apte
- SWS Clinical School, University of New South Wales, and Ingham Institute for Applied Medical Research, Sydney, New South Wales, Australia
| | - Carlo La Vecchia
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Colin D Johnson
- University Surgical Unit, University Hospital Southampton, Southampton, UK
| | - Andrew V Biankin
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Garscube Estate, Bearsden, Glasgow, Scotland, UK
| | - Rachel E Neale
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Margaret Tempero
- UCSF Pancreas Center, University of California San Francisco - Mission Bay Campus/Mission Hall, San Francisco, California, USA
| | - David A Tuveson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, New York, USA
| | - Ralph H Hruban
- The Sol Goldman Pancreatic Cancer Research Center, Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - John P Neoptolemos
- NIHR Pancreas Biomedical Research Unit, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Royal Liverpool and Broadgreen University Hospitals NHS Trust, Duncan Building, Daulby Street, Liverpool L69 3GA, UK
| |
Collapse
|
83
|
Weledji EP, Enoworock G, Mokake M, Sinju M. How Grim is Pancreatic Cancer? Oncol Rev 2016; 10:294. [PMID: 27471581 PMCID: PMC4943093 DOI: 10.4081/oncol.2016.294] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 06/12/2016] [Accepted: 06/29/2016] [Indexed: 12/16/2022] Open
Abstract
Pancreatic ductal carcinoma continues to be the most lethal malignancy with rising incidence. It is the fourth most common cause of cancer death in the western world due to its low treatment success rate. In addition, because of its rapid growth and silent course, diagnosis is often only established in the advanced stages. As one of the most aggressive malignancies, the treatment of this disease is a great challenge to clinicians. This paper reviewed the natural history of pancreatic cancer, the current clinical practice and the future in pancreatic cancer management.
Collapse
Affiliation(s)
| | | | - Martin Mokake
- Department of Surgery and Obstetrics and Gynaecology, University of Buea, Cameroon
| | - Motaze Sinju
- Department of Surgery and Obstetrics and Gynaecology, University of Buea, Cameroon
| |
Collapse
|
84
|
Genrich G, Kruppa M, Lenk L, Helm O, Broich A, Freitag-Wolf S, Röcken C, Sipos B, Schäfer H, Sebens S. The anti-oxidative transcription factor Nuclear factor E2 related factor-2 (Nrf2) counteracts TGF-β1 mediated growth inhibition of pancreatic ductal epithelial cells -Nrf2 as determinant of pro-tumorigenic functions of TGF-β1. BMC Cancer 2016; 16:155. [PMID: 26915435 PMCID: PMC4766703 DOI: 10.1186/s12885-016-2191-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 02/17/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Nuclear factor E2 related factor-2 (Nrf2) is an oxidative stress inducible transcription factor being essential in regulating cell homeostasis. Thus, acute induction of Nrf2 in epithelial cells exposed to inflammation confers protection from oxidative cell damage and mutagenesis supporting an anti-tumorigenic role for Nrf2. However, pancreatic ductal adenocarcinoma (PDAC) is characterized by persistent Nrf2 activity conferring therapy resistance which points to a pro-tumorigenic role of Nrf2. A similar dichotomous role in tumorigenesis is described for the Transforming Growth Factor-beta 1 (TGF-β1). The present study therefore aimed at elucidating whether the switch of Nrf2 function towards a tumor promoting one relates to the modulation of TGF-β1 induced cell responses and whether this might occur early in PDAC development. METHODS In situ analysis comprised immunohistochemical stainings of activated (phosphorylated) Nrf2 and Ki67 in pancreatic tissues containing normal ducts and pancreatic intraepithelial neoplasia (PanINs). In vitro, Nrf2 levels in benign (H6c7-pBp), premalignant (H6c7-kras) and malignant (Colo357) pancreatic ductal epithelial cells were modulated by Nrf2 specific siRNA or Nrf2 overexpression. Then, the effect of Nrf2 alone and in combination with TGF-β1 on cell growth and survival was investigated by cell counting, Ki67 staining and apoptosis assays. The underlying cell signaling was investigated by western blotting. Statistical analysis was performed by Shapiro-Wilk test for normal distribution. Parametric data were analyzed by one-way ANOVA, while non-parametric data were analyzed by Kruskal-Wallis one-way ANOVA on ranks. RESULTS Significantly elevated expression of activated Nrf2 and Ki67 could be detected in PanINs but not in normal pancreatic ductal epithelium. While the effect of Nrf2 on basal cell growth of H6c7-pBp, H6c7-kras and Colo357 cells was minor, it clearly attenuated the growth inhibiting effects of TGF-β1 in all cell lines. This enhanced Nrf2-mediated cell survival was predominantly based on an enhanced proliferative activity. Accordingly, expression of p21 expression along with expression of phospho-p38 and phospho-Smad3 was diminished whereas Erk-phosphorylation was enhanced under these conditions. CONCLUSIONS Overall, our data demonstrate that Nrf2 being elevated in early precursor lesions counteracts the growth inhibiting function of TGF-β1 already in benign and premalignant pancreatic ductal epithelial cells. This could represent one fundamental mechanism underlying the functional switch of both- TGF-β1 and Nrf2 - which may manifest already in early stages of PDAC development.
Collapse
Affiliation(s)
- Geeske Genrich
- Group Inflammatory Carcinogenesis, Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, Arnold-Heller-Str. 3, Building 17, 24105, Kiel, Germany.
| | - Marcus Kruppa
- Group Inflammatory Carcinogenesis, Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, Arnold-Heller-Str. 3, Building 17, 24105, Kiel, Germany.
| | - Lennart Lenk
- Group Inflammatory Carcinogenesis, Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, Arnold-Heller-Str. 3, Building 17, 24105, Kiel, Germany.
| | - Ole Helm
- Group Inflammatory Carcinogenesis, Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, Arnold-Heller-Str. 3, Building 17, 24105, Kiel, Germany.
| | - Anna Broich
- Group Inflammatory Carcinogenesis, Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, Arnold-Heller-Str. 3, Building 17, 24105, Kiel, Germany.
| | - Sandra Freitag-Wolf
- Institute of Medical Informatics and Statistics, UKSH Campus Kiel, Brunswiker Str. 10, 24105, Kiel, Germany.
| | - Christoph Röcken
- Department of Pathology, Christian-Albrechts-University Kiel, Arnold-Heller-Str. 3, Building 14, 24105, Kiel, Germany.
