151
|
Grimes MM, Kenney SR, Dominguez DR, Brayer KJ, Guo Y, Wandinger-Ness A, Hudson LG. The R-enantiomer of ketorolac reduces ovarian cancer tumor burden in vivo. BMC Cancer 2021; 21:40. [PMID: 33413202 PMCID: PMC7791840 DOI: 10.1186/s12885-020-07716-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 12/08/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Rho-family GTPases, including Ras-related C3 botulinum toxin substrate 1 (Rac1) and cell division control protein 42 (Cdc42), are important modulators of cancer-relevant cell functions and are viewed as promising therapeutic targets. Based on high-throughput screening and cheminformatics we identified the R-enantiomer of an FDA-approved drug (ketorolac) as an inhibitor of Rac1 and Cdc42. The corresponding S-enantiomer is a non-steroidal anti-inflammatory drug (NSAID) with selective activity against cyclooxygenases. We reported previously that R-ketorolac, but not the S-enantiomer, inhibited Rac1 and Cdc42-dependent downstream signaling, growth factor stimulated actin cytoskeleton rearrangements, cell adhesion, migration and invasion in ovarian cancer cell lines and patient-derived tumor cells. METHODS In this study we treated mice with R-ketorolac and measured engraftment of tumor cells to the omentum, tumor burden, and target GTPase activity. In order to gain insights into the actions of R-ketorolac, we also performed global RNA-sequencing (RNA-seq) analysis on tumor samples. RESULTS Treatment of mice with R-ketorolac decreased omental engraftment of ovarian tumor cells at 18 h post tumor cell injection and tumor burden after 2 weeks of tumor growth. R-ketorolac treatment inhibited tumor Rac1 and Cdc42 activity with little impact on mRNA or protein expression of these GTPase targets. RNA-seq analysis revealed that R-ketorolac decreased expression of genes in the HIF-1 signaling pathway. R-ketorolac treatment also reduced expression of additional genes associated with poor prognosis in ovarian cancer. CONCLUSION These findings suggest that R-ketorolac may represent a novel therapeutic approach for ovarian cancer based on its pharmacologic activity as a Rac1 and Cdc42 inhibitor. R-ketorolac modulates relevant pathways and genes associated with disease progression and worse outcome.
Collapse
Affiliation(s)
- Martha M. Grimes
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, New Mexico USA
| | - S. Ray Kenney
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, New Mexico USA
- Division of Molecular Medicine, School of Medicine, University of New Mexico, Albuquerque, New Mexico USA
| | - Dayna R. Dominguez
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, New Mexico USA
| | - Kathryn J. Brayer
- Analytical and Translational Genomics Shared Resource, Comprehensive Cancer Center, University of New Mexico, Albuquerque, New Mexico USA
- Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, New Mexico USA
| | - Yuna Guo
- Department of Pathology, School of Medicine, University of New Mexico, Albuquerque, New Mexico USA
| | - Angela Wandinger-Ness
- Department of Pathology, School of Medicine, University of New Mexico, Albuquerque, New Mexico USA
| | - Laurie G. Hudson
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, New Mexico USA
| |
Collapse
|
152
|
Stromal Protein-Mediated Immune Regulation in Digestive Cancers. Cancers (Basel) 2021; 13:cancers13010146. [PMID: 33466303 PMCID: PMC7795083 DOI: 10.3390/cancers13010146] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/21/2020] [Accepted: 12/24/2020] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Solid cancers are surrounded by a network of non-cancerous cells comprising different cell types, including fibroblasts, and acellular protein structures. This entire network is called the tumor microenvironment (TME) and it provides a physical barrier to the tumor shielding it from infiltrating immune cells, such as lymphocytes, or therapeutic agents. In addition, the TME has been shown to dampen efficient immune responses of infiltrated immune cells, which are key in eliminating cancer cells from the organism. In this review, we will discuss how TME proteins in particular are involved in this dampening effect, known as immunosuppression. We will focus on three different types of digestive cancers: pancreatic cancer, colorectal cancer, and gastric cancer. Moreover, we will discuss current therapeutic approaches using TME proteins as targets to reverse their immunosuppressive effects. Abstract The stromal tumor microenvironment (TME) consists of immune cells, vascular and neural structures, cancer-associated fibroblasts (CAFs), as well as extracellular matrix (ECM), and favors immune escape mechanisms promoting the initiation and progression of digestive cancers. Numerous ECM proteins released by stromal and tumor cells are crucial in providing physical rigidity to the TME, though they are also key regulators of the immune response against cancer cells by interacting directly with immune cells or engaging with immune regulatory molecules. Here, we discuss current knowledge of stromal proteins in digestive cancers including pancreatic cancer, colorectal cancer, and gastric cancer, focusing on their functions in inhibiting tumor immunity and enabling drug resistance. Moreover, we will discuss the implication of stromal proteins as therapeutic targets to unleash efficient immunotherapy-based treatments.
Collapse
|
153
|
Abdul Pari AA, Singhal M, Augustin HG. Emerging paradigms in metastasis research. J Exp Med 2021; 218:e20190218. [PMID: 33601416 PMCID: PMC7754674 DOI: 10.1084/jem.20190218] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 09/17/2020] [Accepted: 11/04/2020] [Indexed: 12/15/2022] Open
Abstract
Historically, therapy of metastatic disease has essentially been limited to using strategies that were identified and established to shrink primary tumors. The limited efficacy of such treatments on overall patient survival stems from diverging intrinsic and extrinsic characteristics of a primary tumor and metastases originating therefrom. To develop better therapeutic strategies to treat metastatic disease, there is an urgent need to shift the paradigm in preclinical metastasis research by conceptualizing metastatic dissemination, colonization, and growth as spatiotemporally dynamic processes and identifying rate-limiting vulnerabilities of the metastatic cascade. Clinically, while metastatic colonization remains the most attractive therapeutic avenue, comprehensive understanding of earlier steps may unravel novel metastasis-restricting therapies for presurgical neoadjuvant application. Moving beyond a primary tumor-centric view, this review adopts a holistic approach to understanding the spatial and temporal progression of metastasis. After reviewing recent developments in metastasis research, we highlight some of the grand challenges and propose a framework to expedite mechanism-based discovery research feeding the translational pipeline.
Collapse
Affiliation(s)
- Ashik Ahmed Abdul Pari
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
- Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Mahak Singhal
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
- Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Hellmut G. Augustin
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
- Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
154
|
Rummler M, Ziouti F, Bouchard AL, Brandl A, Duda GN, Bogen B, Beilhack A, Lynch ME, Jundt F, Willie BM. Mechanical loading prevents bone destruction and exerts anti-tumor effects in the MOPC315.BM.Luc model of myeloma bone disease. Acta Biomater 2021; 119:247-258. [PMID: 33130307 DOI: 10.1016/j.actbio.2020.10.041] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 10/05/2020] [Accepted: 10/27/2020] [Indexed: 02/08/2023]
Abstract
Bone continually adapts to changing external loading conditions via (re)modeling (modeling and remodeling) processes. While physical activity is known to beneficially enhance bone mass in healthy individuals, little is known in how physical stimuli affect osteolytic bone destruction in patients suffering from multiple myeloma bone disease. Multiple myeloma (MM) is caused by malignant plasma cells in the bone marrow, shifting the balance in bone remodeling towards massive resorption. We hypothesized that in vivo tibial mechanical loading has anabolic effects in mice with locally injected MOPC315.BM.Luc cells. Conventional microCT analysis revealed enhanced cortical bone mass and microstructure in loaded compared to nonloaded mice. State-of-the-art time-lapse microCT based image analysis demonstrated bone (re)modeling processes at the endosteal and periosteal surfaces as the underlying causes of increased bone mass. Loading prevented the progression and development of osteolytic destruction. Physical stimuli also diminished local MM cell growth and dissemination evidenced by quantification of MM cell-specific immunoglobulin A levels in the serum of mice and by bioluminescence analysis. These data indicate that mechanical loading not only rescues the bone phenotype, but also exerts cell-extrinsic anti-myeloma effects in the MOPC315.BM.Luc model. In conclusion, the use of physical stimuli should be further investigated as an anabolic treatment for osteolytic bone destruction in patients with MM.
Collapse
|
155
|
Cacho-Díaz B, García-Botello DR, Wegman-Ostrosky T, Ortiz-Sánchez E, Herrera-Montalvo LA. Tumor Microenvironment: Comparison Between Primary Origin Tumors and Corresponding Brain Metastasis. PRINCIPLES OF NEURO-ONCOLOGY 2021:27-41. [DOI: 10.1007/978-3-030-54879-7_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
156
|
Kast V, Loessner D. 3D Models for Ovarian Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1330:139-149. [PMID: 34339035 DOI: 10.1007/978-3-030-73359-9_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The main reasons for the slow progress in improving survival outcomes for ovarian cancer are the 'one-size-fits-all' therapy and lack of clinically relevant experimental models that represent the advanced stages of the human disease. The interaction of tumour cells with their surrounding niche, the tumour microenvironment, influences the spread of ovarian cancer cells within the peritoneum and their responses to therapeutics. Scientists are increasingly using 3D cell culture models to dissect the role of the tumour microenvironment in cancer development and progression and the treatment of this disease. In this chapter, we will briefly describe the tumour microenvironment of ovarian cancer. Then, we will review some of the clinically relevant experimental approaches, such as spheroid, organoid and organotypic models, that have been developed for the 3D culture of ovarian cancer cells using different tools, including hydrogels, scaffolds and cancer-on-a-chip devices, to mimic selected components of the tumour microenvironment.
Collapse
Affiliation(s)
- Verena Kast
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials Dresden, Hohe Straβe, Dresden, Germany
| | - Daniela Loessner
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials Dresden, Hohe Straβe, Dresden, Germany. .,Department of Chemical Engineering, Faculty of Engineering, Monash University, Melbourne, VIC, Australia. .,Department of Materials Science and Engineering, Faculty of Engineering, Monash University, Melbourne, VIC, Australia. .,Department of Anatomy and Developmental Biology, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
157
|
Boulter L, Bullock E, Mabruk Z, Brunton VG. The fibrotic and immune microenvironments as targetable drivers of metastasis. Br J Cancer 2021; 124:27-36. [PMID: 33239677 PMCID: PMC7782519 DOI: 10.1038/s41416-020-01172-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 10/29/2020] [Accepted: 11/02/2020] [Indexed: 02/06/2023] Open
Abstract
Although substantial progress has been made over the past 40 years in treating patients with cancer, effective therapies for those who are diagnosed with advanced metastatic disease are still few and far between. Cancer cells do not exist in isolation: rather, they exist within a complex microenvironment composed of stromal cells and extracellular matrix. Within this tumour microenvironment exists an interplay between the two main stromal cell subtypes, cancer-associated fibroblasts (CAFs) and immune cells, that are important in controlling metastasis. A complex network of paracrine signalling pathways between CAFs, immune cells and tumour cells are involved at multiple stages of the metastatic process, from invasion and intravasation at the primary tumour site to extravasation and colonisation in the metastatic site. Heterogeneity and plasticity within stromal cell populations also contribute to the complexity. Although many of these processes are likely to be common to a number of metastatic sites, we will describe in detail the interplay within the liver, a preferred site of metastasis for many tumours. A greater understanding of these networks provides opportunities for the design of new therapeutic approaches for targeting the metastatic disease.
Collapse
Affiliation(s)
- Luke Boulter
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XU, UK
| | - Esme Bullock
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XU, UK
| | - Zeanap Mabruk
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XU, UK
| | - Valerie G Brunton
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XU, UK.
| |
Collapse
|
158
|
Li K, Fan J, Qin X, Wei Q. Novel therapeutic compounds for prostate adenocarcinoma treatment: An analysis using bioinformatic approaches and the CMap database. Medicine (Baltimore) 2020; 99:e23768. [PMID: 33371142 PMCID: PMC7748316 DOI: 10.1097/md.0000000000023768] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 11/17/2020] [Indexed: 01/15/2023] Open
Abstract
INTRODUCTION Prostate adenocarcinoma is the most frequently diagnosed malignancy, particularly for people >70 years old. The main challenge in the treatment of advanced neoplasm is bone metastasis and therapeutic resistance for known oncology drugs. Novel treatment methods to prolong the survival time and improve the life quality of these specific patients are required. The present study attempted to screen potential therapeutic compounds for the tumor through bioinformatics approaches, in order to provide conceptual treatment for this malignant disease. METHODS Differentially expressed genes were obtained from the Gene Expression Omnibus database and submitted into the Connectivity Map database for the detection of potentially associated compounds. Target genes were extracted from the search results. Functional annotation and pathway enrichment were performed for the confirmation. Survival analysis was used to measure potential therapeutic effects. RESULTS It was revealed that 3 compounds (vanoxerine, tolnaftate, and gabexate) may help to prolong the disease-free survival time from tumor metastasis of patients with the tumor. A total of 6 genes [also-keto reductase family 1 member C3 (AKR1C3), collagen type III α 1 chain (COL3A1), lipoprotein lipase (LPL), glucuronidase, β pseudogene 11 (GUSBP11), apolipoprotein E (APOE), and collagen type I α 1 chain (COL1A1)] were identified to be the potential therapeutic targets for the aforementioned compounds. CONCLUSION In the present study, it was speculated that 3 compounds may function as the potential therapeutic drugs of bone metastatic prostate adenocarcinoma; however, further studies verifying vitro and in vivo are necessary.
