101
|
Abstract
Fibroblasts regulate the structure and function of healthy tissues, participate transiently in tissue repair after acute inflammation, and assume an aberrant stimulatory role during chronic inflammatory states including cancer. Such cancer-associated fibroblasts (CAFs) modulate the tumor microenvironment and influence the behavior of neoplastic cells in either a tumor-promoting or tumor-inhibiting manner. These pleiotropic functions highlight the inherent plasticity of fibroblasts and may provide new avenues to understand and therapeutically intervene in malignancies. We discuss the emerging themes of CAF biology in the context of tumorigenesis and therapy.
Collapse
Affiliation(s)
- Daniel Öhlund
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - Ela Elyada
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - David Tuveson
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| |
Collapse
|
102
|
Abstract
Fibroblasts regulate the structure and function of healthy tissues, participate transiently in tissue repair after acute inflammation, and assume an aberrant stimulatory role during chronic inflammatory states including cancer. Such cancer-associated fibroblasts (CAFs) modulate the tumor microenvironment and influence the behavior of neoplastic cells in either a tumor-promoting or tumor-inhibiting manner. These pleiotropic functions highlight the inherent plasticity of fibroblasts and may provide new avenues to understand and therapeutically intervene in malignancies. We discuss the emerging themes of CAF biology in the context of tumorigenesis and therapy.
Collapse
Affiliation(s)
- Daniel Öhlund
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - Ela Elyada
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - David Tuveson
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| |
Collapse
|
103
|
Abstract
Fibroblasts regulate the structure and function of healthy tissues, participate transiently in tissue repair after acute inflammation, and assume an aberrant stimulatory role during chronic inflammatory states including cancer. Such cancer-associated fibroblasts (CAFs) modulate the tumor microenvironment and influence the behavior of neoplastic cells in either a tumor-promoting or tumor-inhibiting manner. These pleiotropic functions highlight the inherent plasticity of fibroblasts and may provide new avenues to understand and therapeutically intervene in malignancies. We discuss the emerging themes of CAF biology in the context of tumorigenesis and therapy.
Collapse
Affiliation(s)
- Daniel Öhlund
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - Ela Elyada
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - David Tuveson
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| |
Collapse
|
104
|
Abstract
Fibroblasts regulate the structure and function of healthy tissues, participate transiently in tissue repair after acute inflammation, and assume an aberrant stimulatory role during chronic inflammatory states including cancer. Such cancer-associated fibroblasts (CAFs) modulate the tumor microenvironment and influence the behavior of neoplastic cells in either a tumor-promoting or tumor-inhibiting manner. These pleiotropic functions highlight the inherent plasticity of fibroblasts and may provide new avenues to understand and therapeutically intervene in malignancies. We discuss the emerging themes of CAF biology in the context of tumorigenesis and therapy.
Collapse
Affiliation(s)
- Daniel Öhlund
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - Ela Elyada
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - David Tuveson
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| |
Collapse
|
105
|
Abstract
Fibroblasts regulate the structure and function of healthy tissues, participate transiently in tissue repair after acute inflammation, and assume an aberrant stimulatory role during chronic inflammatory states including cancer. Such cancer-associated fibroblasts (CAFs) modulate the tumor microenvironment and influence the behavior of neoplastic cells in either a tumor-promoting or tumor-inhibiting manner. These pleiotropic functions highlight the inherent plasticity of fibroblasts and may provide new avenues to understand and therapeutically intervene in malignancies. We discuss the emerging themes of CAF biology in the context of tumorigenesis and therapy.
Collapse
Affiliation(s)
- Daniel Öhlund
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - Ela Elyada
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - David Tuveson
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| |
Collapse
|
106
|
Abstract
Fibroblasts regulate the structure and function of healthy tissues, participate transiently in tissue repair after acute inflammation, and assume an aberrant stimulatory role during chronic inflammatory states including cancer. Such cancer-associated fibroblasts (CAFs) modulate the tumor microenvironment and influence the behavior of neoplastic cells in either a tumor-promoting or tumor-inhibiting manner. These pleiotropic functions highlight the inherent plasticity of fibroblasts and may provide new avenues to understand and therapeutically intervene in malignancies. We discuss the emerging themes of CAF biology in the context of tumorigenesis and therapy.
Collapse
Affiliation(s)
- Daniel Öhlund
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - Ela Elyada
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - David Tuveson
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| |
Collapse
|
107
|
Öhlund D, Elyada E, Tuveson D. Fibroblast heterogeneity in the cancer wound. J Exp Med 2014. [DOI: 10.1084/jem.20140692 order by 1-- eloc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Fibroblasts regulate the structure and function of healthy tissues, participate transiently in tissue repair after acute inflammation, and assume an aberrant stimulatory role during chronic inflammatory states including cancer. Such cancer-associated fibroblasts (CAFs) modulate the tumor microenvironment and influence the behavior of neoplastic cells in either a tumor-promoting or tumor-inhibiting manner. These pleiotropic functions highlight the inherent plasticity of fibroblasts and may provide new avenues to understand and therapeutically intervene in malignancies. We discuss the emerging themes of CAF biology in the context of tumorigenesis and therapy.