| | - Bence Sipos
- Department of Pathology and Neuropathology, University Hospital Tübingen, Liebermeisterstr. 8, 72076, Tübingen, Germany.
| | - Heiner Schäfer
- Laboratory of Molecular Gastroenterology & Hepatology, Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, Arnold-Heller-Str. 3, Building 6, 24105, Kiel, Germany.
| | - Susanne Sebens
- Group Inflammatory Carcinogenesis, Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, Arnold-Heller-Str. 3, Building 17, 24105, Kiel, Germany.
| |
Collapse
|
85
|
De Monte L, Wörmann S, Brunetto E, Heltai S, Magliacane G, Reni M, Paganoni AM, Recalde H, Mondino A, Falconi M, Aleotti F, Balzano G, Algül H, Doglioni C, Protti MP. Basophil Recruitment into Tumor-Draining Lymph Nodes Correlates with Th2 Inflammation and Reduced Survival in Pancreatic Cancer Patients. Cancer Res 2016; 76:1792-803. [DOI: 10.1158/0008-5472.can-15-1801-t] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 01/26/2016] [Indexed: 11/16/2022]
|
86
|
Felix K, Gaida MM. Neutrophil-Derived Proteases in the Microenvironment of Pancreatic Cancer -Active Players in Tumor Progression. Int J Biol Sci 2016; 12:302-13. [PMID: 26929737 PMCID: PMC4753159 DOI: 10.7150/ijbs.14996] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
A hallmark of pancreatic ductal adenocarcinoma (PDAC) is the fibro-inflammatory microenvironment, consisting of activated pancreatic stellate cells, extracellular matrix proteins, and a variety of inflammatory cells, such as T cells, macrophages, or neutrophils. Tumor-infiltrating immune cells, which are found in nearly all cancers, including PDAC, often fail to eliminate the tumor, but conversely can promote its progression by altering the tumor microenvironment. Pancreatic cancer cells are able to attract polymorphonuclear neutrophils (PMN) via tumor secreted chemokines and in human PDAC, PMN infiltrates can be observed in the vicinity of tumor cells and in the desmoplastic tumor stroma, which correlate with undifferentiated tumor growth and poor prognosis. The behavior of tumor-infiltrating neutrophils in the tumor micromilieu is not yet understood at a mechanistic level. It has been shown that PMN have the potential to kill tumor cells, either directly or by antibody-dependent cell-mediated cytotoxicity, but on the other side various adverse effects of PMN, such as promotion of aggressive tumor growth with epithelial-to-mesenchymal transition and increased metastatic potential, have been described. Recent therapeutic approaches for PDAC focus not only the tumor cell itself, but also elements of the tumor microenvironment. Therefore, the role of PMN and their derived products (e.g. cytokines, proteases) as a new vein for a therapeutic target should be critically evaluated in this context. This review summarizes the current understanding of the interplay between proteases of tumor-infiltrating neutrophils and pancreatic tumor cells and elements of the desmoplastic stroma.
Collapse
Affiliation(s)
- Klaus Felix
- 1. Department of General Surgery, University of Heidelberg, INF 110, Heidelberg, Germany
| | - Matthias M Gaida
- 2. Institute of Pathology, University of Heidelberg, INF 224, Heidelberg, Germany
| |
Collapse
|
87
|
Bijlsma MF, van Laarhoven HWM. The conflicting roles of tumor stroma in pancreatic cancer and their contribution to the failure of clinical trials: a systematic review and critical appraisal. Cancer Metastasis Rev 2016; 34:97-114. [PMID: 25566685 DOI: 10.1007/s10555-014-9541-1] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
A nearly universal feature of pancreatic ductal adenocarcinoma (PDAC) is an extensive presence of activated stroma. This stroma is thought to aid in various tumor-promoting processes and hampers response to therapy. Here, we aim to evaluate the evidence that supports this role of the stroma in PDAC with functional experiments in relevant models, discuss the clinical trials that have aimed to target the stroma in this disease, and examine recent work that explains why these clinical trials based on stroma-targeting strategies have thus far not achieved the expected success. We systematically searched PubMed through August 2014 with no restrictions to identify published peer-reviewed research articles assessing the effect of targeting the stroma on tumor growth or metastases in preclinical animal models. Five hundred and thirty articles were extracted of which 31 were included in the analysis. Unfortunately, due to the large variety in models and outcome measures, we could not perform a meta-analysis of our data. We find that despite an abundance of positive outcomes reported in previous studies on stroma targeting, a strong discrepancy exists with the outcomes of clinical trials and the more recent preclinical work that is in line with these trials. We explain the incongruities by the duration of stroma targeting and propose that chronic stroma targeting treatment is possibly detrimental in the treatment of this disease.
Collapse
Affiliation(s)
- Maarten F Bijlsma
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental Molecular Medicine, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | | |
Collapse
|
88
|
Delitto D, Perez C, Han S, Gonzalo DH, Pham K, Knowlton AE, Graves CL, Behrns KE, Moldawer LL, Thomas RM, Liu C, George TJ, Trevino JG, Wallet SM, Hughes SJ. Downstream mediators of the intratumoral interferon response suppress antitumor immunity, induce gemcitabine resistance and associate with poor survival in human pancreatic cancer. Cancer Immunol Immunother 2015; 64:1553-1563. [PMID: 26423423 PMCID: PMC5129167 DOI: 10.1007/s00262-015-1760-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 09/17/2015] [Indexed: 02/07/2023]
Abstract
The cancer microenvironment allows tumor cells to evade immune surveillance through a variety of mechanisms. While interferon-γ (IFNγ) is central to effective antitumor immunity, its effects on the microenvironment are not as clear and have in some cancers been shown to induce immune checkpoint ligands. The heterogeneity of these responses to IFNγ remains poorly characterized in desmoplastic malignancies with minimal inflammatory cell infiltration, such as pancreatic cancer (PC). Thus, the IFNγ response within and on key cells of the PC microenvironment was evaluated. IFNγ induced expression of human leukocyte antigen (HLA) class I and II on PC cell lines, primary pancreatic cancer epithelial cells (PPCE) and patient-derived tumor-associated stroma, concomitant with an upregulation of PDL1 in the absence of CD80 and CD86 expression. As expected, IFNγ also induced high levels of CXCL10 from all cell types. In addition, significantly higher levels of CXCL10 were observed in PC specimens compared to those from chronic pancreatitis, whereby intratumoral CXCL10 concentration was an independent predictor of poor survival. Immunohistochemical analysis revealed a subset of CXCR3-positive cancer cells in over 90 % of PC specimens, as well as on a subset of cultured PC cell lines and PPCE, whereby exposure to CXCL10 induced resistance to the chemotherapeutic gemcitabine. These findings suggest that IFNγ has multiple effects on many cell types within the PC microenvironment that may lead to immune evasion, chemoresistance and shortened survival.