Collapse
Affiliation(s)
- Kai Li
- Departments of Orthopedics, The First Affiliated Hospital, Guangxi Medical University
| | - Jingyuan Fan
- Departments of Orthopedics, The First Affiliated Hospital, Guangxi Medical University
| | - Xinyi Qin
- Graduate School of Guangxi Medical University, Nanning, Guangxi, PR China
| | - Qingjun Wei
- Departments of Orthopedics, The First Affiliated Hospital, Guangxi Medical University
| |
Collapse
|
159
|
Petersen EV, Chudakova DA, Skorova EY, Anikin V, Reshetov IV, Mynbaev OA. The Extracellular Matrix-Derived Biomarkers for Diagnosis, Prognosis, and Personalized Therapy of Malignant Tumors. Front Oncol 2020; 10:575569. [PMID: 33425730 PMCID: PMC7793707 DOI: 10.3389/fonc.2020.575569] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 11/10/2020] [Indexed: 01/18/2023] Open
Abstract
The tumor biomarkers already have proven clinical value and have become an integral part in cancer management and modern translational oncology. The tumor tissue microenvironment (TME), which includes extracellular matrix (ECM), signaling molecules, immune and stromal cells, and adjacent non-tumorous tissue, contributes to cancer pathogenesis. Thus, TME-derived biomarkers have many clinical applications. This review is predominately based on the most recent publications (manuscripts published in a last 5 years, or seminal publications published earlier) and fills a gap in the current literature on the cancer biomarkers derived from the TME, with particular attention given to the ECM and products of its processing and degradation, ECM-associated extracellular vesicles (EVs), biomechanical characteristics of ECM, and ECM-derived biomarkers predicting response to the immunotherapy. We discuss the clinical utility of the TME-incorporating three-dimensional in vitro and ex vivo cell culture models for personalized therapy. We conclude that ECM is a critical driver of malignancies and ECM-derived biomarkers should be included in diagnostics and prognostics panels of markers in the clinic.
Collapse
Affiliation(s)
- Elena V. Petersen
- Department of Molecular and Bio Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Daria A. Chudakova
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Ekaterina Yu. Skorova
- Department of Molecular and Bio Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Vladimir Anikin
- Harefield Hospital, The Royal Brompton and Harefield Hospitals NHS Foundation Trust, Harefield, United Kingdom
- Department of Oncology and Reconstructive Surgery, Sechenov Medical University, Moscow, Russia
| | - Igor V. Reshetov
- Department of Molecular and Bio Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
- Department of Oncology and Reconstructive Surgery, Sechenov Medical University, Moscow, Russia
| | - Ospan A. Mynbaev
- Department of Molecular and Bio Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| |
Collapse
|
160
|
Chen Z, Huang H, Wang Y, Zhan F, Quan Z. Identification of Immune-Related Genes MSR1 and TLR7 in Relation to Macrophage and Type-2 T-Helper Cells in Osteosarcoma Tumor Micro-Environments as Anti-metastasis Signatures. Front Mol Biosci 2020; 7:576298. [PMID: 33381518 PMCID: PMC7768026 DOI: 10.3389/fmolb.2020.576298] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 11/24/2020] [Indexed: 12/19/2022] Open
Abstract
Metastasis of osteosarcoma (OS) is an essential factor affecting the prognosis and survival of patients. The tumor microenvironment, including tumor immune-infiltrating cells (TIIC), is closely related to tumor progression. The purpose of this study was to investigate the differences between metastatic and non-metastatic immune-infiltrating cells in OS and to identify key immune-related genes. The differences in immune infiltration in OS metastasis were calculated based on the ssGSEA algorithm of 28 immuno-infiltrating cells. Weighted gene co-expression network analysis (WGCNA) and intersection analysis were used to screen immune-related modules and hubgenes. Univariate/multivariate/Lasso Cox regressions were used for models construction and signatures screening. The receiver operating characteristic (ROC) and Kaplan-Meier (K-M) curves were constructed to observe the metastases of different groups. Both internal and external data were verified. We found that macrophages and Type-2 T-helper cells were significantly decreased in patients with OS metastases. The high-risk groups obtained from multivariate/Lasso Cox models constructed with 11 immune-related hubgenes almost all underwent distant metastases within 5 years. Interestingly and importantly, two genes, MSR1 and TLR7, appeared in various models and various hubgenes, which play an anti-metastasis role and may prolong overall survival in OS. Our study may help elucidate the impact of TIIC on OS metastasis outcomes and to identify biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Zhiyu Chen
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- The First Clinical College, Chongqing Medical University, Chongqing, China
| | - Huanhuan Huang
- The First Clinical College, Chongqing Medical University, Chongqing, China
| | - Yang Wang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- The First Clinical College, Chongqing Medical University, Chongqing, China
| | - Fangbiao Zhan
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- The First Clinical College, Chongqing Medical University, Chongqing, China
| | - Zhengxue Quan
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
161
|
Levi-Galibov O, Lavon H, Wassermann-Dozorets R, Pevsner-Fischer M, Mayer S, Wershof E, Stein Y, Brown LE, Zhang W, Friedman G, Nevo R, Golani O, Katz LH, Yaeger R, Laish I, Porco JA, Sahai E, Shouval DS, Kelsen D, Scherz-Shouval R. Heat Shock Factor 1-dependent extracellular matrix remodeling mediates the transition from chronic intestinal inflammation to colon cancer. Nat Commun 2020; 11:6245. [PMID: 33288768 PMCID: PMC7721883 DOI: 10.1038/s41467-020-20054-x] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/09/2020] [Indexed: 12/25/2022] Open
Abstract
In the colon, long-term exposure to chronic inflammation drives colitis-associated colon cancer (CAC) in patients with inflammatory bowel disease. While the causal and clinical links are well established, molecular understanding of how chronic inflammation leads to the development of colon cancer is lacking. Here we deconstruct the evolving microenvironment of CAC by measuring proteomic changes and extracellular matrix (ECM) organization over time in a mouse model of CAC. We detect early changes in ECM structure and composition, and report a crucial role for the transcriptional regulator heat shock factor 1 (HSF1) in orchestrating these events. Loss of HSF1 abrogates ECM assembly by colon fibroblasts in cell-culture, prevents inflammation-induced ECM remodeling in mice and inhibits progression to CAC. Establishing relevance to human disease, we find high activation of stromal HSF1 in CAC patients, and detect the HSF1-dependent proteomic ECM signature in human colorectal cancer. Thus, HSF1-dependent ECM remodeling plays a crucial role in mediating inflammation-driven colon cancer.
Collapse
Affiliation(s)
- Oshrat Levi-Galibov
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Hagar Lavon
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | | | | | - Shimrit Mayer
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | | | - Yaniv Stein
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Lauren E Brown
- Department of Chemistry and Center for Molecular Discovery (BU-CMD), Boston University, Boston, MA, USA
| | - Wenhan Zhang
- Department of Chemistry and Center for Molecular Discovery (BU-CMD), Boston University, Boston, MA, USA
| | - Gil Friedman
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Reinat Nevo
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Ofra Golani
- Department of Life Sciences Core Facilities, The Weizmann Institute of Science, Rehovot, Israel
| | - Lior H Katz
- Gastroenterology Institute, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
- Department of Gastroenterology and Hepatology, Hadassah Medical Center, Jerusalem, Israel
| | - Rona Yaeger
- Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, and Weil Cornell Medical College, New York, NY, USA
| | - Ido Laish
- Gastroenterology Institute, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - John A Porco
- Department of Chemistry and Center for Molecular Discovery (BU-CMD), Boston University, Boston, MA, USA
| | | | - Dror S Shouval
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
- Pediatric Gastroenterology Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - David Kelsen
- Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, and Weil Cornell Medical College, New York, NY, USA
| | - Ruth Scherz-Shouval
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
162
|
Landolt L, Spagnoli GC, Hertig A, Brocheriou I, Marti HP. Fibrosis and cancer: shared features and mechanisms suggest common targeted therapeutic approaches. Nephrol Dial Transplant 2020; 37:1024-1032. [PMID: 33280031 DOI: 10.1093/ndt/gfaa301] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Indexed: 12/17/2022] Open
Abstract
Epidemiological studies support a strong link between organ fibrosis and epithelial cancers. Moreover, clinical and experimental investigations consistently indicate that these diseases intertwine and share strikingly overlapping features. As a deregulated response to injury occurring in all body tissues, fibrosis is characterized by activation of fibroblasts and immune cells, contributing to progressive deposition of extracellular matrix (ECM) and inflammation. Cancers are driven by genetic alterations resulting in dysregulated cell survival, proliferation and dissemination. However, non-cancerous components of tumour tissues including fibroblasts, inflammatory cells and ECM play key roles in oncogenesis and cancer progression by providing a pro-mutagenic environment where cancer cells can develop, favouring their survival, expansion and invasiveness. Additional commonalities of fibrosis and cancer are also represented by overproduction of growth factors, like transforming growth factor β, epithelial-to-mesenchymal transition, high oxidative stress, Hippo pathway dysfunctions and enhanced cellular senescence. Here, we review advances in the analysis of cellular and molecular mechanisms involved in the pathogenesis of both organ fibrosis and cancer, with particular reference to chronic kidney diseases and renal cell cancers. Most importantly, improved understanding of common features is contributing to the development of innovative treatment strategies targeting shared mechanisms.
Collapse
Affiliation(s)
- Lea Landolt
- Department of Clinical Medicine, University of Bergen, Bergen, Norway.,Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Giulio C Spagnoli
- National Research Council, Institute of Translational Pharmacology, Rome, Italy
| | - Alexandre Hertig
- Sorbonne Université, INSERM UMR S1155, Pitié-Salpêtrière Hospital, APHP6, Paris, France and
| | - Isabelle Brocheriou
- Sorbonne Université, INSERM UMR S1155, Pitié-Salpêtrière Hospital, APHP6, Paris, France and.,Department of Pathology, Pitié-Salpêtrière Hospital, Paris, France
| | - Hans-Peter Marti
- Department of Clinical Medicine, University of Bergen, Bergen, Norway.,Department of Medicine, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
163
|
Sadeghi Rad H, Bazaz SR, Monkman J, Ebrahimi Warkiani M, Rezaei N, O'Byrne K, Kulasinghe A. The evolving landscape of predictive biomarkers in immuno-oncology with a focus on spatial technologies. Clin Transl Immunology 2020; 9:e1215. [PMID: 33251010 PMCID: PMC7680923 DOI: 10.1002/cti2.1215] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/16/2020] [Accepted: 10/25/2020] [Indexed: 02/06/2023] Open
Abstract
Immunotherapies have shown long-lasting and unparalleled responses for cancer patients compared to conventional therapy. However, they seem to only be effective in a subset of patients. Therefore, it has become evident that a greater understanding of the tumor microenvironment (TME) is required to understand the nuances which may be at play for a favorable outcome to therapy. The immune contexture of the TME is an important factor in dictating how well a tumor may respond to immune checkpoint inhibitors. While traditional immunohistochemistry techniques allow for the profiling of cells in the tumor, this is often lost when tumors are analysed using bulk tissue genomic approaches. Moreover, the actual cellular proportions, cellular heterogeneity and deeper spatial distribution are lacking in characterisation. Advances in tissue interrogation technologies have given rise to spatially resolved characterisation of the TME. This review aims to provide an overview of the current methodologies that are used to profile the TME, which may provide insights into the immunopathology associated with a favorable outcome to immunotherapy.
Collapse
Affiliation(s)
| | - Sajad Razavi Bazaz
- School of Biomedical EngineeringUniversity of Technology SydneySydneyNSWAustralia
| | - James Monkman
- The School of Biomedical Science, Institute of Health and Biomedical InnovationQueensland University of TechnologyBrisbaneQLDAustralia
- Translational Research InstituteWoolloongabbaQLDAustralia
| | - Majid Ebrahimi Warkiani
- School of Biomedical EngineeringUniversity of Technology SydneySydneyNSWAustralia
- Institute of Molecular MedicineSechenov UniversityMoscowRussia
| | - Nima Rezaei
- School of MedicineTehran University of Medical SciencesTehranIran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA)Universal Scientific Education and Research Network (USERN)TehranIran
- Research Center for ImmunodeficienciesChildren's Medical CenterTehran University of Medical SciencesTehranIran
| | - Ken O'Byrne
- The School of Biomedical Science, Institute of Health and Biomedical InnovationQueensland University of TechnologyBrisbaneQLDAustralia
- Translational Research InstituteWoolloongabbaQLDAustralia
- Princess Alexandra HospitalWoolloongabbaQLDAustralia
| | - Arutha Kulasinghe
- The School of Biomedical Science, Institute of Health and Biomedical InnovationQueensland University of TechnologyBrisbaneQLDAustralia
- Translational Research InstituteWoolloongabbaQLDAustralia
- Institute for Molecular BiosciencesUniversity of QueenslandBrisbaneQLDAustralia
| |
Collapse
|
164
|
Li R, Qi Y, Han M, Geng B, Wang G, Han M. Computed tomography reveals microenvironment changes in premetastatic lung. Eur Radiol 2020; 31:4340-4349. [PMID: 33219849 DOI: 10.1007/s00330-020-07500-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 10/03/2020] [Accepted: 11/11/2020] [Indexed: 11/28/2022]
Abstract
OBJECTIVES Microenvironment changes had occurred in the metastatic organs before the arriving of the metastatic tumor cells. In this study, we evaluated the effectiveness of computed tomography (CT) images in quantifying the microenvironment changes in the premetastatic lung under both laboratory and clinical conditions. METHOD Free-breathing Balb/c mice underwent micro-CT repeatedly after the implantation of 4T1 breast tumor. CT-derived indicators (aerated lung volume, lung tissue volume, total lung volume, mean lung density, and the ratio of aerated lung volume to the total lung volume) were quantified. Hematoxylin-eosin staining was used to display the microenvironment changes in premetastatic lung. Moreover, we examined healthy adult women, adult women with histopathologically confirmed primary breast cancer, and adult women with histopathologically confirmed primary breast cancer and lung metastases in our institution to test whether the indicators derived from lung CT images changed with the growth of breast cancer. RESULTS In 4T1 tumor-bearing mice, lung density is increased before lung masses can be recognized on CT images and is correlated with the severity of inflammation in the lung microenvironment. In primary breast tumor-bearing patients, lung density is also increased before the clinical diagnosis of pulmonary metastasis and is correlated with disease score, which represents tumor progression. CONCLUSIONS CT is a reliable and quantitative tool that yields dynamic information on metastatic processes. Microenvironmental changes had occurred in patients' lung tissue before the clinical diagnosis of pulmonary metastasis. Our research will provide new insight for clinical research on the premetastatic niche. KEY POINTS • CT, which provides dynamic information on metastatic processes, is a reliable and quantitative tool to bridge laboratory and clinical studies of the premetastatic niche. • We confirmed that microenvironmental changes occurred in patients' lung tissue before clinicians could diagnose pulmonary metastasis. • Our results provide evidence for the study of the premetastatic niche by analyzing information obtained from CT images.