Collapse
Affiliation(s)
- Daniel Öhlund
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - Ela Elyada
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - David Tuveson
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| |
Collapse
|
108
|
Abstract
Fibroblasts regulate the structure and function of healthy tissues, participate transiently in tissue repair after acute inflammation, and assume an aberrant stimulatory role during chronic inflammatory states including cancer. Such cancer-associated fibroblasts (CAFs) modulate the tumor microenvironment and influence the behavior of neoplastic cells in either a tumor-promoting or tumor-inhibiting manner. These pleiotropic functions highlight the inherent plasticity of fibroblasts and may provide new avenues to understand and therapeutically intervene in malignancies. We discuss the emerging themes of CAF biology in the context of tumorigenesis and therapy.
Collapse
Affiliation(s)
- Daniel Öhlund
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - Ela Elyada
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - David Tuveson
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| |
Collapse
|
109
|
Abstract
Fibroblasts regulate the structure and function of healthy tissues, participate transiently in tissue repair after acute inflammation, and assume an aberrant stimulatory role during chronic inflammatory states including cancer. Such cancer-associated fibroblasts (CAFs) modulate the tumor microenvironment and influence the behavior of neoplastic cells in either a tumor-promoting or tumor-inhibiting manner. These pleiotropic functions highlight the inherent plasticity of fibroblasts and may provide new avenues to understand and therapeutically intervene in malignancies. We discuss the emerging themes of CAF biology in the context of tumorigenesis and therapy.
Collapse
Affiliation(s)
- Daniel Öhlund
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - Ela Elyada
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - David Tuveson
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| |
Collapse
|
110
|
Abstract
Fibroblasts regulate the structure and function of healthy tissues, participate transiently in tissue repair after acute inflammation, and assume an aberrant stimulatory role during chronic inflammatory states including cancer. Such cancer-associated fibroblasts (CAFs) modulate the tumor microenvironment and influence the behavior of neoplastic cells in either a tumor-promoting or tumor-inhibiting manner. These pleiotropic functions highlight the inherent plasticity of fibroblasts and may provide new avenues to understand and therapeutically intervene in malignancies. We discuss the emerging themes of CAF biology in the context of tumorigenesis and therapy.
Collapse
Affiliation(s)
- Daniel Öhlund
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - Ela Elyada
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - David Tuveson
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| |
Collapse
|
111
|
Abstract
Fibroblasts regulate the structure and function of healthy tissues, participate transiently in tissue repair after acute inflammation, and assume an aberrant stimulatory role during chronic inflammatory states including cancer. Such cancer-associated fibroblasts (CAFs) modulate the tumor microenvironment and influence the behavior of neoplastic cells in either a tumor-promoting or tumor-inhibiting manner. These pleiotropic functions highlight the inherent plasticity of fibroblasts and may provide new avenues to understand and therapeutically intervene in malignancies. We discuss the emerging themes of CAF biology in the context of tumorigenesis and therapy.
Collapse
Affiliation(s)
- Daniel Öhlund
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - Ela Elyada
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - David Tuveson
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| |
Collapse
|
112
|
Abstract
Fibroblasts regulate the structure and function of healthy tissues, participate transiently in tissue repair after acute inflammation, and assume an aberrant stimulatory role during chronic inflammatory states including cancer. Such cancer-associated fibroblasts (CAFs) modulate the tumor microenvironment and influence the behavior of neoplastic cells in either a tumor-promoting or tumor-inhibiting manner. These pleiotropic functions highlight the inherent plasticity of fibroblasts and may provide new avenues to understand and therapeutically intervene in malignancies. We discuss the emerging themes of CAF biology in the context of tumorigenesis and therapy.
Collapse
Affiliation(s)
- Daniel Öhlund
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - Ela Elyada
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - David Tuveson
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| |
Collapse
|
113
|
Abstract
Fibroblasts regulate the structure and function of healthy tissues, participate transiently in tissue repair after acute inflammation, and assume an aberrant stimulatory role during chronic inflammatory states including cancer. Such cancer-associated fibroblasts (CAFs) modulate the tumor microenvironment and influence the behavior of neoplastic cells in either a tumor-promoting or tumor-inhibiting manner. These pleiotropic functions highlight the inherent plasticity of fibroblasts and may provide new avenues to understand and therapeutically intervene in malignancies. We discuss the emerging themes of CAF biology in the context of tumorigenesis and therapy.
Collapse
Affiliation(s)
- Daniel Öhlund
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - Ela Elyada
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - David Tuveson
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| |
Collapse
|
114
|
Abstract
Fibroblasts regulate the structure and function of healthy tissues, participate transiently in tissue repair after acute inflammation, and assume an aberrant stimulatory role during chronic inflammatory states including cancer. Such cancer-associated fibroblasts (CAFs) modulate the tumor microenvironment and influence the behavior of neoplastic cells in either a tumor-promoting or tumor-inhibiting manner. These pleiotropic functions highlight the inherent plasticity of fibroblasts and may provide new avenues to understand and therapeutically intervene in malignancies. We discuss the emerging themes of CAF biology in the context of tumorigenesis and therapy.