Collapse
Affiliation(s)
- Daniel Delitto
- Department of Surgery, College of Medicine, University of Florida, PO BOX 100109, Gainesville, FL, 32610, USA
| | - Chelsey Perez
- Department of Surgery, College of Medicine, University of Florida, PO BOX 100109, Gainesville, FL, 32610, USA
| | - Song Han
- Department of Surgery, College of Medicine, University of Florida, PO BOX 100109, Gainesville, FL, 32610, USA
| | - David H Gonzalo
- Department of Pathology, Immunology, Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Kien Pham
- Department of Pathology, Immunology, Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Andrea E Knowlton
- Department of Oral Biology, College of Dentistry, University of Florida, PO BOX 100434, Gainesville, FL, 32610, USA
| | - Christina L Graves
- Department of Oral Biology, College of Dentistry, University of Florida, PO BOX 100434, Gainesville, FL, 32610, USA
| | - Kevin E Behrns
- Department of Surgery, College of Medicine, University of Florida, PO BOX 100109, Gainesville, FL, 32610, USA
| | - Lyle L Moldawer
- Department of Surgery, College of Medicine, University of Florida, PO BOX 100109, Gainesville, FL, 32610, USA
| | - Ryan M Thomas
- Department of Surgery, College of Medicine, University of Florida, PO BOX 100109, Gainesville, FL, 32610, USA
| | - Chen Liu
- Department of Pathology, Immunology, Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Thomas J George
- Department of Medicine, University of Florida, Gainesville, FL, USA
| | - Jose G Trevino
- Department of Surgery, College of Medicine, University of Florida, PO BOX 100109, Gainesville, FL, 32610, USA
| | - Shannon M Wallet
- Department of Oral Biology, College of Dentistry, University of Florida, PO BOX 100434, Gainesville, FL, 32610, USA.
| | - Steven J Hughes
- Department of Surgery, College of Medicine, University of Florida, PO BOX 100109, Gainesville, FL, 32610, USA.
| |
Collapse
|
89
|
Selective inhibition of the p38 alternative activation pathway in infiltrating T cells inhibits pancreatic cancer progression. Nat Med 2015; 21:1337-43. [PMID: 26479921 PMCID: PMC4636461 DOI: 10.1038/nm.3957] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 08/27/2015] [Indexed: 12/11/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive neoplasm characterized by a marked fibro-inflammatory microenvironment1, the presence of which can promote both cancer induction and growth2–4. Therefore, selective manipulation of local cytokines is an attractive if unrealized therapeutic approach. T cells possess a unique mechanism of activation of p38 MAPK downstream of T cell receptor (TCR) engagement by phosphorylation of Tyr-323 (pY323). This alternative p38 activation pathway is required for pro-inflammatory cytokine production5,6. Here we show in human PDAC that a high percentage of infiltrating pY323+ T cells was associated with large numbers of TNFα and IL-17-producing CD4+ tumor-infiltrating lymphocytes (TIL) and aggressive disease. The growth of murine pancreatic tumors was inhibited by genetic ablation of the alternative p38 pathway, and transfer of wild type CD4+ T cells but not those lacking the alternative pathway enhanced tumor growth in T cell-deficient mice. Strikingly, a plasma membrane-permeable peptide derived from Gadd45α, the naturally-occurring inhibitor of p38 pY323+ (ref. 7), reduced CD4+ TIL production of TNFα, IL-17A, IL-10, and secondary cytokines, halted growth of implanted tumors, and inhibited progression of spontaneous K-ras-driven adenocarcinoma in mice. Thus, TCR-mediated activation of CD4+ TIL results in alternative p38 activation and production of pro-tumorigenic factors, and can be targeted for therapeutic benefit.
Collapse
|
90
|
Pang W, Su J, Wang Y, Feng H, Dai X, Yuan Y, Chen X, Yao W. Pancreatic cancer-secreted miR-155 implicates in the conversion from normal fibroblasts to cancer-associated fibroblasts. Cancer Sci 2015. [PMID: 26195069 PMCID: PMC4638007 DOI: 10.1111/cas.12747] [Citation(s) in RCA: 148] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cancer-associated fibroblasts (CAF) are a major constituent of the pancreatic cancer microenvironment and that the meaning is as intended. Pancreatic cancer cells can induce normal fibroblasts to convert into CAF and, reciprocally, CAF promote tumor invasions and proliferations. The mechanism of the conversion from normal fibroblasts (NF) to CAF remains unclear. MicroRNA are short non-coding RNA involved in the post-transcription gene regulation, which have been defined as an imperative controller in tumor invasions, proliferations and colony formations. Microvesicles (MV) have been proved to be an important mediator of intercellular communication and can selectively transport secreted microRNA from a donor cell into a recipient cell. In this study, we isolated primary pancreatic fibroblasts from wild type C57 mice and co-cultured them with pancreatic cancer cell lines, BxPC-3 and SW1990, and observed the conversion from NF to CAF, or at least CAF-like cells. This phenomenon could also be replicated in primary fibroblasts treated with MV separated from a cancer cell media. We identified that miR-155 was upregulated in PaC-derived MV and we confirmed that normal fibroblasts could convert into CAF after MV containing miR-155 had been taken up. TP53INP1 is a target of miR-155 in fibroblasts and a downregulation of TP53INP1 protein levels could contribute to the fibroblasts' activation. These results indicated that pancreatic cancer cells might reprogram normal adjacent fibroblasts into CAF by means of secreted MV containing miR-155. Targeting the circulating microRNA might be a potential therapy for malignant tumors.