Collapse
Affiliation(s)
- Ranran Li
- Cancer Therapy and Research Center, Shandong Provincial Hospital affiliated to Shandong University, Shandong University, 324 Jingwuweiqi Road, Jinan, 250021, People's Republic of China
| | - Yana Qi
- Cancer Therapy and Research Center, Shandong Provincial Hospital affiliated to Shandong University, Shandong University, 324 Jingwuweiqi Road, Jinan, 250021, People's Republic of China
| | - Meng Han
- School of Basic Medical Sciences, Shandong First Medical University, Jinan, People's Republic of China
| | - Baocheng Geng
- Cancer Therapy and Research Center, Shandong Provincial Hospital affiliated to Shandong University, Shandong University, 324 Jingwuweiqi Road, Jinan, 250021, People's Republic of China
| | - Guangyu Wang
- Cancer Therapy and Research Center, Shandong Provincial Hospital affiliated to Shandong University, Shandong University, 324 Jingwuweiqi Road, Jinan, 250021, People's Republic of China
| | - Mingyong Han
- Cancer Therapy and Research Center, Shandong Provincial Hospital affiliated to Shandong University, Shandong University, 324 Jingwuweiqi Road, Jinan, 250021, People's Republic of China.
| |
Collapse
|
165
|
Micek HM, Visetsouk MR, Masters KS, Kreeger PK. Engineering the Extracellular Matrix to Model the Evolving Tumor Microenvironment. iScience 2020; 23:101742. [PMID: 33225247 PMCID: PMC7666341 DOI: 10.1016/j.isci.2020.101742] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Clinical evidence supports a role for the extracellular matrix (ECM) in cancer risk and prognosis across multiple tumor types, and numerous studies have demonstrated that individual ECM components impact key hallmarks of tumor progression (e.g., proliferation, migration, angiogenesis). However, the ECM is a complex network of fibrillar proteins, glycoproteins, and proteoglycans that undergoes dramatic changes in composition and organization during tumor development. In this review, we will highlight how engineering approaches can be used to examine the impact of changes in tissue architecture, ECM composition (i.e., identity and levels of individual ECM components), and cellular- and tissue-level mechanics on tumor progression. In addition, we will discuss recently developed methods to model the ECM that have not yet been applied to the study of cancer.
Collapse
Affiliation(s)
- Hannah M. Micek
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Mike R. Visetsouk
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Kristyn S. Masters
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Pamela K. Kreeger
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Obstetrics and Gynecology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| |
Collapse
|
166
|
He G, Fu S, Li Y, Li T, Mei P, Feng L, Cai L, Cheng Y, Zhou C, Tang Y, Huang W, Liu H, Cen B, Pan M, Gao Y. TCGA and ESTIMATE data mining to identify potential prognostic biomarkers in HCC patients. Aging (Albany NY) 2020; 12:21544-21558. [PMID: 33177245 PMCID: PMC7695391 DOI: 10.18632/aging.103943] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 08/08/2020] [Indexed: 12/23/2022]
Abstract
Hepatocellular carcinoma (HCC) is an aggressive form of cancer characterized by a high recurrence rate following resection. Studies have implicated stromal and immune cells, which form part of the tumor microenvironment, as significant contributors to the poor prognoses of HCC patients. In the present study, we first downloaded gene expression datasets for HCC patients from The Cancer Genome Atlas database and categorized the patients into low and high stromal or immune score groups. By comparing those groups, we identified differentially expressed genes significantly associated with HCC prognosis. The Gene Ontology database was then used to perform functional enrichment analysis, and the STRING network database was used to construct protein-protein interaction networks. Our results show that most of the differentially expressed genes were involved in immune processes and responses and the plasma membrane. Those results were then validated using another a dataset from a HCC cohort in the Gene Expression Omnibus database and in 10 pairs of HCC tumor tissue and adjacent nontumor tissue. These findings enabled us to identify several tumor microenvironment-related genes that associate with HCC prognosis, and some those appear to have the potential to serve as HCC biomarkers.
Collapse
Affiliation(s)
- Guolin He
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Shunjun Fu
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Yang Li
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Ting Li
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Purong Mei
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Lei Feng
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Lei Cai
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Yuan Cheng
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Chenjie Zhou
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Yujun Tang
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Wenbin Huang
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Haiyan Liu
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Bohong Cen
- Department of Pharmacy, Zhujiang Hospital of Southern Medical University, Guangzhou 510282, Guangdong, China.,Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou 510095, Guangdong, China
| | - Mingxin Pan
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Yi Gao
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| |
Collapse
|
167
|
Barba D, León-Sosa A, Lugo P, Suquillo D, Torres F, Surre F, Trojman L, Caicedo A. Breast cancer, screening and diagnostic tools: All you need to know. Crit Rev Oncol Hematol 2020; 157:103174. [PMID: 33249359 DOI: 10.1016/j.critrevonc.2020.103174] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 09/18/2020] [Accepted: 11/05/2020] [Indexed: 02/06/2023] Open
Abstract
Breast cancer is one of the most frequent malignancies among women worldwide. Methods for screening and diagnosis allow health care professionals to provide personalized treatments that improve the outcome and survival. Scientists and physicians are working side-by-side to develop evidence-based guidelines and equipment to detect cancer earlier. However, the lack of comprehensive interdisciplinary information and understanding between biomedical, medical, and technology professionals makes innovation of new screening and diagnosis tools difficult. This critical review gathers, for the first time, information concerning normal breast and cancer biology, established and emerging methods for screening and diagnosis, staging and grading, molecular and genetic biomarkers. Our purpose is to address key interdisciplinary information about these methods for physicians and scientists. Only the multidisciplinary interaction and communication between scientists, health care professionals, technical experts and patients will lead to the development of better detection tools and methods for an improved screening and early diagnosis.
Collapse
Affiliation(s)
- Diego Barba
- Escuela de Medicina, Universidad San Francisco de Quito USFQ, Quito, Ecuador; Instituto de Investigaciones en Biomedicina, Universidad San Francisco de Quito USFQ, Quito, Ecuador; Mito-Act Research Consortium, Quito, Ecuador
| | - Ariana León-Sosa
- Escuela de Medicina, Universidad San Francisco de Quito USFQ, Quito, Ecuador; Instituto de Investigaciones en Biomedicina, Universidad San Francisco de Quito USFQ, Quito, Ecuador; Mito-Act Research Consortium, Quito, Ecuador
| | - Paulina Lugo
- Hospital de los Valles HDLV, Quito, Ecuador; Fundación Ayuda Familiar y Comunitaria AFAC, Quito, Ecuador
| | - Daniela Suquillo
- Instituto de Investigaciones en Biomedicina, Universidad San Francisco de Quito USFQ, Quito, Ecuador; Mito-Act Research Consortium, Quito, Ecuador; Ingeniería en Procesos Biotecnológicos, Colegio de Ciencias Biológicas y Ambientales COCIBA, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - Fernando Torres
- Escuela de Medicina, Universidad San Francisco de Quito USFQ, Quito, Ecuador; Hospital de los Valles HDLV, Quito, Ecuador
| | - Frederic Surre
- University of Glasgow, James Watt School of Engineering, Glasgow, G12 8QQ, United Kingdom
| | - Lionel Trojman
- LISITE, Isep, 75006, Paris, France; Universidad San Francisco de Quito USFQ, Colegio de Ciencias e Ingenierías Politécnico - USFQ, Instituto de Micro y Nanoelectrónica, IMNE, USFQ, Quito, Ecuador
| | - Andrés Caicedo
- Escuela de Medicina, Universidad San Francisco de Quito USFQ, Quito, Ecuador; Instituto de Investigaciones en Biomedicina, Universidad San Francisco de Quito USFQ, Quito, Ecuador; Mito-Act Research Consortium, Quito, Ecuador; Sistemas Médicos SIME, Universidad San Francisco de Quito USFQ, Quito, Ecuador.
| |
Collapse
|
168
|
Zaghdoudi S, Decaup E, Belhabib I, Samain R, Cassant‐Sourdy S, Rochotte J, Brunel A, Schlaepfer D, Cros J, Neuzillet C, Strehaiano M, Alard A, Tomasini R, Rajeeve V, Perraud A, Mathonnet M, Pearce OMT, Martineau Y, Pyronnet S, Bousquet C, Jean C. FAK activity in cancer-associated fibroblasts is a prognostic marker and a druggable key metastatic player in pancreatic cancer. EMBO Mol Med 2020; 12:e12010. [PMID: 33025708 PMCID: PMC7645544 DOI: 10.15252/emmm.202012010] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 09/04/2020] [Accepted: 09/08/2020] [Indexed: 12/22/2022] Open
Abstract
Cancer-associated fibroblasts (CAFs) are considered the most abundant type of stromal cells in pancreatic ductal adenocarcinoma (PDAC), playing a critical role in tumour progression and chemoresistance; however, a druggable target on CAFs has not yet been identified. Here we report that focal adhesion kinase (FAK) activity (evaluated based on 397 tyrosine phosphorylation level) in CAFs is highly increased compared to its activity in fibroblasts from healthy pancreas. Fibroblastic FAK activity is an independent prognostic marker for disease-free and overall survival of PDAC patients (cohort of 120 PDAC samples). Genetic inactivation of FAK within fibroblasts (FAK kinase-dead, KD) reduces fibrosis and immunosuppressive cell number within primary tumours and dramatically decreases tumour spread. FAK pharmacologic or genetic inactivation reduces fibroblast migration/invasion, decreases extracellular matrix (ECM) expression and deposition by CAFs, modifies ECM track generation and negatively impacts M2 macrophage polarization and migration. Thus, FAK activity within CAFs appears as an independent PDAC prognostic marker and a druggable driver of tumour cell invasion.
Collapse
Affiliation(s)
- Sonia Zaghdoudi
- Cancer Research Center of Toulouse (CRCT)team 6 “Protein synthesis & secretion in carcinogenesis”Equipe labellisée Ligue Contre Le CancerLabex TOUCANINSERM UMR 1037‐ University Toulouse III Paul SabatierToulouseFrance
| | - Emilie Decaup
- Cancer Research Center of Toulouse (CRCT)team 6 “Protein synthesis & secretion in carcinogenesis”Equipe labellisée Ligue Contre Le CancerLabex TOUCANINSERM UMR 1037‐ University Toulouse III Paul SabatierToulouseFrance
| | - Ismahane Belhabib
- Cancer Research Center of Toulouse (CRCT)team 6 “Protein synthesis & secretion in carcinogenesis”Equipe labellisée Ligue Contre Le CancerLabex TOUCANINSERM UMR 1037‐ University Toulouse III Paul SabatierToulouseFrance
| | - Rémi Samain
- Cancer Research Center of Toulouse (CRCT)team 6 “Protein synthesis & secretion in carcinogenesis”Equipe labellisée Ligue Contre Le CancerLabex TOUCANINSERM UMR 1037‐ University Toulouse III Paul SabatierToulouseFrance
| | - Stéphanie Cassant‐Sourdy
- Cancer Research Center of Toulouse (CRCT)team 6 “Protein synthesis & secretion in carcinogenesis”Equipe labellisée Ligue Contre Le CancerLabex TOUCANINSERM UMR 1037‐ University Toulouse III Paul SabatierToulouseFrance
| | - Julia Rochotte
- Cancer Research Center of Toulouse (CRCT)team 6 “Protein synthesis & secretion in carcinogenesis”Equipe labellisée Ligue Contre Le CancerLabex TOUCANINSERM UMR 1037‐ University Toulouse III Paul SabatierToulouseFrance
| | - Alexia Brunel
- Cancer Research Center of Toulouse (CRCT)team 6 “Protein synthesis & secretion in carcinogenesis”Equipe labellisée Ligue Contre Le CancerLabex TOUCANINSERM UMR 1037‐ University Toulouse III Paul SabatierToulouseFrance
| | - David Schlaepfer
- Department of Reproductive Medicine Moores Cancer CenterUniversity of California San DiegoLa JollaCAUSA
| | - Jérome Cros
- Department of PathologyBeaujon HospitalINSERM U1149ClichyFrance
| | - Cindy Neuzillet
- Medical Oncology DepartmentCurie InstituteVersailles Saint‐Quentin UniversitySaint‐CloudFrance
| | - Manon Strehaiano
- Cancer Research Center of Toulouse (CRCT)team 6 “Protein synthesis & secretion in carcinogenesis”Equipe labellisée Ligue Contre Le CancerLabex TOUCANINSERM UMR 1037‐ University Toulouse III Paul SabatierToulouseFrance
| | - Amandine Alard
- Cancer Research Center of Toulouse (CRCT)team 6 “Protein synthesis & secretion in carcinogenesis”Equipe labellisée Ligue Contre Le CancerLabex TOUCANINSERM UMR 1037‐ University Toulouse III Paul SabatierToulouseFrance
| | | | - Vinothini Rajeeve
- Centre for Genomics and Computational BiologyBarts Cancer InstituteQueen Mary University of LondonLondonUK
| | - Aurélie Perraud
- EA 3842 Laboratory, Medicine and Pharmacy FacultiesLimoges UniversityLimogesFrance
| | - Muriel Mathonnet
- EA 3842 Laboratory, Medicine and Pharmacy FacultiesLimoges UniversityLimogesFrance
| | - Oliver MT Pearce
- Centre for Tumour MicroenvironmentBarts Cancer InstituteLondonUK
| | - Yvan Martineau
- Cancer Research Center of Toulouse (CRCT)team 6 “Protein synthesis & secretion in carcinogenesis”Equipe labellisée Ligue Contre Le CancerLabex TOUCANINSERM UMR 1037‐ University Toulouse III Paul SabatierToulouseFrance
| | - Stéphane Pyronnet
- Cancer Research Center of Toulouse (CRCT)team 6 “Protein synthesis & secretion in carcinogenesis”Equipe labellisée Ligue Contre Le CancerLabex TOUCANINSERM UMR 1037‐ University Toulouse III Paul SabatierToulouseFrance
| | - Corinne Bousquet
- Cancer Research Center of Toulouse (CRCT)team 6 “Protein synthesis & secretion in carcinogenesis”Equipe labellisée Ligue Contre Le CancerLabex TOUCANINSERM UMR 1037‐ University Toulouse III Paul SabatierToulouseFrance
| | - Christine Jean
- Cancer Research Center of Toulouse (CRCT)team 6 “Protein synthesis & secretion in carcinogenesis”Equipe labellisée Ligue Contre Le CancerLabex TOUCANINSERM UMR 1037‐ University Toulouse III Paul SabatierToulouseFrance
| |
Collapse
|
169
|
Etzerodt A, Moulin M, Doktor TK, Delfini M, Mossadegh-Keller N, Bajenoff M, Sieweke MH, Moestrup SK, Auphan-Anezin N, Lawrence T. Tissue-resident macrophages in omentum promote metastatic spread of ovarian cancer. J Exp Med 2020; 217:133611. [PMID: 31951251 PMCID: PMC7144521 DOI: 10.1084/jem.20191869] [Citation(s) in RCA: 201] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/23/2019] [Accepted: 12/16/2019] [Indexed: 12/17/2022] Open
Abstract
Experimental and clinical evidence suggests that tumor-associated macrophages (TAMs) play important roles in cancer progression. Here, we have characterized the ontogeny and function of TAM subsets in a mouse model of metastatic ovarian cancer that is representative for visceral peritoneal metastasis. We show that the omentum is a critical premetastatic niche for development of invasive disease in this model and define a unique subset of CD163+ Tim4+ resident omental macrophages responsible for metastatic spread of ovarian cancer cells. Transcriptomic analysis showed that resident CD163+ Tim4+ omental macrophages were phenotypically distinct and maintained their resident identity during tumor growth. Selective depletion of CD163+ Tim4+ macrophages in omentum using genetic and pharmacological tools prevented tumor progression and metastatic spread of disease. These studies describe a specific role for tissue-resident macrophages in the invasive progression of metastatic ovarian cancer. The molecular pathways of cross-talk between tissue-resident macrophages and disseminated cancer cells may represent new targets to prevent metastasis and disease recurrence.