Collapse
Affiliation(s)
- Daniel Öhlund
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - Ela Elyada
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - David Tuveson
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| |
Collapse
|
115
|
Abstract
Fibroblasts regulate the structure and function of healthy tissues, participate transiently in tissue repair after acute inflammation, and assume an aberrant stimulatory role during chronic inflammatory states including cancer. Such cancer-associated fibroblasts (CAFs) modulate the tumor microenvironment and influence the behavior of neoplastic cells in either a tumor-promoting or tumor-inhibiting manner. These pleiotropic functions highlight the inherent plasticity of fibroblasts and may provide new avenues to understand and therapeutically intervene in malignancies. We discuss the emerging themes of CAF biology in the context of tumorigenesis and therapy.
Collapse
Affiliation(s)
- Daniel Öhlund
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - Ela Elyada
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - David Tuveson
- D. Öhlund, E. Elyada, and D. Tuveson are at the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| |
Collapse
|
116
|
Coleman SJ, Watt J, Arumugam P, Solaini L, Carapuca E, Ghallab M, Grose RP, Kocher HM. Pancreatic cancer organotypics: High throughput, preclinical models for pharmacological agent evaluation. World J Gastroenterol 2014; 20:8471-8481. [PMID: 25024603 PMCID: PMC4093698 DOI: 10.3748/wjg.v20.i26.8471] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 01/15/2014] [Accepted: 04/01/2014] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer carries a terrible prognosis, as the fourth most common cause of cancer death in the Western world. There is clearly a need for new therapies to treat this disease. One of the reasons no effective treatment has been developed in the past decade may in part, be explained by the diverse influences exerted by the tumour microenvironment. The tumour stroma cross-talk in pancreatic cancer can influence chemotherapy delivery and response rate. Thus, appropriate preclinical in vitro models which can bridge simple 2D in vitro cell based assays and complex in vivo models are required to understand the biology of pancreatic cancer. Here we discuss the evolution of 3D organotypic models, which recapitulare the morphological and functional features of pancreatic ductal adenocarcinoma (PDAC). Organotypic cultures are a valid high throughput preclinical in vitro model that maybe a useful tool to help establish new therapies for PDAC. A huge advantage of the organotypic model system is that any component of the model can be easily modulated in a short time-frame. This allows new therapies that can target the cancer, the stromal compartment or both to be tested in a model that mirrors the in vivo situation. A major challenge for the future is to expand the cellular composition of the organotypic model to further develop a system that mimics the PDAC environment more precisely. We discuss how this challenge is being met to increase our understanding of this terrible disease and develop novel therapies that can improve the prognosis for patients.
Collapse
|
117
|
Herrmann D, Conway JRW, Vennin C, Magenau A, Hughes WE, Morton JP, Timpson P. Three-dimensional cancer models mimic cell-matrix interactions in the tumour microenvironment. Carcinogenesis 2014; 35:1671-9. [PMID: 24903340 DOI: 10.1093/carcin/bgu108] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Basic in vitro systems can be used to model and assess complex diseases, such as cancer. Recent advances in this field include the incorporation of multiple cell types and extracellular matrix proteins into three-dimensional (3D) models to recapitulate the structure, organization and functionality of live tissue in situ. Cells within such a 3D environment behave very differently from cells on two-dimensional (2D) substrates, as cell-matrix interactions trigger signalling pathways and cellular responses in 3D, which may not be observed in 2D. Thus, the use of 3D systems can be advantageous for the assessment of disease progression over 2D set-ups alone. Here, we highlight the current advantages and challenges of employing 3D systems in the study of cancer and provide an overview to guide the appropriate use of distinct models in cancer research.
Collapse
Affiliation(s)
- David Herrmann
- Cancer Division, Garvan Institute of Medical Research, The Kinghorn Cancer Centre, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia, Diabetes and Obesity Division, Garvan Institute of Medical Research, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia and The Beatson Institute for Cancer Research, Garscube Estate, Glasgow G61 1BD, UK
| | - James R W Conway
- Cancer Division, Garvan Institute of Medical Research, The Kinghorn Cancer Centre, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia, Diabetes and Obesity Division, Garvan Institute of Medical Research, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia and The Beatson Institute for Cancer Research, Garscube Estate, Glasgow G61 1BD, UK
| | - Claire Vennin
- Cancer Division, Garvan Institute of Medical Research, The Kinghorn Cancer Centre, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia, Diabetes and Obesity Division, Garvan Institute of Medical Research, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia and The Beatson Institute for Cancer Research, Garscube Estate, Glasgow G61 1BD, UK
| | - Astrid Magenau
- Cancer Division, Garvan Institute of Medical Research, The Kinghorn Cancer Centre, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia, Diabetes and Obesity Division, Garvan Institute of Medical Research, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia and The Beatson Institute for Cancer Research, Garscube Estate, Glasgow G61 1BD, UK
| | - William E Hughes