Collapse
Affiliation(s)
- Wenjing Pang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Jiaojiao Su
- Department of Gastroenterology, First-affiliated Hospital, Anhui Medical School, Hefei, China.,Department of Gastroenterology, Lúan People's Hospital, Lúan Affiliated Hospital of Anhui Medical University, 21, Wanxi Road, 237000, Lúan, China
| | - Yalei Wang
- Department of Gastroenterology, First-affiliated Hospital, Anhui Medical School, Hefei, China
| | - Hui Feng
- Department of Gastroenterology, First-affiliated Hospital, Anhui Medical School, Hefei, China
| | - Xin Dai
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Yaozong Yuan
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Xi Chen
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical, Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Weiyan Yao
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
91
|
Zhou H, Qian W, Uckun FM, Wang L, Wang YA, Chen H, Kooby D, Yu Q, Lipowska M, Staley CA, Mao H, Yang L. IGF1 Receptor Targeted Theranostic Nanoparticles for Targeted and Image-Guided Therapy of Pancreatic Cancer. ACS NANO 2015; 9:7976-91. [PMID: 26242412 PMCID: PMC4908958 DOI: 10.1021/acsnano.5b01288] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Overcoming resistance to chemotherapy is a major and unmet medical challenge in the treatment of pancreatic cancer. Poor drug delivery due to stromal barriers in the tumor microenvironment and aggressive tumor biology are additional impediments toward a more successful treatment of pancreatic cancer. In attempts to address these challenges, we developed IGF1 receptor (IGF1R)-directed, multifunctional theranostic nanoparticles for targeted delivery of therapeutic agents into IGF1R-expressing drug-resistant tumor cells and tumor-associated stromal cells. These nanoparticles were prepared by conjugating recombinant human IGF1 to magnetic iron oxide nanoparticles (IONPs) carrying the anthracycline doxorubicin (Dox) as the chemotherapeutic payload. Intravenously administered IGF1-IONPs exhibited excellent tumor targeting and penetration in an orthotopic patient-derived xenograft (PDX) model of pancreatic cancer featuring enriched tumor stroma and heterogeneous cancer cells. IGF1R-targeted therapy using the theranostic IGF1-IONP-Dox significantly inhibited the growth of pancreatic PDX tumors. The effects of the intratumoral nanoparticle delivery and therapeutic responses in the orthotopic pancreatic PDX tumors could be detected by magnetic resonance imaging (MRI) with IONP-induced contrasts. Histological analysis showed that IGF1R-targeted delivery of Dox significantly inhibited cell proliferation and induced apoptotic cell death of pancreatic cancer cells. Therefore, further development of IGF1R-targeted theranostic IONPs and MRI-guided cancer therapy as a precision nanomedicine may provide the basis for more effective treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Hongyu Zhou
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Weiping Qian
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Fatih M. Uckun
- University of Southern California Norris Comprehensive Cancer Center, Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, Los Angeles, California 90027, United States
| | - Liya Wang
- Departments of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Y. Andrew Wang
- Ocean Nanotech, LLC, San Diego, California 92126, United States
| | - Hongyu Chen
- Ocean Nanotech, LLC, San Diego, California 92126, United States
| | - David Kooby
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Qian Yu
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Malgorzata Lipowska
- Departments of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Charles A. Staley
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Hui Mao
- Departments of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Lily Yang
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia 30322, United States
- Departments of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia 30322, United States
- Address correspondence to .
| |
Collapse
|
92
|
Arfmann-Knübel S, Struck B, Genrich G, Helm O, Sipos B, Sebens S, Schäfer H. The Crosstalk between Nrf2 and TGF-β1 in the Epithelial-Mesenchymal Transition of Pancreatic Duct Epithelial Cells. PLoS One 2015. [PMID: 26226105 PMCID: PMC4520686 DOI: 10.1371/journal.pone.0132978] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Nrf2 and TGF-β1 both affect tumorigenesis in a dual fashion, either by preventing carcinogen induced carcinogenesis and suppressing tumor growth, respectively, or by conferring cytoprotection and invasiveness to tumor cells during malignant transformation. Given the involvement of Nrf2 and TGF-β1 in the adaptation of epithelial cells to persistent inflammatory stress, e.g. of the pancreatic duct epithelium during chronic pancreatitis, a crosstalk between Nrf2 and TGF-β1 can be envisaged. By using premalignant human pancreatic duct cells (HPDE) and the pancreatic ductal adenocarcinoma cell line Colo357, we could show that Nrf2 and TGF-β1 independently but additively conferred an invasive phenotype to HPDE cells, whereas acting synergistically in Colo357 cells. This was accompanied by differential regulation of EMT markers like vimentin, Slug, L1CAM and E-cadherin. Nrf2 activation suppressed E-cadherin expression through an as yet unidentified ARE related site in the E-cadherin promoter, attenuated TGF-β1 induced Smad2/3-activity and enhanced JNK-signaling. In Colo357 cells, TGF-β1 itself was capable of inducing Nrf2 whereas in HPDE cells TGF-β1 per-se did not affect Nrf2 activity, but enhanced Nrf2 induction by tBHQ. In Colo357, but not in HPDE cells, the effects of TGF-β1 on invasion were sensitive to Nrf2 knock-down. In both cell lines, E-cadherin re-expression inhibited the proinvasive effect of Nrf2. Thus, the increased invasion of both cell lines relates to the Nrf2-dependent downregulation of E-cadherin expression. In line, immunohistochemistry analysis of human pancreatic intraepithelial neoplasias in pancreatic tissues from chronic pancreatitis patients revealed strong Nrf2 activity already in premalignant epithelial duct cells, accompanied by partial loss of E-cadherin expression. Our findings indicate that Nrf2 and TGF-β1 both contribute to malignant transformation through distinct EMT related mechanisms accounting for an invasive phenotype. Provided a crosstalk between both pathways, Nrf2 and TGF-β1 mutually promote their tumorigenic potential, a condition manifesting already at an early stage during inflammation induced carcinogenesis of the pancreas.