Collapse
Affiliation(s)
- Anders Etzerodt
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France.,Department of Biomedicine, University of Aarhus, Aarhus, Denmark
| | - Morgane Moulin
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France.,Centre for Inflammation Biology and Cancer Immunology, School of Immunology & Microbial Sciences, King's College London, London, UK
| | - Thomas Koed Doktor
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | | | | | - Marc Bajenoff
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | - Michael H Sieweke
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France.,Centre for Regenerative Therapies, TU Dresden, Dresden, Germany
| | - Søren Kragh Moestrup
- Department of Biomedicine, University of Aarhus, Aarhus, Denmark.,Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | | - Toby Lawrence
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France.,Centre for Inflammation Biology and Cancer Immunology, School of Immunology & Microbial Sciences, King's College London, London, UK.,Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
170
|
Modulation of Immune Infiltration of Ovarian Cancer Tumor Microenvironment by Specific Subpopulations of Fibroblasts. Cancers (Basel) 2020; 12:cancers12113184. [PMID: 33138184 PMCID: PMC7692816 DOI: 10.3390/cancers12113184] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 10/20/2020] [Accepted: 10/24/2020] [Indexed: 12/15/2022] Open
Abstract
Tumor immune infiltration plays a key role in the progression of solid tumors, including ovarian cancer, and immunotherapies are rapidly emerging as effective treatment modalities. However, the role of cancer-associated fibroblasts (CAFs), a predominant stromal constituent, in determining the tumor-immune microenvironment and modulating efficacy of immunotherapies remains poorly understood. We have conducted an extensive bioinformatic analysis of our and other publicly available ovarian cancer datasets (GSE137237, GSE132289 and GSE71340), to determine the correlation of fibroblast subtypes within the tumor microenvironment (TME) with the characteristics of tumor-immune infiltration. We identified (1) four functional modules of CAFs in ovarian cancer that are associated with the TME and metastasis of ovarian cancer, (2) immune-suppressive function of the collagen 1,3,5-expressing CAFs in primary ovarian cancer and omental metastases, and (3) consistent positive correlations between the functional modules of CAFs with anti-immune response genes and negative correlation with pro-immune response genes. Our study identifies a specific fibroblast subtype, fibroblast functional module (FFM)2, in the ovarian cancer tumor microenvironment that can potentially modulate a tumor-promoting immune microenvironment, which may be detrimental toward the effectiveness of ovarian cancer immunotherapies.
Collapse
|
171
|
Modelling Pancreatic Neuroendocrine Cancer: From Bench Side to Clinic. Cancers (Basel) 2020; 12:cancers12113170. [PMID: 33126717 PMCID: PMC7693644 DOI: 10.3390/cancers12113170] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/20/2020] [Accepted: 10/23/2020] [Indexed: 12/11/2022] Open
Abstract
Pancreatic neuroendocrine tumours (pNETs) are a heterogeneous group of epithelial tumours with neuroendocrine differentiation. Although rare (incidence of <1 in 100,000), they are the second most common group of pancreatic neoplasms after pancreatic ductal adenocarcinoma (PDAC). pNET incidence is however on the rise and patient outcomes, although variable, have been linked with 5-year survival rates as low as 40%. Improvement of diagnostic and treatment modalities strongly relies on disease models that reconstruct the disease ex vivo. A key constraint in pNET research, however, is the absence of human pNET models that accurately capture the original tumour phenotype. In attempts to more closely mimic the disease in its native environment, three-dimensional culture models as well as in vivo models, such as genetically engineered mouse models (GEMMs), have been developed. Despite adding significant contributions to our understanding of more complex biological processes associated with the development and progression of pNETs, factors such as ethical considerations and low rates of clinical translatability limit their use. Furthermore, a role for the site-specific extracellular matrix (ECM) in disease development and progression has become clear. Advances in tissue engineering have enabled the use of tissue constructs that are designed to establish disease ex vivo within a close to native ECM that can recapitulate tumour-associated tissue remodelling. Yet, such advanced models for studying pNETs remain underdeveloped. This review summarises the most clinically relevant disease models of pNETs currently used, as well as future directions for improved modelling of the disease.
Collapse
|
172
|
Chin MW, Norman MDA, Gentleman E, Coppens MO, Day RM. A Hydrogel-Integrated Culture Device to Interrogate T Cell Activation with Physicochemical Cues. ACS APPLIED MATERIALS & INTERFACES 2020; 12:47355-47367. [PMID: 33027591 PMCID: PMC7586298 DOI: 10.1021/acsami.0c16478] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
The recent rise of adoptive T cell therapy (ATCT) as a promising cancer immunotherapy has triggered increased interest in therapeutic T cell bioprocessing. T cell activation is a critical processing step and is known to be modulated by physical parameters, such as substrate stiffness. Nevertheless, relatively little is known about how biophysical factors regulate immune cells, such as T cells. Understanding how T cell activation is modulated by physical and biochemical cues may offer novel methods to control cell behavior for therapeutic cell processing. Inspired by T cell mechanosensitivity, we developed a multiwell, reusable, customizable, two-dimensional (2D) polyacrylamide (PA) hydrogel-integrated culture device to study the physicochemical stimulation of Jurkat T cells. Substrate stiffness and ligand density were tuned by concentrations of the hydrogel cross-linker and antibody in the coating solution, respectively. We cultured Jurkat T cells on 2D hydrogels of different stiffnesses that presented surface-immobilized stimulatory antibodies against CD3 and CD28 and demonstrated that Jurkat T cells stimulated by stiff hydrogels (50.6 ± 15.1 kPa) exhibited significantly higher interleukin-2 (IL-2) secretion, but lower proliferation, than those stimulated by softer hydrogels (7.1 ± 0.4 kPa). In addition, we found that increasing anti-CD3 concentration from 10 to 30 μg/mL led to a significant increase in IL-2 secretion from cells stimulated on 7.1 ± 0.4 and 9.3 ± 2.4 kPa gels. Simultaneous tuning of substrate stiffness and stimulatory ligand density showed that the two parameters synergize (two-way ANOVA interaction effect: p < 0.001) to enhance IL-2 secretion. Our results demonstrate the importance of physical parameters in immune cell stimulation and highlight the potential of designing future immunostimulatory biomaterials that are mechanically tailored to balance stimulatory strength and downstream proliferative capacity of therapeutic T cells.
Collapse
Affiliation(s)
- Matthew
H. W. Chin
- Centre
for Precision Healthcare, Division of Medicine, University College London, London WC1E 6BT, United Kingdom
- Centre
for Nature Inspired Engineering, University
College London, London WC1E 6BT, United Kingdom
| | - Michael D. A. Norman
- Centre
for Craniofacial and Regenerative Biology, King’s College London, London SE1 9RT, United Kingdom
| | - Eileen Gentleman
- Centre
for Craniofacial and Regenerative Biology, King’s College London, London SE1 9RT, United Kingdom
| | - Marc-Olivier Coppens
- Centre
for Nature Inspired Engineering, University
College London, London WC1E 6BT, United Kingdom
- Department
of Chemical Engineering, University College
London, London WC1E 7JE, United Kingdom
| | - Richard M. Day
- Centre
for Precision Healthcare, Division of Medicine, University College London, London WC1E 6BT, United Kingdom
- Centre
for Nature Inspired Engineering, University
College London, London WC1E 6BT, United Kingdom
| |
Collapse
|
173
|
Huang YL, Liang CY, Ritz D, Coelho R, Septiadi D, Estermann M, Cumin C, Rimmer N, Schötzau A, Núñez López M, Fedier A, Konantz M, Vlajnic T, Calabrese D, Lengerke C, David L, Rothen-Rutishauser B, Jacob F, Heinzelmann-Schwarz V. Collagen-rich omentum is a premetastatic niche for integrin α2-mediated peritoneal metastasis. eLife 2020; 9:59442. [PMID: 33026975 PMCID: PMC7541088 DOI: 10.7554/elife.59442] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 09/15/2020] [Indexed: 02/07/2023] Open
Abstract
The extracellular matrix (ECM) plays critical roles in tumor progression and metastasis. However, the contribution of ECM proteins to early metastatic onset in the peritoneal cavity remains unexplored. Here, we suggest a new route of metastasis through the interaction of integrin alpha 2 (ITGA2) with collagens enriched in the tumor coinciding with poor outcome in patients with ovarian cancer. Using multiple gene-edited cell lines and patient-derived samples, we demonstrate that ITGA2 triggers cancer cell adhesion to collagen, promotes cell migration, anoikis resistance, mesothelial clearance, and peritoneal metastasis in vitro and in vivo. Mechanistically, phosphoproteomics identify an ITGA2-dependent phosphorylation of focal adhesion kinase and mitogen-activated protein kinase pathway leading to enhanced oncogenic properties. Consequently, specific inhibition of ITGA2-mediated cancer cell-collagen interaction or targeting focal adhesion signaling may present an opportunity for therapeutic intervention of metastatic spread in ovarian cancer.
Collapse
Affiliation(s)
- Yen-Lin Huang
- Ovarian Cancer Research, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Ching-Yeu Liang
- Ovarian Cancer Research, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Danilo Ritz
- Proteomics core facility, Biozentrum, University of Basel, Basel, Switzerland
| | - Ricardo Coelho
- Differentiation and Cancer group, Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal.,Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal.,Faculty of Medicine, University of Porto, Porto, Portugal
| | - Dedy Septiadi
- Adolphe Merkle Institute, University of Fribourg, Fribourg, Switzerland
| | - Manuela Estermann
- Adolphe Merkle Institute, University of Fribourg, Fribourg, Switzerland
| | - Cécile Cumin
- Ovarian Cancer Research, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Natalie Rimmer
- Ovarian Cancer Research, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Andreas Schötzau
- Ovarian Cancer Research, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Mónica Núñez López
- Ovarian Cancer Research, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - André Fedier
- Ovarian Cancer Research, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Martina Konantz
- Stem Cells and Hematopoiesis, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Tatjana Vlajnic
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Diego Calabrese
- Histology Core Facility, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Claudia Lengerke
- Stem Cells and Hematopoiesis, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland.,Department of Internal Medicine, Internal Medicine II, Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Leonor David
- Differentiation and Cancer group, Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal.,Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal.,Faculty of Medicine, University of Porto, Porto, Portugal
| | | | - Francis Jacob
- Ovarian Cancer Research, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Viola Heinzelmann-Schwarz
- Ovarian Cancer Research, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland.,Gynecological Cancer Center, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
174
|
Hedegaard CL, Redondo-Gómez C, Tan BY, Ng KW, Loessner D, Mata A. Peptide-protein coassembling matrices as a biomimetic 3D model of ovarian cancer. SCIENCE ADVANCES 2020; 6:eabb3298. [PMID: 33008910 PMCID: PMC7852381 DOI: 10.1126/sciadv.abb3298] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 08/20/2020] [Indexed: 05/04/2023]
Abstract
Bioengineered three-dimensional (3D) matrices expand our experimental repertoire to study tumor growth and progression in a biologically relevant, yet controlled, manner. Here, we used peptide amphiphiles (PAs) to coassemble with and organize extracellular matrix (ECM) proteins producing tunable 3D models of the tumor microenvironment. The matrix was designed to mimic physical and biomolecular features of tumors present in patients. We included specific epitopes, PA nanofibers, and ECM macromolecules for the 3D culture of human ovarian cancer, endothelial, and mesenchymal stem cells. The multicellular constructs supported the formation of tumor spheroids with extensive F-actin networks surrounding the spheroids, enabling cell-cell communication, and comparative cell-matrix interactions and encapsulation response to those observed in Matrigel. We conducted a proof-of-concept study with clinically used chemotherapeutics to validate the functionality of the multicellular constructs. Our study demonstrates that peptide-protein coassembling matrices serve as a defined model of the multicellular tumor microenvironment of primary ovarian tumors.