- Diabetes and Obesity Division, Garvan Institute of Medical Research, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia and
| | - Jennifer P Morton
- The Beatson Institute for Cancer Research, Garscube Estate, Glasgow G61 1BD, UK
| | - Paul Timpson
- Cancer Division, Garvan Institute of Medical Research, The Kinghorn Cancer Centre, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia, Diabetes and Obesity Division, Garvan Institute of Medical Research, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia and The Beatson Institute for Cancer Research, Garscube Estate, Glasgow G61 1BD, UK
| |
Collapse
|
118
|
Sirica AE, Gores GJ. Desmoplastic stroma and cholangiocarcinoma: clinical implications and therapeutic targeting. Hepatology 2014; 59:2397-402. [PMID: 24123296 PMCID: PMC3975806 DOI: 10.1002/hep.26762] [Citation(s) in RCA: 150] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 09/15/2013] [Indexed: 12/12/2022]
Affiliation(s)
- Alphonse E. Sirica
- Division of Cellular and Molecular Pathogenesis, Department of Pathology, Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Gregory J. Gores
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, MN
| |
Collapse
|
119
|
Abstract
FGFR (fibroblast growth factor receptor) signalling plays critical roles in embryogensis, adult physiology, tissue repair and many pathologies. Of particular interest over recent years, it has been implicated in a wide range of cancers, and concerted efforts are underway to target different aspects of FGFR signalling networks. A major focus has been identifying the canonical downstream signalling pathways in cancer cells, and these are now relatively well understood. In the present review, we focus on two distinct but emerging hot topics in FGF biology: its role in stromal cross-talk during cancer progression and the potential roles of FGFR signalling in the nucleus. These neglected areas are proving to be of great interest clinically and are intimately linked, at least in pancreatic cancer. The importance of the stroma in cancer is well accepted, both as a conduit/barrier for treatment and as a target in its own right. Nuclear receptors are less acknowledged as targets, largely due to historical scepticism as to their existence or importance. However, increasing evidence from across the receptor tyrosine kinase field is now strong enough to make the study of nuclear growth factor receptors a major area of interest.
Collapse
|
120
|
Håkanson M, Cukierman E, Charnley M. Miniaturized pre-clinical cancer models as research and diagnostic tools. Adv Drug Deliv Rev 2014; 69-70:52-66. [PMID: 24295904 PMCID: PMC4019677 DOI: 10.1016/j.addr.2013.11.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 10/09/2013] [Accepted: 11/24/2013] [Indexed: 12/14/2022]
Abstract
Cancer is one of the most common causes of death worldwide. Consequently, important resources are directed towards bettering treatments and outcomes. Cancer is difficult to treat due to its heterogeneity, plasticity and frequent drug resistance. New treatment strategies should strive for personalized approaches. These should target neoplastic and/or activated microenvironmental heterogeneity and plasticity without triggering resistance and spare host cells. In this review, the putative use of increasingly physiologically relevant microfabricated cell-culturing systems intended for drug development is discussed. There are two main reasons for the use of miniaturized systems. First, scaling down model size allows for high control of microenvironmental cues enabling more predictive outcomes. Second, miniaturization reduces reagent consumption, thus facilitating combinatorial approaches with little effort and enables the application of scarce materials, such as patient-derived samples. This review aims to give an overview of the state-of-the-art of such systems while predicting their application in cancer drug development.
Collapse
Affiliation(s)
- Maria Håkanson
- CSEM SA, Section for Micro-Diagnostics, 7302 Landquart, Switzerland
| | - Edna Cukierman
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.
| | - Mirren Charnley
- Centre for Micro-Photonics and Industrial Research Institute Swinburne, Swinburne University of Technology, Victoria 3122, Australia.
| |
Collapse
|
121
|
Rucki AA, Zheng L. Pancreatic cancer stroma: Understanding biology leads to new therapeutic strategies. World J Gastroenterol 2014; 20:2237-2246. [PMID: 24605023 PMCID: PMC3942829 DOI: 10.3748/wjg.v20.i9.2237] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 12/14/2013] [Accepted: 01/20/2014] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) is among the deadliest cancers in the United States and in the world. Late diagnosis, early metastasis and lack of effective therapy are among the reasons why only 6% of patients diagnosed with PDA survive past 5 years. Despite development of targeted therapy against other cancers, little progression has been made in the treatment of PDA. Therefore, there is an urgent need for the development of new treatments. However, in order to proceed with treatments, the complicated biology of PDA needs to be understood first. Interestingly, majority of the tumor volume is not made of malignant epithelial cells but of stroma. In recent years, it has become evident that there is an important interaction between the stromal compartment and the less prevalent malignant cells, leading to cancer progression. The stroma not only serves as a growth promoting source of signals but it is also a physical barrier to drug delivery. Understanding the tumor-stroma signaling leading to development of desmoplastic reaction and tumor progression can lead to the development of therapies to decrease stromal activity and improve drug delivery. In this review, we focus on how the current understanding of biology of the pancreatic tumor microenvironment can be translated into the development of targeted therapy.