Collapse
Affiliation(s)
- Sarah Arfmann-Knübel
- Laboratory of Molecular Gastroenterology, Dept. of Internal Medicine I, UKSH Campus Kiel, Arnold-Heller-Str. 3, Bldg. 6, 24105, Kiel, Germany
| | - Birte Struck
- Laboratory of Molecular Gastroenterology, Dept. of Internal Medicine I, UKSH Campus Kiel, Arnold-Heller-Str. 3, Bldg. 6, 24105, Kiel, Germany
| | - Geeske Genrich
- Group Inflammatory Carcinogenesis, Institute of Experimental Medicine, CAU Kiel, Arnold-Heller-Str. 3, Bldg. 17, 24105, Kiel, Germany
| | - Ole Helm
- Group Inflammatory Carcinogenesis, Institute of Experimental Medicine, CAU Kiel, Arnold-Heller-Str. 3, Bldg. 17, 24105, Kiel, Germany
| | - Bence Sipos
- Department of Pathology and Neuropathology, University Hospital Tübingen, Liebermeisterstraße 8, 72076, Tübingen, Germany
| | - Susanne Sebens
- Group Inflammatory Carcinogenesis, Institute of Experimental Medicine, CAU Kiel, Arnold-Heller-Str. 3, Bldg. 17, 24105, Kiel, Germany
| | - Heiner Schäfer
- Laboratory of Molecular Gastroenterology, Dept. of Internal Medicine I, UKSH Campus Kiel, Arnold-Heller-Str. 3, Bldg. 6, 24105, Kiel, Germany
- * E-mail:
| |
Collapse
|
93
|
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; 36:5617-5626. [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
|
94
|
Whipple CA. Tumor talk: understanding the conversation between the tumor and its microenvironment. ACTA ACUST UNITED AC 2015; 2:e773. [PMID: 26023680 DOI: 10.14800/ccm.773] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
It was once believed that tumor growth, progression, and metastasis were intrinsically driven by the tumor. Instead, recent research has demonstrated that a solid tumor is surrounded by a complex matrix of cells, particularly fibroblasts, which support and even promote tumor progression. This matrix of stromal cells, also known as the tumor microenvironment (TME), plays a critical role in cancer and may represent a novel therapeutic target. As such, understanding the complex nature of how the tumor initiates and maintains communication, or a "conversation", with the TME is the focus of current investigations. We have previously shown that the most prevalent mutation found in melanoma, BRAFV600E, results in increased expression and secretion of several growth factors, cytokines, and matrix metalloproteinases, including factors that are able to activate fibroblasts. Targeted inhibition of the BRAFV600E mutation resulted in a decrease of secreted proteins into the TME and suggests that targeting the tumor also modifies the TME. Overall, this work, in combination with several additional studies discussed herein, provides strong evidence for the potential therapeutic benefits of targeting the TME, particularly signaling pathways within the fibroblasts, in conjunction with the tumor. This approach may result in extended drug resistance free survival, reduction in metastasis, and improved cytotoxic drug delivery.
Collapse
Affiliation(s)
- Chery A Whipple
- Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756
| |
Collapse
|
95
|
Hollander LL, Guo X, Salem RR, Cha CH. The novel tumor angiogenic factor, adrenomedullin-2, predicts survival in pancreatic adenocarcinoma. J Surg Res 2015; 197:219-24. [PMID: 25982376 DOI: 10.1016/j.jss.2014.11.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 10/19/2014] [Accepted: 11/03/2014] [Indexed: 01/09/2023]
Abstract
BACKGROUND Tumor angiogenesis has been demonstrated to have an important role in the development, progression, and metastasis of pancreas cancer. Adrenomedullin-2 (ADM2) is a calcitonin gene-related peptide that has recently been shown to be a novel tumor angiogenesis factor, acting via mitogen-activated protein kinase/extracellular signal-regulated kinase, phosphoinositide 3-kinase/Akt, and vascular endothelial growth factor/vascular endothelial growth factor-2 signaling pathways. Through the use of tissue microarray (TMA) technology, we hypothesize that ADM2 is an important tumor angiogenesis factor in pancreatic cancer. METHODS Multiple TMAs were created using tissue from pancreatic cancer patients resected between January 1996 and December 2006. Core tissue samples of formalin-fixed, paraffin-embedded blocks of pancreatic cancer tissue were collected through an institutional review board-approved protocol and linked to available clinicopathologic data. Two TMAs consisting of 112 and 60 patients with pancreatic adenocarcinoma were studied for ADM2 protein expression using a quantitative, automated immunofluorescent microscopy system, a technology that removes potential observer bias in TMA analysis. The results were analyzed using independent Student t-test, chi-square, log-rank regression, and Kaplan-Meier methods. RESULTS One hundred sixteen patients were identified for complete analysis, and 56 patients had complete survival data. Median follow-up for survivors was 14.5 mo. Total cellular levels of ADM2 were found to be a predictor of survival in pancreatic cancer. Low ADM2 levels were associated with a higher 5-y survival compared with high ADM2 levels (18% versus 6%, P = 0.05). Median survival was also worse in high ADM2 expressers (18.7 versus 8.6 mo). In accordance with prior-published pancreatic cancer data, a worse histologic grade (P = 0.001), tumor (T) stage (P = 0.009), and overall disease stage (P = 0.004), all portended a worse survival. CONCLUSIONS For the first time, we have demonstrated that high levels of ADM2 expression predict a poorer survival in patients with pancreatic adenocarcinoma. This suggests a possible role of ADM2 in pancreas cancer and as a novel biomarker that predicts poorer survival. Additional study of ADM2 in pancreatic cancer will help reveal its true angiogenic role in pancreas cancer and its potential role as a novel therapeutic target.
Collapse
Affiliation(s)
- Lindsay L Hollander
- Department of Surgery, Yale University School of Medicine, New Haven, Connecticut; Department of Surgery, University of Connecticut School of Medicine, Farmington, Connecticut
| | - Xiaojia Guo
- Department of Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - Ronald R Salem
- Department of Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - Charles H Cha
- Department of Surgery, Yale University School of Medicine, New Haven, Connecticut.
| |
Collapse
|
96
|
Sclafani F, Iyer R, Cunningham D, Starling N. Management of metastatic pancreatic cancer: Current treatment options and potential new therapeutic targets. Crit Rev Oncol Hematol 2015; 95:318-36. [PMID: 25921418 DOI: 10.1016/j.critrevonc.2015.03.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 03/18/2015] [Accepted: 03/31/2015] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma is a malignancy with a poor prognosis, with the majority of patients diagnosed with advanced disease on presentation. Treatment options remain limited with little progress over the last 40 years. This review will focus on the current management of metastatic pancreatic ductal adenocarcinoma, with a discussion of new and future treatment strategies based on an improved understanding of tumour biology and mechanisms of pathogenesis.