Collapse
Affiliation(s)
- Clara Louise Hedegaard
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, E1 4NS London, UK
- Institute of Bioengineering, Queen Mary University of London, Mile End Road, E1 4NS London, UK
| | - Carlos Redondo-Gómez
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, E1 4NS London, UK
- Institute of Bioengineering, Queen Mary University of London, Mile End Road, E1 4NS London, UK
| | - Bee Yi Tan
- School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Kee Woei Ng
- School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
- Environmental Chemistry and Materials Centre, Nanyang Environment & Water Research Institute, Singapore 637141, Singapore
- Skin Research Institute of Singapore, Singapore 138648, Singapore
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA 02115, USA
| | - Daniela Loessner
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, EC1M 6BQ London, UK
- Department of Chemical Engineering and Department of Materials Science and Engineering, Faculty of Engineering, Monash University, Melbourne, VIC 3800, Australia
- Department of Anatomy and Developmental Biology, Faculty of Medicine, Monash University, Melbourne, VIC 3800, Australia
| | - Alvaro Mata
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, E1 4NS London, UK.
- Institute of Bioengineering, Queen Mary University of London, Mile End Road, E1 4NS London, UK
- School of Pharmacy, University of Nottingham, NG7 2RD Nottingham, UK
- Department of Chemical and Environmental Engineering, University of Nottingham, NG7 2RD Nottingham, UK
- Biodiscovery Institute, University of Nottingham, NG7 2RD Nottingham, UK
| |
Collapse
|
175
|
Zou S, Tong Q, Liu B, Huang W, Tian Y, Fu X. Targeting STAT3 in Cancer Immunotherapy. Mol Cancer 2020; 19:145. [PMID: 32972405 PMCID: PMC7513516 DOI: 10.1186/s12943-020-01258-7] [Citation(s) in RCA: 653] [Impact Index Per Article: 130.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 09/04/2020] [Indexed: 02/08/2023] Open
Abstract
As a point of convergence for numerous oncogenic signaling pathways, signal transducer and activator of transcription 3 (STAT3) is central in regulating the anti-tumor immune response. STAT3 is broadly hyperactivated both in cancer and non-cancerous cells within the tumor ecosystem and plays important roles in inhibiting the expression of crucial immune activation regulators and promoting the production of immunosuppressive factors. Therefore, targeting the STAT3 signaling pathway has emerged as a promising therapeutic strategy for numerous cancers. In this review, we outline the importance of STAT3 signaling pathway in tumorigenesis and its immune regulation, and highlight the current status for the development of STAT3-targeting therapeutic approaches. We also summarize and discuss recent advances in STAT3-based combination immunotherapy in detail. These endeavors provide new insights into the translational application of STAT3 in cancer and may contribute to the promotion of more effective treatments toward malignancies.
Collapse
Affiliation(s)
- Sailan Zou
- Division of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, Sichuan, China
| | - Qiyu Tong
- Division of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, Sichuan, China
| | - Bowen Liu
- College of Life Sciences, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Wei Huang
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yan Tian
- Division of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, Sichuan, China.
| | - Xianghui Fu
- Division of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
176
|
Lin H, Wang J, Wen X, Wen Q, Huang S, Mai Z, Lu L, Liang X, Pan H, Li S, He Y, Ma H. A prognosis-predictive nomogram of ovarian cancer with two immune-related genes: CDC20B and PNPLA5. Oncol Lett 2020; 20:204. [PMID: 32963610 DOI: 10.3892/ol.2020.12067] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 07/29/2020] [Indexed: 12/12/2022] Open
Abstract
Ovarian carcinoma (OV) is one of the most lethal gynecological malignancies globally, and the overall 5-year survival rate of OV was 47% in 2018 according to American data. To increase the survival rate of patients with OV, many researchers have sought to identify biomarkers that act as both prognosis-predictive markers and therapy targets. However, most of these have not been suitable for clinical application. The present study aimed at constructing a predictive prognostic nomogram of OV using the genes identified by combining The Cancer Genome Atlas (TCGA) dataset for OV with the immune score calculated by the Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data algorithm. Firstly, the algorithm was used to calculate the immune score of patients with OV in the TCGA-OV dataset. Secondly, differentially expressed genes (DEGs) between low and high immune score tissues were identified, and Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analysis was performed to predict the functions of these DEGs. Thirdly, univariate, multivariate and Lasso Cox's regression analyses were carried out step by step, and six prognosis-related DEGs were identified. Then, Kaplan-Myer survival curves were generated for these genes and validated by comparing their expression levels to further narrow the range of DEGs and to calculate the risk score. Two genes were identified, cell division cycle 20B and patatin-like phospholipase domain containing 5, which were both shown to have higher expression levels in OV tissues and to be significantly associated with the prognosis of OV. Next, a nomogram was created using these two genes and age, and using the receiver operating characteristic (ROC) curve and calibration curve, the effectiveness of the nomogram was validated. Finally, an external validation was conducted for this nomogram. The ROC showed that the areas under the curve (AUCs) of the 3- and 5-year overall survival predictions for the nomogram were 0.678 and 0.62, respectively. Moreover, the ROC of the external validation model showed that the AUCs of the 3- and 5-year were 0.699 and 0.643, respectively, demonstrating the effectiveness of the generated nomogram. In conclusion, the present study has identified two immune-related genes as biomarkers that reliably predict overall survival in OV. These biomarkers might also be potential molecular targets of immune therapy to treat patients with OV.
Collapse
Affiliation(s)
- Han Lin
- Department of Gynecology of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510000, P.R. China
| | - Jiamin Wang
- Department of Urology and Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510000, P.R. China
| | - Xiaohui Wen
- Department of Gynecology of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510000, P.R. China
| | - Qidan Wen
- Department of Gynecology of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510000, P.R. China
| | - Shiya Huang
- Department of Gynecology of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510000, P.R. China
| | - Zhefen Mai
- Department of Gynecology of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510000, P.R. China
| | - Lingjing Lu
- Department of Gynecology of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510000, P.R. China
| | - Xingyan Liang
- Department of Gynecology of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510000, P.R. China
| | - Haixia Pan
- Department of Gynecology of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510000, P.R. China
| | - Shuna Li
- Department of Gynecology of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510000, P.R. China
| | - Yuhong He
- Department of Gynecology of Traditional Chinese Medicine, The Affiliated Ruikang Hospital of Guangxi University of Chinese Medicine, Nanning, Guangxi 530000, P.R. China
| | - Hongxia Ma
- Department of Gynecology of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510000, P.R. China
| |
Collapse
|
177
|
Omental macrophages secrete chemokine ligands that promote ovarian cancer colonization of the omentum via CCR1. Commun Biol 2020; 3:524. [PMID: 32963283 PMCID: PMC7508838 DOI: 10.1038/s42003-020-01246-z] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 08/06/2020] [Indexed: 12/26/2022] Open
Abstract
The omentum is the most common site of ovarian cancer metastasis. Immune cell clusters called milky spots are found throughout the omentum. It is however unknown if these immune cells contribute to ovarian cancer metastasis. Here we report that omental macrophages promote the migration and colonization of ovarian cancer cells to the omentum through the secretion of chemokine ligands that interact with chemokine receptor 1 (CCR1). We found that depletion of macrophages reduces ovarian cancer colonization of the omentum. RNA-sequencing of macrophages isolated from mouse omentum and mesenteric adipose tissue revealed a specific enrichment of chemokine ligand CCL6 in omental macrophages. CCL6 and the human homolog CCL23 were both necessary and sufficient to promote ovarian cancer migration by activating ERK1/2 and PI3K pathways. Importantly, inhibition of CCR1 reduced ovarian cancer colonization. These findings demonstrate a critical mechanism of omental macrophage induced colonization by ovarian cancer cells via CCR1 signaling. Krishnan et al. find that CCR1 ligands CCL6 and CCL23 secreted by murine and human macrophages, respectively, enhance metastatic colonization of ovarian cancer cells to the omentum in manner dependent on chemokine receptor 1 (CCR1). This study suggests that targeting CCR1 or CCL23 in ovarian cancer may be a therapeutic strategy.
Collapse
|
178
|
Xiong X, Wen YA, Fairchild R, Zaytseva YY, Weiss HL, Evers BM, Gao T. Upregulation of CPT1A is essential for the tumor-promoting effect of adipocytes in colon cancer. Cell Death Dis 2020; 11:736. [PMID: 32913185 PMCID: PMC7484798 DOI: 10.1038/s41419-020-02936-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 08/21/2020] [Accepted: 08/27/2020] [Indexed: 01/28/2023]
Abstract
Colon tumors grow in an adipose tissue-enriched microenvironment. Locally advanced colon cancers often invade into surrounding adipose tissue with a direct contact with adipocytes. We have previously shown that adipocytes promote tumor growth by modulating cellular metabolism. Here we demonstrate that carnitine palmitoyltransferase I (CPT1A), a key enzyme controlling fatty acid oxidation (FAO), was upregulated in colon cancer cells upon exposure to adipocytes or fatty acids. In addition, CPT1A expression was increased in invasive tumor cells within the adipose tissue compared to tumors without direct contact with adipocytes. Silencing CPT1A abolished the protective effect provided by fatty acids against nutrient deprivation and reduced tumor organoid formation in 3D culture and the expression of genes associated with cancer stem cells downstream of Wnt/β-catenin. Mechanistically, CPT1A-dependent FAO promoted the acetylation and nuclear translocation of β-catenin. Furthermore, knockdown of CPT1A blocked the tumor-promoting effect of adipocytes in vivo and inhibited xenograft tumor initiation. Taken together, our findings identify CPT1A-depedent FAO as an essential metabolic pathway that enables the interaction between adipocytes and colon cancer cells.
Collapse
Affiliation(s)
- Xiaopeng Xiong
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40536-0679, USA
| | - Yang-An Wen
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40536-0679, USA
| | - Rachelle Fairchild
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40536-0679, USA
| | - Yekaterina Y Zaytseva
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, 40536-0679, USA
| | - Heidi L Weiss
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40536-0679, USA
| | - B Mark Evers
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40536-0679, USA
- Department of Surgery, University of Kentucky, Lexington, KY, 40536-0679, USA
| | - Tianyan Gao
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40536-0679, USA.
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40536-0679, USA.
| |
Collapse
|
179
|
Liu M, Silva-Sanchez A, Randall TD, Meza-Perez S. Specialized immune responses in the peritoneal cavity and omentum. J Leukoc Biol 2020; 109:717-729. [PMID: 32881077 DOI: 10.1002/jlb.5mir0720-271rr] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 07/13/2020] [Accepted: 07/24/2020] [Indexed: 12/15/2022] Open
Abstract
The peritoneal cavity is a fluid filled space that holds most of the abdominal organs, including the omentum, a visceral adipose tissue that contains milky spots or clusters of leukocytes that are organized similar to those in conventional lymphoid tissues. A unique assortment of leukocytes patrol the peritoneal cavity and migrate in and out of the milky spots, where they encounter Ags or pathogens from the peritoneal fluid and respond accordingly. The principal role of leukocytes in the peritoneal cavity is to preserve tissue homeostasis and secure tissue repair. However, when peritoneal homeostasis is disturbed by inflammation, infection, obesity, or tumor metastasis, specialized fibroblastic stromal cells and mesothelial cells in the omentum regulate the recruitment of peritoneal leukocytes and steer their activation in unique ways. In this review, the types of cells that reside in the peritoneal cavity, the role of the omentum in their maintenance and activation, and how these processes function in response to pathogens and malignancy will be discussed.
Collapse
Affiliation(s)
- Mingyong Liu
- Department of Medicine, Division of Clinical Immunology and Rheumatology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Aaron Silva-Sanchez
- Department of Medicine, Division of Clinical Immunology and Rheumatology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Troy D Randall
- Department of Medicine, Division of Clinical Immunology and Rheumatology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Selene Meza-Perez
- Department of Medicine, Division of Clinical Immunology and Rheumatology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
180
|
Honselmann KC, Finetti P, Birnbaum DJ, Monsalve CS, Wellner UF, Begg SKS, Nakagawa A, Hank T, Li A, Goldsworthy MA, Sharma H, Bertucci F, Birnbaum D, Tai E, Ligorio M, Ting DT, Schilling O, Biniossek ML, Bronsert P, Ferrone CR, Keck T, Mino-Kenudson M, Lillemoe KD, Warshaw AL, Fernández-Del Castillo C, Liss AS. Neoplastic-Stromal Cell Cross-talk Regulates Matrisome Expression in Pancreatic Cancer. Mol Cancer Res 2020; 18:1889-1902. [PMID: 32873625 DOI: 10.1158/1541-7786.mcr-20-0439] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 05/28/2020] [Accepted: 08/25/2020] [Indexed: 11/16/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by a highly desmoplastic reaction, warranting intense cancer-stroma communication. In this study, we interrogated the contribution of the BET family of chromatin adaptors to the cross-talk between PDAC cells and the tumor stroma. Short-term treatment of orthotopic xenograft tumors with CPI203, a small-molecule inhibitor of BET proteins, resulted in broad changes in the expression of genes encoding components of the extracellular matrix (matrisome) in both cancer and stromal cells. Remarkably, more than half of matrisome genes were expressed by cancer cells. In vitro cocultures of PDAC cells and cancer-associated fibroblasts (CAF) demonstrated that matrisome expression was regulated by BET-dependent cancer-CAF cross-talk. Disrupting this cross-talk in vivo resulted in diminished growth of orthotopic patient-derived xenograft tumors, reduced proliferation of cancer cells, and changes in collagen structure consistent with that of patients who experienced better survival. Examination of matrisome gene expression in publicly available data sets of 573 PDAC tumors identified a 65-gene signature that was able to distinguish long- and short-term PDAC survivors. Importantly, the expression of genes predictive of short-term survival was diminished in the cancer cells of orthotopic xenograft tumors of mice treated with CPI203. Taken together, these results demonstrate that inhibiting the activity BET proteins results in transcriptional and structural differences in the matrisome are associated with better patient survival. IMPLICATIONS: These studies highlight the biological relevance of the matrisome program in PDAC and suggest targeting of epigenetically driven tumor-stroma cross-talk as a potential therapeutic avenue.