Collapse
|
122
|
Coleman SJ, Chioni AM, Ghallab M, Anderson RK, Lemoine NR, Kocher HM, Grose RP. Nuclear translocation of FGFR1 and FGF2 in pancreatic stellate cells facilitates pancreatic cancer cell invasion. EMBO Mol Med 2014; 6:467-81. [PMID: 24503018 PMCID: PMC3992074 DOI: 10.1002/emmm.201302698] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Pancreatic cancer is characterised by desmoplasia, driven by activated pancreatic stellate cells (PSCs). Over-expression of FGFs and their receptors is a feature of pancreatic cancer and correlates with poor prognosis, but whether their expression impacts on PSCs is unclear. At the invasive front of human pancreatic cancer, FGF2 and FGFR1 localise to the nucleus in activated PSCs but not cancer cells. In vitro, inhibiting FGFR1 and FGF2 in PSCs, using RNAi or chemical inhibition, resulted in significantly reduced cell proliferation, which was not seen in cancer cells. In physiomimetic organotypic co-cultures, FGFR inhibition prevented PSC as well as cancer cell invasion. FGFR inhibition resulted in cytoplasmic localisation of FGFR1 and FGF2, in contrast to vehicle-treated conditions where PSCs with nuclear FGFR1 and FGF2 led cancer cells to invade the underlying extra-cellular matrix. Strikingly, abrogation of nuclear FGFR1 and FGF2 in PSCs abolished cancer cell invasion. These findings suggest a novel therapeutic approach, where preventing nuclear FGF/FGFR mediated proliferation and invasion in PSCs leads to disruption of the tumour microenvironment, preventing pancreatic cancer cell invasion.
Collapse
Affiliation(s)
- Stacey J Coleman
- Centre for Tumour Biology Barts Cancer Institute - a CRUK Centre of Excellence, Queen Mary University of London, London, UK
| | | | | | | | | | | | | |
Collapse
|
123
|
De Wever O, Van Bockstal M, Mareel M, Hendrix A, Bracke M. Carcinoma-associated fibroblasts provide operational flexibility in metastasis. Semin Cancer Biol 2014; 25:33-46. [PMID: 24406210 DOI: 10.1016/j.semcancer.2013.12.009] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 12/20/2013] [Accepted: 12/30/2013] [Indexed: 01/11/2023]
Abstract
Malignant cancer cells do not act as lone wolves to achieve metastasis, as they exist within a complex ecosystem consisting of an extracellular matrix scaffold populated by carcinoma-associated fibroblasts (CAFs), endothelial cells and immune cells. We recognize local (primary tumor) and distant ecosystems (metastasis). CAFs, also termed myofibroblasts, may have other functions in the primary tumor versus the metastasis. Cellular origin and tumor heterogeneity lead to the expression of specific markers. The molecular characteristics of a CAF remain in evolution since CAFs show operational flexibility. CAFs respond dynamically to a cancer cell's fluctuating demands by shifting profitable signals necessary in metastasis. Local, tissue-resident fibroblasts and mesenchymal stem cells (MSCs) coming from reservoir sites such as bone marrow and adipose tissue are the main progenitor cells of CAFs. CAFs may induce awakening from metastatic dormancy, a major cause of cancer-specific death. Cancer management protocols influence CAF precursor recruitment and CAF activation. Since CAF signatures represent early changes in metastasis, including formation of pre-metastatic niches, we discuss whether liquid biopsies, including exosomes, may detect and monitor CAF reactions allowing optimized prognosis of cancer patients.
Collapse
Affiliation(s)
- Olivier De Wever
- Laboratory of Experimental Cancer Research, Department of Radiotherapy and Experimental Cancer Research, Ghent University Hospital, Ghent, Belgium.
| | | | - Marc Mareel
- Laboratory of Experimental Cancer Research, Department of Radiotherapy and Experimental Cancer Research, Ghent University Hospital, Ghent, Belgium
| | - An Hendrix
- Laboratory of Experimental Cancer Research, Department of Radiotherapy and Experimental Cancer Research, Ghent University Hospital, Ghent, Belgium
| | - Marc Bracke
- Laboratory of Experimental Cancer Research, Department of Radiotherapy and Experimental Cancer Research, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
124
|
Abstract
The progression from normal cells to invasive pancreatic ductal adenocarcinoma (PDAC) requires the accumulation of multiple inherited or acquired mutations. Activating point mutations in the KRAS oncogene are prevalent in pancreatic cancer and result in the stimulation of several pathways including the RAF-mitogen-activated protein kinase pathway and the phosphoinositide 3-kinase pathway. Other genetic alterations, including telomere shortening and the inactivation of tumor suppressor genes such as CDKN2A, TP53, and SMAD4, which encode p16, p53, and SMAD4, respectively, also contribute to the progression of pancreatic cancer. These, and other genetic events, can present at different stages in the development of PDAC at histologically defined precursor lesions known as pancreatic intraepithelial neoplasia, intraductal papillary mucinous neoplasms, or mucinous cystic neoplasms. Each precursor lesion represents alternate routes to PDAC formation and has a unique presentation and somewhat distinct genetic events controlling its development. Despite the advances in the understanding of the genetics of PDAC, the prognosis for this cancer remains poor, and several important aspects of its pathogenesis must be clarified to improve therapeutics, including the timing and method of metastases, as well as the relationship of the tumor cells with the desmoplastic stroma, which is a characteristic feature of the cancer. This review discusses the principal genetic alterations in PDAC and its precursor lesions, including their effects on promoting carcinogenesis.