Collapse
Affiliation(s)
| | - Ridhima Iyer
- The Royal Marsden NHS Foundation Trust, London and Surrey, UK
| | | | | |
Collapse
|
97
|
Schmitz-Winnenthal FH, Hohmann N, Niethammer AG, Friedrich T, Lubenau H, Springer M, Breiner KM, Mikus G, Weitz J, Ulrich A, Buechler MW, Pianka F, Klaiber U, Diener M, Leowardi C, Schimmack S, Sisic L, Keller AV, Koc R, Springfeld C, Knebel P, Schmidt T, Ge Y, Bucur M, Stamova S, Podola L, Haefeli WE, Grenacher L, Beckhove P. Anti-angiogenic activity of VXM01, an oral T-cell vaccine against VEGF receptor 2, in patients with advanced pancreatic cancer: A randomized, placebo-controlled, phase 1 trial. Oncoimmunology 2015; 4:e1001217. [PMID: 26137397 DOI: 10.1080/2162402x.2014.1001217] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 12/17/2014] [Accepted: 12/17/2014] [Indexed: 12/22/2022] Open
Abstract
VEGFR-2 is expressed on tumor vasculature and a target for anti-angiogenic intervention. VXM01 is a first in kind orally applied tumor vaccine based on live, attenuated Salmonella bacteria carrying an expression plasmid, encoding VEGFR-2. We here studied the safety, tolerability, T effector (Teff), T regulatory (Treg) and humoral responses to VEGFR2 and anti-angiogenic effects in advanced pancreatic cancer patients in a randomized, dose escalation phase I clinical trial. Results of the first 3 mo observation period are reported. Locally advanced or metastatic, pancreatic cancer patients were enrolled. In five escalating dose groups, 30 patients received VXM01 and 15 placebo on days 1, 3, 5, and 7. Treatment was well tolerated at all dose levels. No dose-limiting toxicities were observed. Salmonella excretion and salmonella-specific humoral immune responses occurred in the two highest dose groups. VEGFR2 specific Teff, but not Treg responses were overall increased in vaccinated patients. We furthermore observed a significant reduction of tumor perfusion after 38 d in vaccinated patients together with increased levels of serum biomarkers indicative of anti-angiogenic activity, VEGF-A, and collagen IV. Vaccine specific Teff responses significantly correlated with reductions of tumor perfusion and high levels of preexisting VEGFR2-specific Teff while those showing no antiangiogenic activity had low levels of preexisting VEGFR2 specific Teff, showed a transient early increase of VEGFR2-specific Treg and reduced levels of VEGFR2-specific Teff at later time points - pointing to the possibility that early anti-angiogenic activity might be based at least in part on specific reactivation of preexisting memory T cells.
Collapse
Affiliation(s)
| | - Nicolas Hohmann
- Clinical Pharmacology and Pharmacoepidemiology; University Clinics of Heidelberg ; Heidelberg, Germany
| | | | - Tobias Friedrich
- Diagnostic and Interventional Radiology; University Clinics of Heidelberg ; Heidelberg, Germany
| | | | | | | | - Gerd Mikus
- Clinical Pharmacology and Pharmacoepidemiology; University Clinics of Heidelberg ; Heidelberg, Germany
| | - Jürgen Weitz
- General, Visceral and Transplantation Surgery; University Clinics of Heidelberg ; Heidelberg, Germany
| | - Alexis Ulrich
- General, Visceral and Transplantation Surgery; University Clinics of Heidelberg ; Heidelberg, Germany
| | - Markus W Buechler
- General, Visceral and Transplantation Surgery; University Clinics of Heidelberg ; Heidelberg, Germany
| | - Frank Pianka
- General, Visceral and Transplantation Surgery; University Clinics of Heidelberg ; Heidelberg, Germany
| | - Ulla Klaiber
- General, Visceral and Transplantation Surgery; University Clinics of Heidelberg ; Heidelberg, Germany
| | - Markus Diener
- General, Visceral and Transplantation Surgery; University Clinics of Heidelberg ; Heidelberg, Germany
| | - Christine Leowardi
- General, Visceral and Transplantation Surgery; University Clinics of Heidelberg ; Heidelberg, Germany
| | - Simon Schimmack
- General, Visceral and Transplantation Surgery; University Clinics of Heidelberg ; Heidelberg, Germany
| | - Leila Sisic
- General, Visceral and Transplantation Surgery; University Clinics of Heidelberg ; Heidelberg, Germany
| | - Anne-Valerie Keller
- General, Visceral and Transplantation Surgery; University Clinics of Heidelberg ; Heidelberg, Germany
| | - Ruhan Koc
- General, Visceral and Transplantation Surgery; University Clinics of Heidelberg ; Heidelberg, Germany
| | | | - Philipp Knebel
- General, Visceral and Transplantation Surgery; University Clinics of Heidelberg ; Heidelberg, Germany
| | - Thomas Schmidt
- General, Visceral and Transplantation Surgery; University Clinics of Heidelberg ; Heidelberg, Germany
| | - Yingzi Ge
- Translational Immunology; National Center for Tumor Diseases ; Heidelberg, Germany
| | - Mariana Bucur
- Translational Immunology; National Center for Tumor Diseases ; Heidelberg, Germany
| | - Slava Stamova
- Translational Immunology; National Center for Tumor Diseases ; Heidelberg, Germany
| | - Lilli Podola
- Translational Immunology; National Center for Tumor Diseases ; Heidelberg, Germany
| | - Walter E Haefeli
- Clinical Pharmacology and Pharmacoepidemiology; University Clinics of Heidelberg ; Heidelberg, Germany
| | - Lars Grenacher
- Diagnostic and Interventional Radiology; University Clinics of Heidelberg ; Heidelberg, Germany
| | - Philipp Beckhove
- Translational Immunology; National Center for Tumor Diseases ; Heidelberg, Germany
| |
Collapse
|
98
|
Goebel L, Grage-Griebenow E, Gorys A, Helm O, Genrich G, Lenk L, Wesch D, Ungefroren H, Freitag-Wolf S, Sipos B, Röcken C, Schäfer H, Sebens S. CD4 + T cells potently induce epithelial-mesenchymal-transition in premalignant and malignant pancreatic ductal epithelial cells-novel implications of CD4 + T cells in pancreatic cancer development. Oncoimmunology 2015; 4:e1000083. [PMID: 26137395 DOI: 10.1080/2162402x.2014.1000083] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 12/15/2014] [Accepted: 12/16/2014] [Indexed: 01/06/2023] Open
Abstract
Chronic pancreatitis (CP) is a risk factor of pancreatic ductal adenocarcinoma (PDAC) and characterized by a pronounced desmoplastic reaction with CD4+ T cells accounting for the majority of the stromal T cell infiltrate. Epithelial-mesenchymal-transition (EMT) is a critical process for metastasis by which epithelial/carcinoma cells become enabled to disseminate probably prior to tumor formation. To investigate whether CD4+ T cells induce EMT in human pancreatic ductal epithelial cells, premalignant H6c7 cells were mono- or co-cultured with human CD4+CD25+CD127-CD49d- regulatory T cells (T-regs) or CD4+CD25- T-effector cells (T-effs) being isolated by negative magnetic bead separation from blood of healthy donors. Particularly in the presence of activated T-effs, H6c7 cells acquired a spindle-shaped morphology, reduced E-cadherin expression, and elevated expression of the mesenchymal proteins vimentin, L1CAM, and ZEB-1. This was accompanied by an increased invasive behavior. Moreover, activated T-effs exerted similar effects in the PDAC cell line T3M4. Blocking of TNF-α and IL-6 being released at greater amounts into supernatants during co-cultures with activated T-effs attenuated the EMT-associated alterations in H6c7 cells. Supporting these findings, EMT-associated alterations (exemplified by reduced E-cadherin expression and enhanced expression of vimentin and L1CAM) were predominantly detected in ductal epithelium of CP tissues surrounded by a dense stroma enriched with CD4+ T cells. Overall this study points to a novel role of CD4+ T cells beyond their immune function in pancreatic tumorigenesis and underscores the view that EMT induction in pancreatic ductal epithelial cells represents an early event in PDAC development being essentially promoted by inflammatory processes.