Collapse
Affiliation(s)
- Kim C Honselmann
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Pascal Finetti
- Laboratoire d'Oncologie Prédictive, Centre de Recherche en Cancérologie de Marseille, INSERM UMR1068, CNRS UMR7258, Aix-Marseille University, Marseille, France
| | - David J Birnbaum
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.,Laboratoire d'Oncologie Prédictive, Centre de Recherche en Cancérologie de Marseille, INSERM UMR1068, CNRS UMR7258, Aix-Marseille University, Marseille, France.,Département de Chirurgie Générale et Viscérale, AP-HM, Marseille, France
| | - Christian S Monsalve
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ulrich F Wellner
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Sebastian K S Begg
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Akifumi Nakagawa
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Thomas Hank
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Annie Li
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Mathew A Goldsworthy
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Himanshu Sharma
- Partners Healthcare Personalized Medicine Center, Cambridge, Massachusetts
| | - François Bertucci
- Laboratoire d'Oncologie Prédictive, Centre de Recherche en Cancérologie de Marseille, INSERM UMR1068, CNRS UMR7258, Aix-Marseille University, Marseille, France.,Département d'Oncologie Médicale, Institut Paoli-Calmettes, Marseille, France
| | - Daniel Birnbaum
- Laboratoire d'Oncologie Prédictive, Centre de Recherche en Cancérologie de Marseille, INSERM UMR1068, CNRS UMR7258, Aix-Marseille University, Marseille, France
| | - Eric Tai
- MGH Cancer Research Center, Harvard Medical School, Boston, Massachusetts
| | - Matteo Ligorio
- MGH Cancer Research Center, Harvard Medical School, Boston, Massachusetts
| | - David T Ting
- MGH Cancer Research Center, Harvard Medical School, Boston, Massachusetts
| | - Oliver Schilling
- Institute of Surgical Pathology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK) and Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martin L Biniossek
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Peter Bronsert
- Institute of Surgical Pathology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK) and Cancer Research Center (DKFZ), Heidelberg, Germany.,Comprehensive Cancer Center Freiburg, Medical Center - University of Freiburg, Freiburg, Germany
| | - Cristina R Ferrone
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Tobias Keck
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Mari Mino-Kenudson
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Keith D Lillemoe
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Andrew L Warshaw
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | | | - Andrew S Liss
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
181
|
Wen YC, Lin YW, Chu CY, Yang YC, Yang SF, Liu YF, Hsiao M, Lee WJ, Chien MH. Melatonin-triggered post-transcriptional and post-translational modifications of ADAMTS1 coordinately retard tumorigenesis and metastasis of renal cell carcinoma. J Pineal Res 2020; 69:e12668. [PMID: 32408377 DOI: 10.1111/jpi.12668] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 03/25/2020] [Accepted: 05/07/2020] [Indexed: 12/27/2022]
Abstract
A disintegrin and metalloprotease with thrombospondin motifs (ADAMTS) family are widely implicated in tissue remodeling events manifested in cancer development. ADAMTS1, the most fully characterized ADAMTS, plays conflicting roles in different cancer types; however, the role of ADAMTS1 in renal cell carcinoma (RCC) remains unclear. Herein, we found that ADAMTS1 is highly expressed in RCC tissues compared to normal renal tissues, and its expression was correlated with an advanced stage and a poor prognosis of RCC patients. In vitro, we observed higher expression of ADAMTS1 in metastatic (m)RCC cells compared to primary cells, and manipulation of ADAMTS1 expression affected cell invasion and clonogenicity. Results from protease array showed that ADAMTS1 is modulated by melatonin through mechanisms independent of the MT1 receptor in mRCC cells, and overexpression of ADAMTS1 relieved the invasion/clonogenicity and growth/metastasis inhibition imposed by melatonin treatment in vitro and in an orthotopic xenograft model. The human microRNA (miR) OneArray showed that miR-181d and miR-let-7f were induced by melatonin and, respectively, targeted the 3'-UTR and non-3'-UTR of ADAMTS1 to suppress its expression and mRCC invasive ability. Clinically, RCC patients with high levels of miR-181d or miR-let-7f and a low level of ADAMTS1 had the most favorable prognoses. In addition, ubiquitin/proteasome-mediated degradation of ADAMTS1 can also be triggered by melatonin. Together, our study indicates that ADAMTS1 may be a useful biomarker for predicting RCC progression. The novel convergence between melatonin and ADAMTS1 post-transcriptional and post-translational regulation provides new insights into the role of melatonin-induced molecular regulation in suppressing RCC progression.
Collapse
Affiliation(s)
- Yu-Ching Wen
- Department of Urology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Urology, School of Medicine, College of Medicine and TMU Research Center of Urology and Kidney (TMU-RCUK), Taipei Medical University, Taipei, Taiwan
| | - Yung-Wei Lin
- Department of Urology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Urology, School of Medicine, College of Medicine and TMU Research Center of Urology and Kidney (TMU-RCUK), Taipei Medical University, Taipei, Taiwan
| | - Chih-Ying Chu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Chieh Yang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Medical Research, Tungs' Taichung MetroHarbor Hospital, Taichung, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yu-Fan Liu
- Department of Biomedical Sciences, College of Medicine Sciences and Technology, Chung Shan Medical University, Taichung, Taiwan
- Division of Allergy, Department of Pediatrics, Chung-Shan Medical University Hospital, Taichung, Taiwan
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Wei-Jiunn Lee
- Department of Urology, School of Medicine, College of Medicine and TMU Research Center of Urology and Kidney (TMU-RCUK), Taipei Medical University, Taipei, Taiwan
- Department of Medical Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Ming-Hsien Chien
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Pulmonary Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital Taipei, Taiwan
| |
Collapse
|
182
|
Arolt C, Meyer M, Hoffmann F, Wagener-Ryczek S, Schwarz D, Nachtsheim L, Beutner D, Odenthal M, Guntinas-Lichius O, Buettner R, von Eggeling F, Klußmann JP, Quaas A. Expression Profiling of Extracellular Matrix Genes Reveals Global and Entity-Specific Characteristics in Adenoid Cystic, Mucoepidermoid and Salivary Duct Carcinomas. Cancers (Basel) 2020; 12:cancers12092466. [PMID: 32878206 PMCID: PMC7564650 DOI: 10.3390/cancers12092466] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 08/25/2020] [Accepted: 08/25/2020] [Indexed: 02/06/2023] Open
Abstract
Simple Summary The extracellular matrix (ECM), an important factor in tumour metastasis and therapy resistance, has not been studied in salivary gland carcinomas (SGC), so far. In this retrospective study, we profiled the RNA expression of 28 ECM-related genes in 11 adenoid cystic (AdCy), 14 mucoepidermoid (MuEp) and 9 salivary duct carcinomas (SaDu). Also, we validated our results in a multimodal approach. MuEp and SaDu shared a common gene signature involving an overexpression of COL11A1. In contrast, nonhierarchical clustering revealed a more specific gene expression pattern for AdCy, characterized by overexpression of COL27A1. In situ studies at RNA level indicated that in AdCy, ECM production results from tumour cells and not from cancer-associated fibroblasts as is the case in MuEp and SaDu. For the first time, we characterized the ECM composition in SGC and identified several differentially expressed genes, which are potential therapeutic targets. Abstract The composition of the extracellular matrix (ECM) plays a pivotal role in tumour initiation, metastasis and therapy resistance. Until now, the ECM composition of salivary gland carcinomas (SGC) has not been studied. We quantitatively analysed the mRNA of 28 ECM-related genes of 34 adenoid cystic (AdCy; n = 11), mucoepidermoid (MuEp; n = 14) and salivary duct carcinomas (SaDu; n = 9). An incremental overexpression of six collagens (including COL11A1) and four glycoproteins from MuEp and SaDu suggested a common ECM alteration. Conversely, AdCy and MuEp displayed a distinct overexpression of COL27A1 and LAMB3, respectively. Nonhierarchical clustering and principal component analysis revealed a more specific pattern for AdCy with low expression of the common gene signature. In situ studies at the RNA and protein level confirmed these results and indicated that, in contrast to MuEp and SaDu, ECM production in AdCy results from tumour cells and not from cancer-associated fibroblasts (CAFs). Our findings reveal different modes of ECM production leading to common and distinct RNA signatures in SGC. Of note, an overexpression of COL27A1, as in AdCy, has not been linked to any other neoplasm so far. Here, we contribute to the dissection of the ECM composition in SGC and identified a panel of deferentially expressed genes, which could be putative targets for SGC therapy and overcoming therapeutic resistance.
Collapse
Affiliation(s)
- Christoph Arolt
- Institute of Pathology, Medical Faculty, University of Cologne, 50937 Cologne, Germany; (S.W.-R.); (M.O.); (R.B.); (A.Q.)
- Correspondence: ; Tel.: +49-221-478-4726
| | - Moritz Meyer
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty, University of Cologne, 50937 Cologne, Germany; (M.M.); (D.S.); (L.N.); (J.P.K.)
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
| | - Franziska Hoffmann
- Department of Otorhinolaryngology, MALDI Imaging and Innovative Biophotonics, Jena University Hospital, 07747 Jena, Germany;
| | - Svenja Wagener-Ryczek
- Institute of Pathology, Medical Faculty, University of Cologne, 50937 Cologne, Germany; (S.W.-R.); (M.O.); (R.B.); (A.Q.)
| | - David Schwarz
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty, University of Cologne, 50937 Cologne, Germany; (M.M.); (D.S.); (L.N.); (J.P.K.)
| | - Lisa Nachtsheim
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty, University of Cologne, 50937 Cologne, Germany; (M.M.); (D.S.); (L.N.); (J.P.K.)
| | - Dirk Beutner
- Department of Otorhinolaryngology, Head and Neck Surgery, University Medical Center Göttingen, 37075 Göttingen, Germany;
| | - Margarete Odenthal
- Institute of Pathology, Medical Faculty, University of Cologne, 50937 Cologne, Germany; (S.W.-R.); (M.O.); (R.B.); (A.Q.)
| | - Orlando Guntinas-Lichius
- Department of Otorhinolaryngology, Head and Neck Surgery, Jena University Hospital, 07747 Jena, Germany;
| | - Reinhard Buettner
- Institute of Pathology, Medical Faculty, University of Cologne, 50937 Cologne, Germany; (S.W.-R.); (M.O.); (R.B.); (A.Q.)
| | - Ferdinand von Eggeling
- Department of Otorhinolaryngology, MALDI Imaging, Core Unit Proteome Analysis, DFG Core Unit Jena Biophotonic and Imaging Laboratory (JBIL), Jena University Hospital, 07747 Jena, Germany;
| | - Jens Peter Klußmann
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty, University of Cologne, 50937 Cologne, Germany; (M.M.); (D.S.); (L.N.); (J.P.K.)
| | - Alexander Quaas
- Institute of Pathology, Medical Faculty, University of Cologne, 50937 Cologne, Germany; (S.W.-R.); (M.O.); (R.B.); (A.Q.)
| |
Collapse
|
183
|
Yang Y, Yang Y, Yang J, Zhao X, Wei X. Tumor Microenvironment in Ovarian Cancer: Function and Therapeutic Strategy. Front Cell Dev Biol 2020; 8:758. [PMID: 32850861 PMCID: PMC7431690 DOI: 10.3389/fcell.2020.00758] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/20/2020] [Indexed: 02/05/2023] Open
Abstract
Ovarian cancer is one of the leading causes of death in patients with gynecological malignancy. Despite optimal cytoreductive surgery and platinum-based chemotherapy, ovarian cancer disseminates and relapses frequently, with poor prognosis. Hence, it is urgent to find new targeted therapies for ovarian cancer. Recently, the tumor microenvironment has been reported to play a vital role in the tumorigenesis of ovarian cancer, especially with discoveries from genome-, transcriptome- and proteome-wide studies; thus tumor microenvironment may present potential therapeutic target for ovarian cancer. Here, we review the interactions between the tumor microenvironment and ovarian cancer and various therapies targeting the tumor environment.
Collapse
Affiliation(s)
- Yanfei Yang
- Department of Gynecology and Obstetrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Yang Yang
- Department of Gynecology and Obstetrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Jing Yang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xia Zhao
- Department of Gynecology and Obstetrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
184
|
James NE, Woodman M, DiSilvestro PA, Ribeiro JR. The Perfect Combination: Enhancing Patient Response to PD-1-Based Therapies in Epithelial Ovarian Cancer. Cancers (Basel) 2020; 12:E2150. [PMID: 32756436 PMCID: PMC7466102 DOI: 10.3390/cancers12082150] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/28/2020] [Accepted: 08/01/2020] [Indexed: 12/17/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecologic malignancy, with an overall 5-year survival of only 47%. As the development of novel targeted therapies is drastically necessary in order to improve patient survival, current EOC clinical trials have heavily focused on immunotherapeutic approaches, centered upon programmed cell death 1 (PD-1) inhibitors. While PD-1 monotherapies have only exhibited modest responses for patients, it has been theorized that in order to enhance EOC patient response to immunotherapy, combinatorial regimens must be investigated. In this review, unique challenges to EOC PD-1 response will be discussed, along with a comprehensive description of both preclinical and clinical studies evaluating PD-1-based combinatorial therapies. Promising aspects of PD-1-based combinatorial approaches are highlighted, while also discussing specific preclinical and clinical areas of research that need to be addressed, in order to optimize EOC patient immunotherapy response.