Collapse
Affiliation(s)
- Robert W Cowan
- From the Department of Pathology and Translational Molecular Pathology, Sheikh Ahmed Bin Zayed Al Nahyan Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | |
Collapse
|
125
|
Wen Z, Liao Q, Zhao J, Hu Y, You L, Lu Z, Jia C, Wei Y, Zhao Y. High expression of interleukin-22 and its receptor predicts poor prognosis in pancreatic ductal adenocarcinoma. Ann Surg Oncol 2014; 21:125-32. [PMID: 24132627 DOI: 10.1245/s10434-013-3322-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Indexed: 12/22/2022]
Abstract
BACKGROUND The cytokine interleukin-22 (IL-22) and its receptor are present in the tumor microenvironment. Their function in pancreatic ductal adenocarcinoma (PDAC) remains largely unknown. The goal of the present study was to measure the expression of IL-22 and IL-22R in PDAC and assess their relationship with clinicopathological features and prognosis. METHODS The expression of IL-22 and IL-22R was evaluated by immunohistochemistry in PDAC tissues from 57 patients and by Western blotting in six tumors and adjacent nontumor tissues. A statistical analysis was conducted to assess the relationship between levels of expression, clinicopathological factors, and overall survival. In addition, the relationship between the expression of IL-22 and IL-22R and invasion was assessed by Western blotting and transwell assay with the PDAC cell lines PANC1 and BxPC3. RESULTS Positive IL-22 staining was detected in PDAC tissues and adjacent nontumor tissues. Positive IL-22R staining was detected in PDAC cells. High expression of IL-22 and IL-22R correlated significantly with lymph node involvement. IL-22 increased the phosphorylation of signal transducer and activator of transcription3, the expression of matrix metalloproteinase 9, and the invasion in PANC1 and BxPC3 cells in vitro while silencing of IL-22R RNA caused opposite effects. Most importantly, overall survival was significantly poorer in patients with high expression of IL-22 and IL-22R than in those with low expression. CONCLUSIONS These findings reveal the positive role of IL-22 and IL-22R in invasion and metastasis in human PDAC. IL-22 and IL-22R may be suitable independent prognostic markers in PDAC.
Collapse
Affiliation(s)
- Zhang Wen
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tsinghua University, Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
126
|
Neesse A, Krug S, Gress TM, Tuveson DA, Michl P. Emerging concepts in pancreatic cancer medicine: targeting the tumor stroma. Onco Targets Ther 2013; 7:33-43. [PMID: 24379681 PMCID: PMC3872146 DOI: 10.2147/ott.s38111] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma is a stroma-rich and highly challenging cancer to treat. Over recent years, it has become increasingly evident that the complex network of soluble cytokines, growth factors, proteases, and components of the extracellular matrix collaboratively interact within the tumor microenvironment, sustaining and driving cancer cell proliferation, invasion, and early metastasis. More recently, the tumor microenvironment has also been appreciated to mediate therapeutic resistance in pancreatic ductal adenocarcinoma, thus opening numerous avenues for novel therapeutic explorations. Inert and soluble components of the tumor stroma have been targeted in order to break down the extracellular matrix scaffold, relieve vessel compression, and increase drug delivery to hypovascular tumors. Moreover, targeting of antiapoptotic, immunosuppressive, and pro-proliferative effects of the tumor stroma provides novel vantage points of attack. This review focuses on current and future developments in pancreatic cancer medicine, with a particular emphasis on biophysical and biochemical approaches that target the tumor microenvironment.
Collapse
Affiliation(s)
- Albrecht Neesse
- Department of Gastroenterology, Endocrinology, Infectiology and Metabolism, Philipps University Marburg, Marburg, Germany
| | - Sebastian Krug
- Department of Gastroenterology, Endocrinology, Infectiology and Metabolism, Philipps University Marburg, Marburg, Germany
| | - Thomas M Gress
- Department of Gastroenterology, Endocrinology, Infectiology and Metabolism, Philipps University Marburg, Marburg, Germany
| | | | - Patrick Michl
- Department of Gastroenterology, Endocrinology, Infectiology and Metabolism, Philipps University Marburg, Marburg, Germany
| |
Collapse
|
127
|
Li NF, Gemenetzidis E, Marshall FJ, Davies D, Yu Y, Frese K, Froeling FEM, Woolf AK, Feakins RM, Naito Y, Iacobuzio-Donahue C, Tuveson DA, Hart IR, Kocher HM. RhoC interacts with integrin α5β1 and enhances its trafficking in migrating pancreatic carcinoma cells. PLoS One 2013; 8:e81575. [PMID: 24312560 PMCID: PMC3849283 DOI: 10.1371/journal.pone.0081575] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 10/15/2013] [Indexed: 02/08/2023] Open
Abstract
Human pancreatic ductal adenocarcinoma (PDAC) is characterized by early systemic dissemination. Although RhoC has been implicated in cancer cell migration, the relevant underlying molecular mechanisms remain unknown. RhoC has been implicated in the enhancement of cancer cell migration and invasion, with actions which are distinct from RhoA (84% homology), and are possibly attributed to the divergent C-terminus domain. Here, we confirm that RhoC significantly enhances the migratory and invasive properties of pancreatic carcinoma cells. In addition, we show that RhoC over-expression decreases cancer cell adhesion and, in turn, accelerates cellular body movement and focal adhesion turnover, especially, on fibronectin-coated surfaces. Whilst RhoC over-expression did not alter integrin expression patterns, we show that it enhanced integrin α5β1 internalization and re-cycling (trafficking), an effect that was dependent specifically on the C-terminus (180-193 amino acids) of RhoC protein. We also report that RhoC and integrin α5β1 co-localize within the peri-nuclear region of pancreatic tumor cells, and by masking the CAAX motif at the C-terminal of RhoC protein, we were able to abolish this interaction in vitro and in vivo. Co-localization of integrin α5β1 and RhoC was demonstrable in invading cancer cells in 3D-organotypic cultures, and further mimicked in vivo analyses of, spontaneous human, (two distinct sources: operated patients and rapid autopsy programme) and transgenic murine (LSL-KrasG12D/+;LSL-Trp53R172H/+;Pdx-1-Cre), pancreatic cancers. In both cases, co-localization of integrin α5β1 and RhoC correlated with poor differentiation status and metastatic potential. We propose that RhoC facilitates tumor cell invasion and promotes subsequent metastasis, in part, by enhancing integrin α5β1 trafficking. Thus, RhoC may serve as a biomarker and a therapeutic target.