Collapse
Affiliation(s)
- Lisa Goebel
- Group Inflammatory Carcinogenesis; Institute for Experimental Medicine; Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein (UKSH) Campus Kiel ; Kiel, Germany
| | - Evelin Grage-Griebenow
- Group Inflammatory Carcinogenesis; Institute for Experimental Medicine; Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein (UKSH) Campus Kiel ; Kiel, Germany
| | - Artur Gorys
- Group Inflammatory Carcinogenesis; Institute for Experimental Medicine; Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein (UKSH) Campus Kiel ; Kiel, Germany
| | - Ole Helm
- Group Inflammatory Carcinogenesis; Institute for Experimental Medicine; Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein (UKSH) Campus Kiel ; Kiel, Germany
| | - Geeske Genrich
- Group Inflammatory Carcinogenesis; Institute for Experimental Medicine; Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein (UKSH) Campus Kiel ; Kiel, Germany
| | - Lennart Lenk
- Group Inflammatory Carcinogenesis; Institute for Experimental Medicine; Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein (UKSH) Campus Kiel ; Kiel, Germany
| | - Daniela Wesch
- Institute of Immunology; Christian-Albrechts-University and UKSH Campus Kiel ; Kiel, Germany
| | | | - Sandra Freitag-Wolf
- Institute of Medical Informatics and Statistics; UKSH Campus Kiel ; Kiel, Germany
| | - Bence Sipos
- Department of Pathology and Neuropathology; University Hospital Tübingen ; Tübingen, Germany
| | | | - Heiner Schäfer
- Laboratory of Molecular Gastroenterology & Hepatology; Department of Internal Medicine I; UKSH Campus Kiel ; Kiel, Germany
| | - Susanne Sebens
- Group Inflammatory Carcinogenesis; Institute for Experimental Medicine; Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein (UKSH) Campus Kiel ; Kiel, Germany
| |
Collapse
|
99
|
Secq V, Leca J, Bressy C, Guillaumond F, Skrobuk P, Nigri J, Lac S, Lavaut MN, Bui TT, Thakur AK, Callizot N, Steinschneider R, Berthezene P, Dusetti N, Ouaissi M, Moutardier V, Calvo E, Bousquet C, Garcia S, Bidaut G, Vasseur S, Iovanna JL, Tomasini R. Stromal SLIT2 impacts on pancreatic cancer-associated neural remodeling. Cell Death Dis 2015; 6:e1592. [PMID: 25590802 PMCID: PMC4669755 DOI: 10.1038/cddis.2014.557] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 11/10/2014] [Accepted: 11/20/2014] [Indexed: 02/04/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDA) is a critical health issue in the field of cancer, with few therapeutic options. Evidence supports an implication of the intratumoral microenvironment (stroma) on PDA progression. However, its contribution to the role of neuroplastic changes within the pathophysiology and clinical course of PDA, through tumor recurrence and neuropathic pain, remains unknown, neglecting a putative, therapeutic window. Here, we report that the intratumoral microenvironment is a mediator of PDA-associated neural remodeling (PANR), and we highlight factors such as 'SLIT2' (an axon guidance molecule), which is expressed by cancer-associated fibroblasts (CAFs), that impact on neuroplastic changes in human PDA. We showed that 'CAF-secreted SLIT2' increases neurite outgrowth from dorsal root ganglia neurons as well as from Schwann cell migration/proliferation by modulating N-cadherin/β-catenin signaling. Importantly, SLIT2/ROBO signaling inhibition disrupts this stromal/neural connection. Finally, we revealed that SLIT2 expression and CAFs are correlated with neural remodeling within human and mouse PDA. All together, our data demonstrate the implication of CAFs, through the secretion of axon guidance molecule, in PANR. Furthermore, it provides rationale to investigate the disruption of the stromal/neural compartment connection with SLIT2/ROBO inhibitors for the treatment of pancreatic cancer recurrence and pain.