Collapse
Affiliation(s)
- Nicole E. James
- Program in Women’s Oncology, Department of Obstetrics and Gynecology, Women and Infants Hospital, Providence, RI 02905, USA; (N.E.J.); (M.W.); (P.A.D.)
| | - Morgan Woodman
- Program in Women’s Oncology, Department of Obstetrics and Gynecology, Women and Infants Hospital, Providence, RI 02905, USA; (N.E.J.); (M.W.); (P.A.D.)
| | - Paul A. DiSilvestro
- Program in Women’s Oncology, Department of Obstetrics and Gynecology, Women and Infants Hospital, Providence, RI 02905, USA; (N.E.J.); (M.W.); (P.A.D.)
- Department of Obstetrics and Gynecology, Warren Alpert School of Medicine, Brown University, Providence, RI 02903, USA
| | - Jennifer R. Ribeiro
- Program in Women’s Oncology, Department of Obstetrics and Gynecology, Women and Infants Hospital, Providence, RI 02905, USA; (N.E.J.); (M.W.); (P.A.D.)
- Department of Obstetrics and Gynecology, Warren Alpert School of Medicine, Brown University, Providence, RI 02903, USA
| |
Collapse
|
185
|
Dasari S, Pandhiri T, Grassi T, Visscher DW, Multinu F, Agarwal K, Mariani A, Shridhar V, Mitra AK. Signals from the Metastatic Niche Regulate Early and Advanced Ovarian Cancer Metastasis through miR-4454 Downregulation. Mol Cancer Res 2020; 18:1202-1217. [PMID: 32350057 PMCID: PMC10788085 DOI: 10.1158/1541-7786.mcr-19-1162] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/16/2020] [Accepted: 04/24/2020] [Indexed: 11/16/2022]
Abstract
Treatment of ovarian cancer is limited by extensive metastasis and yet it remains poorly understood. We have studied the critical step of metastatic colonization in the context of the productive interactions with the metastatic microenvironment with a goal of identifying key regulators. By combining miRNA expression analysis using an organotypic 3D culture model of early ovarian cancer metastasis with that of matched primary and metastatic tumors from 42 patients with ovarian cancer, we identified miR-4454 as a key regulator of both early colonization and advanced metastasis in patients with ovarian cancer. miR-4454 was downregulated in the metastasizing ovarian cancer cells through paracrine signals from microenvironmental fibroblasts, which promoted migration, invasion, proliferation, and clonogenic growth in ovarian cancer cells as well as their ability to penetrate through the outer layers of the omentum. Stable overexpression of miR-4454 decreased metastasis in ovarian cancer xenografts. Its mechanism of action was through the upregulation of its targets, secreted protein acidic and cysteine rich (SPARC) and BCL2 associated athanogene 5 (BAG5), which activated focal adhesion kinase (FAK) signaling, promoted mutant p53 gain of function by its stabilization, and inhibited apoptosis. Because microenvironment-induced downregulation of miR-4454 is essential for early and advanced metastasis, targeting it could be a promising therapeutic approach. IMPLICATIONS: This study identifies a miRNA, miR-4454, which is downregulated by signals from the microenvironment and promotes early and advanced ovarian cancer metastasis through its effects on FAK activation, mutant p53 stabilization, and apoptosis inhibition.
Collapse
Affiliation(s)
- Subramanyam Dasari
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, Indiana
| | - Taruni Pandhiri
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, Indiana
| | - Tommaso Grassi
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota
| | - Daniel W Visscher
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Francesco Multinu
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota
| | - Komal Agarwal
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, Indiana
- Department of Obstetrics and Gynecology, St. Vincent Dunn Hospital, Bedford, Indiana
| | - Andrea Mariani
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota
| | - Viji Shridhar
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Anirban K Mitra
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, Indiana.
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| |
Collapse
|
186
|
D'Angelo E, Lindoso RS, Sensi F, Pucciarelli S, Bussolati B, Agostini M, Collino F. Intrinsic and Extrinsic Modulators of the Epithelial to Mesenchymal Transition: Driving the Fate of Tumor Microenvironment. Front Oncol 2020; 10:1122. [PMID: 32793478 PMCID: PMC7393251 DOI: 10.3389/fonc.2020.01122] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 06/04/2020] [Indexed: 12/11/2022] Open
Abstract
The epithelial to mesenchymal transition (EMT) is an evolutionarily conserved process. In cancer, EMT can activate biochemical changes in tumor cells that enable the destruction of the cellular polarity, leading to the acquisition of invasive capabilities. EMT regulation can be triggered by intrinsic and extrinsic signaling, allowing the tumor to adapt to the microenvironment demand in the different stages of tumor progression. In concomitance, tumor cells undergoing EMT actively interact with the surrounding tumor microenvironment (TME) constituted by cell components and extracellular matrix as well as cell secretome elements. As a result, the TME is in turn modulated by the EMT process toward an aggressive behavior. The current review presents the intrinsic and extrinsic modulators of EMT and their relationship with the TME, focusing on the non-cell-derived components, such as secreted metabolites, extracellular matrix, as well as extracellular vesicles. Moreover, we explore how these modulators can be suitable targets for anticancer therapy and personalized medicine.
Collapse
Affiliation(s)
- Edoardo D'Angelo
- First Surgical Clinic, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
- LIFELAB Program, Consorzio per la Ricerca Sanitaria–CORIS, Veneto Region, Padua, Italy
- Institute of Pediatric Research, Fondazione Citta della Speranza, Padua, Italy
| | - Rafael Soares Lindoso
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Regenerative Medicine–REGENERA, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Francesca Sensi
- Institute of Pediatric Research, Fondazione Citta della Speranza, Padua, Italy
- Department of Molecular Sciences and Nanosystems, Cà Foscari University of Venice, Venice, Italy
| | - Salvatore Pucciarelli
- First Surgical Clinic, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Benedetta Bussolati
- Department of Medical Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Marco Agostini
- First Surgical Clinic, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
- LIFELAB Program, Consorzio per la Ricerca Sanitaria–CORIS, Veneto Region, Padua, Italy
- Institute of Pediatric Research, Fondazione Citta della Speranza, Padua, Italy
| | - Federica Collino
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Biomedical Sciences, University of Padova, Padua, Italy
- Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione Ca' Granda, IRCCS Policlinico di Milano, Milan, Italy
| |
Collapse
|
187
|
Izzi V, Davis MN, Naba A. Pan-Cancer Analysis of the Genomic Alterations and Mutations of the Matrisome. Cancers (Basel) 2020; 12:E2046. [PMID: 32722287 PMCID: PMC7463652 DOI: 10.3390/cancers12082046] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/17/2020] [Accepted: 07/22/2020] [Indexed: 12/13/2022] Open
Abstract
The extracellular matrix (ECM) is a master regulator of all cellular functions and a major component of the tumor microenvironment. We previously defined the "matrisome" as the ensemble of genes encoding ECM proteins and proteins modulating ECM structure or function. While compositional and biomechanical changes in the ECM regulate cancer progression, no study has investigated the genomic alterations of matrisome genes in cancers and their consequences. Here, mining The Cancer Genome Atlas (TCGA) data, we found that copy number alterations and mutations are frequent in matrisome genes, even more so than in the rest of the genome. We also found that these alterations are predicted to significantly impact gene expression and protein function. Moreover, we identified matrisome genes whose mutational burden is an independent predictor of survival. We propose that studying genomic alterations of matrisome genes will further our understanding of the roles of this compartment in cancer progression and will lead to the development of innovative therapeutic strategies targeting the ECM.
Collapse
Affiliation(s)
- Valerio Izzi
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, FI-90014 Oulu, Finland;
- Finnish Cancer Institute, 00130 Helsinki, Finland
| | - Martin N. Davis
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, USA;
| | - Alexandra Naba
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, USA;
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| |
Collapse
|
188
|
Modelling Epithelial Ovarian Cancer in Mice: Classical and Emerging Approaches. Int J Mol Sci 2020; 21:ijms21134806. [PMID: 32645943 PMCID: PMC7370285 DOI: 10.3390/ijms21134806] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/03/2020] [Accepted: 07/05/2020] [Indexed: 12/31/2022] Open
Abstract
High-grade serous epithelial ovarian cancer (HGSC) is the most aggressive subtype of epithelial ovarian cancer. The identification of germline and somatic mutations along with genomic information unveiled by The Cancer Genome Atlas (TCGA) and other studies has laid the foundation for establishing preclinical models with high fidelity to the molecular features of HGSC. Notwithstanding such progress, the field of HGSC research still lacks a model that is both robust and widely accessible. In this review, we discuss the recent advancements and utility of HGSC genetically engineered mouse models (GEMMs) to date. Further analysis and critique on alternative approaches to modelling HGSC considers technological advancements in somatic gene editing and modelling prototypic organs, capable of tumorigenesis, on a chip.
Collapse
|
189
|
Fogg KC, Renner CM, Christian H, Walker A, Marty-Santos L, Khan A, Olson WR, Parent C, O'Shea A, Wellik DM, Weisman PS, Kreeger PK. Ovarian Cells Have Increased Proliferation in Response to Heparin-Binding Epidermal Growth Factor as Collagen Density Increases. Tissue Eng Part A 2020; 26:747-758. [PMID: 32598229 DOI: 10.1089/ten.tea.2020.0001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
It is well known that during ovarian cancer progression, the omentum transforms from a thin lacy organ to a thick tougher tissue. However, the mechanisms regulating this transformation and the implications of the altered microenvironment on ovarian cancer progression remain unclear. To address these questions, the global and local concentrations of collagen I were determined for normal and metastatic human omentum. Collagen I was increased 5.3-fold in omenta from ovarian cancer patients and localized to areas of activated fibroblasts rather than regions with a high density of cancer cells. Transforming growth factor beta 1 (TGFβ1) was detected in ascites from ovarian cancer patients (4 ng/mL), suggesting a potential role for TGFβ1 in the observed increase in collagen. Treatment with TGFβ1 induced fibroblast activation, proliferation, and collagen deposition in mouse omental explants and an in vitro model with human omental fibroblasts. Finally, the impact of increased collagen I on ovarian cancer cells was determined by examining proliferation on collagen I gels formulated to mimic normal and cancerous omenta. While collagen density alone had no impact on proliferation, a synergistic effect was observed with collagen density and heparin-binding epidermal growth factor treatment. These results suggest that TGFβ1 induces collagen deposition from the resident fibroblasts in the omentum and that this altered microenvironment impacts cancer cell response to growth factors found in ascites. Impact statement Using quantitative analysis of patient samples, in vitro models of the metastatic ovarian cancer microenvironment were designed with pathologically relevant collagen densities and growth factor concentrations. Studies in these models support a mechanism where transforming growth factor β1 in the ascites fluid induces omental fibroblast proliferation, activation, and deposition of collagen I, which then impacts tumor cell proliferation in response to additional ascites growth factors such as heparin-binding epidermal growth factor. This approach can be used to dissect mechanisms involved in microenvironmental modeling in multiple disease applications.
Collapse
Affiliation(s)
- Kaitlin C Fogg
- Department of Biomedical Engineering, University of Wisconsin - Madison, Madison, Wisconsin, USA
| | - Carine M Renner
- Department of Biomedical Engineering, University of Wisconsin - Madison, Madison, Wisconsin, USA
| | - Hannah Christian
- Department of Biomedical Engineering, University of Wisconsin - Madison, Madison, Wisconsin, USA
| | - Alyssa Walker
- Department of Biomedical Engineering, University of Wisconsin - Madison, Madison, Wisconsin, USA
| | - Leilani Marty-Santos
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Aisha Khan
- Department of Biomedical Engineering, University of Wisconsin - Madison, Madison, Wisconsin, USA
| | - Will R Olson
- Department of Biomedical Engineering, University of Wisconsin - Madison, Madison, Wisconsin, USA
| | - Carl Parent
- Department of Biomedical Engineering, University of Wisconsin - Madison, Madison, Wisconsin, USA
| | - Andrea O'Shea
- Department of Obstetrics and Gynecology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Deneen M Wellik
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA.,University of Wisconsin Carbone Cancer Center, and University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Paul S Weisman
- University of Wisconsin Carbone Cancer Center, and University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA.,Department of Pathology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Pamela K Kreeger
- Department of Biomedical Engineering, University of Wisconsin - Madison, Madison, Wisconsin, USA.,Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA.,Department of Obstetrics and Gynecology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA.,University of Wisconsin Carbone Cancer Center, and University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| |
Collapse
|
190
|
Dey S, Trau M, Koo KM. Surface-Enhanced Raman Spectroscopy for Cancer Immunotherapy Applications: Opportunities, Challenges, and Current Progress in Nanomaterial Strategies. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E1145. [PMID: 32545182 PMCID: PMC7353228 DOI: 10.3390/nano10061145] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/05/2020] [Accepted: 06/06/2020] [Indexed: 02/06/2023]
Abstract
Cancer immunotherapy encompasses a variety of approaches which target or use a patient's immune system components to eliminate cancer. Notably, the current use of immune checkpoint inhibitors to target immune checkpoint receptors such as CTLA-4 or PD-1 has led to remarkable treatment responses in a variety of cancers. To predict cancer patients' immunotherapy responses effectively and efficiently, multiplexed immunoassays have been shown to be advantageous in sensing multiple immunomarkers of the tumor microenvironment simultaneously for patient stratification. Surface-enhanced Raman spectroscopy (SERS) is well-regarded for its capabilities in multiplexed bioassays and has been increasingly demonstrated in cancer immunotherapy applications in recent years. This review focuses on SERS-active nanomaterials in the modern literature which have shown promise for enabling cancer patient-tailored immunotherapies, including multiplexed in vitro and in vivo immunomarker sensing and imaging, as well as immunotherapy drug screening and delivery.