Collapse
Affiliation(s)
- Ningfeng Fiona Li
- Barts Cancer Institute - a CR-United Kingdom Centre of Excellence, Queen Mary University of London, Centre for Tumour Biology, London, United Kingdom
| | - Emilios Gemenetzidis
- Barts Cancer Institute - a CR-United Kingdom Centre of Excellence, Queen Mary University of London, Centre for Tumour Biology, London, United Kingdom
| | - Francis J. Marshall
- Barts Cancer Institute - a CR-United Kingdom Centre of Excellence, Queen Mary University of London, Centre for Tumour Biology, London, United Kingdom
| | - Derek Davies
- Cancer Research United Kingdom London Research Institute, London, United Kingdom
| | - Yongwei Yu
- Changhai Hospital of Shanghai Second Military Medical University, Pathology Department, Shanghai, China
| | - Kristopher Frese
- Cancer Research United Kingdom Cambridge Research Institute, Cambridge, United Kingdom
| | - Fieke E. M. Froeling
- Barts Cancer Institute - a CR-United Kingdom Centre of Excellence, Queen Mary University of London, Centre for Tumour Biology, London, United Kingdom
| | - Adam K. Woolf
- Barts Cancer Institute - a CR-United Kingdom Centre of Excellence, Queen Mary University of London, Centre for Tumour Biology, London, United Kingdom
| | - Roger M. Feakins
- Barts and the London HPB Centre, The Royal London Hospital, London, United Kingdom
| | - Yoshiki Naito
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Christine Iacobuzio-Donahue
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - David A. Tuveson
- Cancer Research United Kingdom Cambridge Research Institute, Cambridge, United Kingdom
| | - Ian R. Hart
- Barts Cancer Institute - a CR-United Kingdom Centre of Excellence, Queen Mary University of London, Centre for Tumour Biology, London, United Kingdom
| | - Hemant M. Kocher
- Barts Cancer Institute - a CR-United Kingdom Centre of Excellence, Queen Mary University of London, Centre for Tumour Biology, London, United Kingdom
- Barts and the London HPB Centre, The Royal London Hospital, London, United Kingdom
| |
Collapse
|
128
|
Ene-Obong A, Clear AJ, Watt J, Wang J, Fatah R, Riches JC, Marshall JF, Chin-Aleong J, Chelala C, Gribben JG, Ramsay AG, Kocher HM. Activated pancreatic stellate cells sequester CD8+ T cells to reduce their infiltration of the juxtatumoral compartment of pancreatic ductal adenocarcinoma. Gastroenterology 2013; 145:1121-32. [PMID: 23891972 PMCID: PMC3896919 DOI: 10.1053/j.gastro.2013.07.025] [Citation(s) in RCA: 441] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Revised: 06/24/2013] [Accepted: 07/17/2013] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Pancreatic ductal adenocarcinoma (PDAC) is characterized by a prominent desmoplastic microenvironment that contains many different immune cells. Activated pancreatic stellate cells (PSCs) contribute to the desmoplasia. We investigated whether distinct stromal compartments are differentially infiltrated by different types of immune cells. METHODS We used tissue microarray analysis to compare immune cell infiltration of different pancreaticobiliary diseased tissues (PDAC, ampullary carcinoma, cholangiocarcinoma, mucinous cystic neoplasm, chronic inflammation, and chronic pancreatitis) and juxtatumoral stromal (<100 μm from tumor) and panstromal compartments. We investigated the association between immune infiltrate and patient survival times. We also analyzed T-cell migration and tumor infiltration in LSL-KrasG12D/+; LSL-Trp53R172H/+; Pdx-1-Cre (KPC) mice and the effects of all-trans retinoic acid (ATRA) on these processes. RESULTS Juxtatumoral compartments in PDAC samples from 2 independent groups of patients contained increased numbers of myeloperoxidase(+) and CD68(+) cells compared with panstromal compartments. However, juxtatumoral compartments of PDACs contained fewer CD8(+), FoxP3(+), CD56(+), or CD20(+) cells than panstromal compartments, a distinction absent in ampullary carcinomas and cholangiocarcinomas. Patients with PDACs that had high densities of CD8(+) T cells in the juxtatumoral compartment had longer survival times than patients with lower densities. In KPC mice, administration of ATRA, which renders PSCs quiescent, increased numbers of CD8(+) T cells in juxtatumoral compartments. We found that activated PSCs express cytokines, chemokines, and adhesion molecules that regulate T-cell migration. In vitro migration assays showed that CD8(+) T cells, from patients with PDAC, had increased chemotaxis toward activated PSCs, which secrete CXCL12, compared with quiescent PSCs or tumor cells. These effects could be reversed by knockdown of CXCL12 or treatment of PSCs with ATRA. CONCLUSIONS Based on studies of human PDAC samples and KPC mice, activated PSCs appear to reduce migration of CD8(+) T cells to juxtatumoral stromal compartments, preventing their access to cancer cells. Deregulated signaling by activated PSCs could prevent an effective antitumor immune response.