Collapse
Affiliation(s)
- V Secq
- CRCM, Cellular Stress, INSERM, U1068, Parc scientifique de Luminy, Paoli-Calmettes Institute, Aix-Marseille University, UM 105, CNRS, UMR7258, Marseille 13009, France
- Department of Pathology, Hospital North/Mediterranean University, Marseille, France
| | - J Leca
- CRCM, Cellular Stress, INSERM, U1068, Parc scientifique de Luminy, Paoli-Calmettes Institute, Aix-Marseille University, UM 105, CNRS, UMR7258, Marseille 13009, France
| | - C Bressy
- CRCM, Cellular Stress, INSERM, U1068, Parc scientifique de Luminy, Paoli-Calmettes Institute, Aix-Marseille University, UM 105, CNRS, UMR7258, Marseille 13009, France
| | - F Guillaumond
- CRCM, Cellular Stress, INSERM, U1068, Parc scientifique de Luminy, Paoli-Calmettes Institute, Aix-Marseille University, UM 105, CNRS, UMR7258, Marseille 13009, France
| | - P Skrobuk
- CRCM, Cellular Stress, INSERM, U1068, Parc scientifique de Luminy, Paoli-Calmettes Institute, Aix-Marseille University, UM 105, CNRS, UMR7258, Marseille 13009, France
| | - J Nigri
- CRCM, Cellular Stress, INSERM, U1068, Parc scientifique de Luminy, Paoli-Calmettes Institute, Aix-Marseille University, UM 105, CNRS, UMR7258, Marseille 13009, France
| | - S Lac
- CRCM, Cellular Stress, INSERM, U1068, Parc scientifique de Luminy, Paoli-Calmettes Institute, Aix-Marseille University, UM 105, CNRS, UMR7258, Marseille 13009, France
| | - M-N Lavaut
- CRCM, Cellular Stress, INSERM, U1068, Parc scientifique de Luminy, Paoli-Calmettes Institute, Aix-Marseille University, UM 105, CNRS, UMR7258, Marseille 13009, France
- Department of Pathology, Hospital North/Mediterranean University, Marseille, France
| | - T-t Bui
- CRCM, Cellular Stress, INSERM, U1068, Parc scientifique de Luminy, Paoli-Calmettes Institute, Aix-Marseille University, UM 105, CNRS, UMR7258, Marseille 13009, France
| | - A K Thakur
- CRCM, Cellular Stress, INSERM, U1068, Parc scientifique de Luminy, Paoli-Calmettes Institute, Aix-Marseille University, UM 105, CNRS, UMR7258, Marseille 13009, France
| | - N Callizot
- Neuronexperts, Medical North Faculty, Marseille, France
| | | | - P Berthezene
- CRCM, Cellular Stress, INSERM, U1068, Parc scientifique de Luminy, Paoli-Calmettes Institute, Aix-Marseille University, UM 105, CNRS, UMR7258, Marseille 13009, France
| | - N Dusetti
- CRCM, Cellular Stress, INSERM, U1068, Parc scientifique de Luminy, Paoli-Calmettes Institute, Aix-Marseille University, UM 105, CNRS, UMR7258, Marseille 13009, France
| | - M Ouaissi
- Aix-Marseille University, INSERM, CRO2, UMR 911, Marseille 13385, France
| | - V Moutardier
- CRCM, Cellular Stress, INSERM, U1068, Parc scientifique de Luminy, Paoli-Calmettes Institute, Aix-Marseille University, UM 105, CNRS, UMR7258, Marseille 13009, France
| | - E Calvo
- Molecular Endocrinology and Oncology Research Center, CHUL Research Center, Quebec City, QCue, Canada
| | - C Bousquet
- INSERM UMR 1037, CRCT, University Toulouse III, Toulouse, France
| | - S Garcia
- CRCM, Cellular Stress, INSERM, U1068, Parc scientifique de Luminy, Paoli-Calmettes Institute, Aix-Marseille University, UM 105, CNRS, UMR7258, Marseille 13009, France
- Department of Pathology, Hospital North/Mediterranean University, Marseille, France
| | - G Bidaut
- CRCM, Cellular Stress, INSERM, U1068, Parc scientifique de Luminy, Paoli-Calmettes Institute, Aix-Marseille University, UM 105, CNRS, UMR7258, Marseille 13009, France
| | - S Vasseur
- CRCM, Cellular Stress, INSERM, U1068, Parc scientifique de Luminy, Paoli-Calmettes Institute, Aix-Marseille University, UM 105, CNRS, UMR7258, Marseille 13009, France
| | - J L Iovanna
- CRCM, Cellular Stress, INSERM, U1068, Parc scientifique de Luminy, Paoli-Calmettes Institute, Aix-Marseille University, UM 105, CNRS, UMR7258, Marseille 13009, France
| | - R Tomasini
- CRCM, Cellular Stress, INSERM, U1068, Parc scientifique de Luminy, Paoli-Calmettes Institute, Aix-Marseille University, UM 105, CNRS, UMR7258, Marseille 13009, France
| |
Collapse
|
100
|
Hidalgo M, Cascinu S, Kleeff J, Labianca R, Löhr JM, Neoptolemos J, Real FX, Van Laethem JL, Heinemann V. Addressing the challenges of pancreatic cancer: future directions for improving outcomes. Pancreatology 2015; 15:8-18. [PMID: 25547205 DOI: 10.1016/j.pan.2014.10.001] [Citation(s) in RCA: 365] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 10/01/2014] [Accepted: 10/03/2014] [Indexed: 12/11/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC), which accounts for more than 90% of all pancreatic tumours, is a devastating malignancy with an extremely poor prognosis, as shown by a 1-year survival rate of around 18% for all stages of the disease. The low survival rates associated with PDAC primarily reflect the fact that tumours progress rapidly with few specific symptoms and are thus at an advanced stage at diagnosis in most patients. As a result, there is an urgent need to develop accurate markers of pre-invasive pancreatic neoplasms in order to facilitate prediction of cancer risk and to help diagnose the disease at an earlier stage. However, screening for early diagnosis of prostate cancer remains challenging and identifying a highly accurate, low-cost screening test for early PDAC for use in clinical practice remains an important unmet need. More effective therapies are also crucial in PDAC, since progress in identifying novel therapies has been hampered by the genetic complexity of the disease and treatment remains a major challenge. Presently, the greatest step towards improved treatment efficacy has been made in the field of palliative chemotherapy by introducing FOLFIRINOX (folinic acid, 5-fluorouracil, irinotecan and oxaliplatin) and gemcitabine/nab-paclitaxel. Strategies designed to raise the profile of PDAC in research and clinical practice are a further requirement in order to ensure the best treatment for patients. This article proposes a number of approaches that may help to accelerate progress in treating patients with PDAC, which, in turn, may be expected to improve the quality of life and survival for those suffering from this devastating disease.
Collapse
Affiliation(s)
- Manuel Hidalgo
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain.
| | - Stefano Cascinu
- Department of Medical Oncology, University of Ancona, Ancona, Italy
| | - Jörg Kleeff
- Department of General Surgery, Technische Universität München, Munich, Germany
| | | | - J-Matthias Löhr
- Department of Clinical Science, Intervention and Technology, Karolinska Institute, Stockholm, Sweden
| | - John Neoptolemos
- National Institutes of Health Research Liverpool Pancreas Biomedical Research Unit and Cancer Research UK Liverpool Clinical Trials Unit Director, University of Liverpool and Royal Liverpool University Hospital, Liverpool, UK
| | - Francisco X Real
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid and Universitat Pompeu Fabra, Barcelona, Spain
| | - Jean-Luc Van Laethem
- Department of Gastroenterology-GI Cancer Unit, Erasme University Hospital, Brussels, Belgium
| | - Volker Heinemann
- Comprehensive Cancer Centre Munich, Klinikum der Universität München, Munich, Germany
| |
Collapse
|