Collapse
Affiliation(s)
- Abasi Ene-Obong
- Centre for Tumour Biology, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Andrew J. Clear
- Centre for Hemato-Oncology, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Jennifer Watt
- Centre for Tumour Biology, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
,Department of Surgery, Barts and the London HPB Centre, The Royal London Hospital, Barts Health NHS Trust, London, E1 1BB, UK.
| | - Jun Wang
- Centre for Molecular Oncology, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Rewas Fatah
- Centre for Hemato-Oncology, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - John C. Riches
- Centre for Hemato-Oncology, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - John F. Marshall
- Centre for Tumour Biology, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Joanne Chin-Aleong
- Department of Pathology, Barts and the London HPB Centre, The Royal London Hospital, Barts Health NHS Trust, London, E1 1BB, UK.
| | - Claude Chelala
- Centre for Molecular Oncology, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - John G. Gribben
- Centre for Hemato-Oncology, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Alan G. Ramsay
- Centre for Hemato-Oncology, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Hemant M. Kocher
- Centre for Tumour Biology, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
,Department of Surgery, Barts and the London HPB Centre, The Royal London Hospital, Barts Health NHS Trust, London, E1 1BB, UK.
| |
Collapse
|
129
|
Watt J, Kocher HM. The desmoplastic stroma of pancreatic cancer is a barrier to immune cell infiltration. Oncoimmunology 2013; 2:e26788. [PMID: 24498555 PMCID: PMC3912010 DOI: 10.4161/onci.26788] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 10/11/2013] [Indexed: 12/17/2022] Open
Abstract
A detailed map demonstrating the spatially restricted distribution of immune cells within the desmoplastic stroma of human pancreatic tumors opens up the field of pancreatic oncoimmunology. CD8+ T cells located in the proximity of malignant lesions are associated with a survival advantage, suggesting the existence of immunoediting. Pancreatic stellate cells appear to dictate the infiltration of the juxtatumoral stroma by CD8+ T cells.
Collapse
Affiliation(s)
- Jennifer Watt
- Centre for Tumour Biology; Barts Cancer Institute; Barts and the London School of Medicine and Dentistry; Queen Mary University of London; London, UK ; Barts and the London HPB Centre; The Royal London Hospital; Barts Health NHS Trust; London UK
| | - Hemant M Kocher
- Centre for Tumour Biology; Barts Cancer Institute; Barts and the London School of Medicine and Dentistry; Queen Mary University of London; London, UK ; Barts and the London HPB Centre; The Royal London Hospital; Barts Health NHS Trust; London UK
| |
Collapse
|
130
|
Sirica AE, Gores GJ. Desmoplastic stroma and cholangiocarcinoma: clinical implications and therapeutic targeting. HEPATOLOGY (BALTIMORE, MD.) 2013. [PMID: 24123296 DOI: 10.1002/hep.26762.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Alphonse E Sirica
- Division of Cellular and Molecular Pathogenesis, Department of Pathology, Virginia Commonwealth University School of Medicine, Richmond, VA
| | | |
Collapse
|
131
|
Erkan M. Understanding the stroma of pancreatic cancer: co-evolution of the microenvironment with epithelial carcinogenesis. J Pathol 2013; 231:4-7. [PMID: 23716361 DOI: 10.1002/path.4213] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2013] [Revised: 05/18/2013] [Accepted: 05/22/2013] [Indexed: 12/18/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a notoriously therapy-resistant desmoplastic tumour. Antifibrotic therapy is shown to increase drug delivery in the preclinical setting. However, this approach can be a double-edged sword: first, PDAC is not uniform and some types of (dormant) stroma may in fact be protective; second, conventional chemotherapeutics are not powerful enough to eradicate all cancer cells in the tumour, therefore breaking down the stromal wall non-selectively may also lead to the increased dissemination of cancer cells. Recently, Kadaba et al have analysed the impact of the stromal cells in pancreatic, oesophageal and skin cancers, in bio-engineered, physiomimetic organotypic cultures. These authors show that the maximal effect on increasing cancer cell proliferation and invasion, as well as decreasing cancer cell apoptosis, occurs when pancreatic stellate cells constitute the majority of the cellular population in a three-dimensional (3D) model. This work may be instrumental for better understanding the types of stoma in PDAC before eliminating it non-selectively.
Collapse
Affiliation(s)
- Mert Erkan
- Department of Surgery, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany.
| |
Collapse
|