Collapse
Affiliation(s)
- Shuvashis Dey
- Centre for Personalized Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia
| | - Matt Trau
- Centre for Personalized Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia
- School of Chemistry and Molecular Biosciences, the University of Queensland, Brisbane, QLD 4072, Australia
| | - Kevin M. Koo
- XING Technologies Pty Ltd., Brisbane, QLD 4073, Australia
- The University of Queensland Centre for Clinical Research (UQCCR), Brisbane, QLD 4029, Australia
| |
Collapse
|
191
|
Muñoz-Galván S, Carnero A. Targeting Cancer Stem Cells to Overcome Therapy Resistance in Ovarian Cancer. Cells 2020; 9:cells9061402. [PMID: 32512891 PMCID: PMC7349391 DOI: 10.3390/cells9061402] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 06/02/2020] [Indexed: 12/18/2022] Open
Abstract
Ovarian cancer is the most lethal gynecological malignancy due to its late detection and high recurrence rate. Resistance to conventional platinum-based therapies and metastasis are attributed to a population of cells within tumors called cancer stem cells, which possess stem-like features and are able to recapitulate new tumors. Recent studies have deepened the understanding of the biology of ovarian cancer stem cells and their special properties and have identified multiple markers and signaling pathways responsible for their self-renewal abilities. Targeting cancer stem cells represents the most promising strategy for overcoming therapy resistance and reducing mortality in ovarian cancer, but further efforts must be made to improve our understanding of the mechanisms involved in therapy resistance. In this review, we summarize our current knowledge about ovarian cancer stem cells, their involvement in metastasis and their interactions with the tumor microenvironment; we also discuss the therapeutic approaches that are being developed to target them to prevent tumor relapse.
Collapse
Affiliation(s)
- Sandra Muñoz-Galván
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Avda. Manuel Siurot s/n, 41013 Seville, Spain
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (S.M.-G.); (A.C.); Tel.: +34-955-923-115 (S.M.-G); +34-955-923-110 (A.C.)
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Avda. Manuel Siurot s/n, 41013 Seville, Spain
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (S.M.-G.); (A.C.); Tel.: +34-955-923-115 (S.M.-G); +34-955-923-110 (A.C.)
| |
Collapse
|
192
|
Galli F, Aguilera JV, Palermo B, Markovic SN, Nisticò P, Signore A. Relevance of immune cell and tumor microenvironment imaging in the new era of immunotherapy. J Exp Clin Cancer Res 2020; 39:89. [PMID: 32423420 PMCID: PMC7236372 DOI: 10.1186/s13046-020-01586-y] [Citation(s) in RCA: 181] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 04/28/2020] [Indexed: 02/06/2023] Open
Abstract
Tumor-infiltrating immune cells play a key role against cancer. However, malignant cells are able to evade the immune response and establish a very complex balance in which different immune subtypes may drive tumor progression, metastatization and resistance to therapy. New immunotherapeutic approaches aim at restoring the natural balance and increase immune response against cancer by different mechanisms. The complexity of these interactions and the heterogeneity of immune cell subpopulations are a real challenge when trying to develop new immunotherapeutics and evaluate or predict their efficacy in vivo. To this purpose, molecular imaging can offer non-invasive diagnostic tools like radiopharmaceuticals, contrast agents or fluorescent dyes. These agents can be useful for preclinical and clinical purposes and can overcome [18F]FDG limitations in discriminating between true-progression and pseudo-progression. This review provides a comprehensive overview of immune cells involved in microenvironment, available immunotherapies and imaging agents to highlight the importance of new therapeutic biomarkers and their in vivo evaluation to improve the management of cancer patients.
Collapse
Affiliation(s)
- Filippo Galli
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, "Sapienza" University of Rome, S. Andrea University Hospital, Roma, Italy.
| | - Jesus Vera Aguilera
- Department of oncology and Department of Immunology, Mayo Clinic, (MN), Rochester, USA
| | - Belinda Palermo
- Tumor Immunology and Immunotherapy Unit, Department of Research, Advanced Diagnostics and Technological Innovation, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Svetomir N Markovic
- Department of oncology and Department of Immunology, Mayo Clinic, (MN), Rochester, USA
| | - Paola Nisticò
- Tumor Immunology and Immunotherapy Unit, Department of Research, Advanced Diagnostics and Technological Innovation, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Alberto Signore
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, "Sapienza" University of Rome, S. Andrea University Hospital, Roma, Italy
| |
Collapse
|
193
|
Yang Z, Xu H, Zhao X. Designer Self-Assembling Peptide Hydrogels to Engineer 3D Cell Microenvironments for Cell Constructs Formation and Precise Oncology Remodeling in Ovarian Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1903718. [PMID: 32382486 PMCID: PMC7201262 DOI: 10.1002/advs.201903718] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/08/2020] [Indexed: 02/05/2023]
Abstract
Designer self-assembling peptides form the entangled nanofiber networks in hydrogels by ionic-complementary self-assembly. This type of hydrogel has realistic biological and physiochemical properties to serve as biomimetic extracellular matrix (ECM) for biomedical applications. The advantages and benefits are distinct from natural hydrogels and other synthetic or semisynthetic hydrogels. Designer peptides provide diverse alternatives of main building blocks to form various functional nanostructures. The entangled nanofiber networks permit essential compositional complexity and heterogeneity of engineering cell microenvironments in comparison with other hydrogels, which may reconstruct the tumor microenvironments (TMEs) in 3D cell cultures and tissue-specific modeling in vitro. Either ovarian cancer progression or recurrence and relapse are involved in the multifaceted TMEs in addition to mesothelial cells, fibroblasts, endothelial cells, pericytes, immune cells, adipocytes, and the ECM. Based on the progress in common hydrogel products, this work focuses on the diverse designer self-assembling peptide hydrogels for instructive cell constructs in tissue-specific modeling and the precise oncology remodeling for ovarian cancer, which are issued by several research aspects in a 3D context. The advantages and significance of designer peptide hydrogels are discussed, and some common approaches and coming challenges are also addressed in current complex tumor diseases.
Collapse
Affiliation(s)
- Zehong Yang
- West China School of Basic Medical Sciences and Forensic MedicineSichuan UniversityChengduSichuan610041P. R. China
- Institute for Nanobiomedical Technology and Membrane BiologyWest China HospitalSichuan UniversityChengduSichuan610041P. R. China
| | - Hongyan Xu
- GL Biochem (Shanghai) Ltd.519 Ziyue Rd.Shanghai200241P. R. China
| | - Xiaojun Zhao
- Institute for Nanobiomedical Technology and Membrane BiologyWest China HospitalSichuan UniversityChengduSichuan610041P. R. China
- Wenzhou InstituteUniversity of Chinese Academy of Sciences (Wenzhou Institute of Biomaterials & Engineering)WenzhouZhejiang325001P. R. China
| |
Collapse
|
194
|
Single-cell RNA sequencing reveals compromised immune microenvironment in precursor stages of multiple myeloma. ACTA ACUST UNITED AC 2020; 1:493-506. [PMID: 33409501 DOI: 10.1038/s43018-020-0053-3] [Citation(s) in RCA: 237] [Impact Index Per Article: 47.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Precursor states of Multiple Myeloma (MM) and its native tumor microenvironment need in-depth molecular characterization to better stratify and treat patients at risk. Using single-cell RNA sequencing of bone marrow cells from precursor stages, MGUS and smoldering myeloma (SMM), to full-blown MM alongside healthy donors, we demonstrate early immune changes during patient progression. We find NK cell abundance is frequently increased in early stages, and associated with altered chemokine receptor expression. As early as SMM, we show loss of GrK+ memory cytotoxic T-cells, and show their critical role in MM immunosurveillance in mouse models. Finally, we report MHC class II dysregulation in CD14+ monocytes, which results in T cell suppression in vitro. These results provide a comprehensive map of immune changes at play over the evolution of pre-malignant MM, which will help develop strategies for immune-based patient stratification.
Collapse
|
195
|
Gordon S, Plüddemann A, Mukhopadhyay S. Plasma membrane receptors of tissue macrophages: functions and role in pathology. J Pathol 2020; 250:656-666. [PMID: 32086805 DOI: 10.1002/path.5404] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 02/18/2020] [Indexed: 12/11/2022]
Abstract
The cells of the mononuclear phagocyte system (MPS) constitute a dispersed organ, which is distributed throughout the body. Macrophages in different tissues display distinctive mosaic phenotypes as resident and recruited cells of embryonic and bone marrow origin, respectively. They help to maintain homeostasis during development and throughout adult life, yet contribute to the pathogenesis of many disease processes, including inflammation, innate and adaptive immunity, metabolic disorders, and cancer. Heterogeneous tissue macrophage populations display a wide variety of surface molecules to recognise and respond to host, microbial, and exogenous ligands in their environment; their receptors mediate the uptake and destruction of effete and dying host cells and pathogens, as well as contribute trophic and secretory functions within every organ in the body. Apart from local cellular interactions, macrophage surface molecules and products serve to mobilise and coordinate systemic humoral and cellular responses. Their use as antigen markers in pathogenesis and as potential drug targets has lagged in clinical pathology and human immunotherapy. In this review, we summarise the properties of selected surface molecules expressed on macrophages in different tissues and disease processes, to provide a functional basis for diagnosis, further research, and treatment. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Siamon Gordon
- College of Medicine, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan City, Taiwan.,Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Annette Plüddemann
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Subhankar Mukhopadhyay
- Peter Gorer Department of Immunobiology, Medical Research Council Centre for Transplantation, King's College London, London, UK
| |
Collapse
|
196
|
Demircioglu F, Wang J, Candido J, Costa ASH, Casado P, de Luxan Delgado B, Reynolds LE, Gomez-Escudero J, Newport E, Rajeeve V, Baker AM, Roy-Luzarraga M, Graham TA, Foster J, Wang Y, Campbell JJ, Singh R, Zhang P, Schall TJ, Balkwill FR, Sosabowski J, Cutillas PR, Frezza C, Sancho P, Hodivala-Dilke K. Cancer associated fibroblast FAK regulates malignant cell metabolism. Nat Commun 2020; 11:1290. [PMID: 32157087 PMCID: PMC7064590 DOI: 10.1038/s41467-020-15104-3] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 02/18/2020] [Indexed: 12/19/2022] Open
Abstract
Emerging evidence suggests that cancer cell metabolism can be regulated by cancer-associated fibroblasts (CAFs), but the mechanisms are poorly defined. Here we show that CAFs regulate malignant cell metabolism through pathways under the control of FAK. In breast and pancreatic cancer patients we find that low FAK expression, specifically in the stromal compartment, predicts reduced overall survival. In mice, depletion of FAK in a subpopulation of CAFs regulates paracrine signals that increase malignant cell glycolysis and tumour growth. Proteomic and phosphoproteomic analysis in our mouse model identifies metabolic alterations which are reflected at the transcriptomic level in patients with low stromal FAK. Mechanistically we demonstrate that FAK-depletion in CAFs increases chemokine production, which via CCR1/CCR2 on cancer cells, activate protein kinase A, leading to enhanced malignant cell glycolysis. Our data uncover mechanisms whereby stromal fibroblasts regulate cancer cell metabolism independent of genetic mutations in cancer cells.
Collapse
Affiliation(s)
- Fevzi Demircioglu
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Jun Wang
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Juliana Candido
- Centre for Cancer and Inflammation, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Ana S H Costa
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge Biomedical Campus, Cambridge, CB2 0XZ, UK
| | - Pedro Casado
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Beatriz de Luxan Delgado
- Centre for Stem Cells in Cancer and Ageing, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Louise E Reynolds
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Jesus Gomez-Escudero
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Emma Newport
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Vinothini Rajeeve
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Ann-Marie Baker
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Marina Roy-Luzarraga
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Trevor A Graham
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Julie Foster
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Yu Wang
- ChemoCentryx Inc., 850 Maude Ave, Mountain View, CA94043, USA
| | | | - Rajinder Singh
- ChemoCentryx Inc., 850 Maude Ave, Mountain View, CA94043, USA
| | - Penglie Zhang
- ChemoCentryx Inc., 850 Maude Ave, Mountain View, CA94043, USA
| | - Thomas J Schall
- ChemoCentryx Inc., 850 Maude Ave, Mountain View, CA94043, USA
| | - Frances R Balkwill
- Centre for Cancer and Inflammation, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Jane Sosabowski
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Pedro R Cutillas
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Christian Frezza
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge Biomedical Campus, Cambridge, CB2 0XZ, UK
| | - Patricia Sancho
- Centre for Stem Cells in Cancer and Ageing, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
- IIS Aragon, Hospital Universitario Miguel Servet, Zaragoza, 50009, Spain
| | - Kairbaan Hodivala-Dilke
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK.
| |
Collapse
|
197
|
Esophageal Cancer Development: Crucial Clues Arising from the Extracellular Matrix. Cells 2020; 9:cells9020455. [PMID: 32079295 PMCID: PMC7072790 DOI: 10.3390/cells9020455] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 02/05/2020] [Accepted: 02/13/2020] [Indexed: 02/06/2023] Open
Abstract
In the last years, the extracellular matrix (ECM) has been reported as playing a relevant role in esophageal cancer (EC) development, with this compartment being related to several aspects of EC genesis and progression. This sounds very interesting due to the complexity of this highly incident and lethal tumor, which takes the sixth position in mortality among all tumor types worldwide. The well-established increase in ECM stiffness, which is able to trigger mechanotransduction signaling, is capable of regulating several malignant behaviors by converting alteration in ECM mechanics into cytoplasmatic biochemical signals. In this sense, it has been shown that some molecules play a key role in these events, particularly the different collagen isoforms, as well as enzymes related to its turnover, such as lysyl oxidase (LOX) and matrix metalloproteinases (MMPs). In fact, MMPs are not only involved in ECM stiffness, but also in other events related to ECM homeostasis, which includes ECM remodeling. Therefore, the crucial role of distinct MMPs isoform has already been reported, especially MMP-2, -3, -7, and -9, along EC development, thus strongly associating these proteins with the control of important cellular events during tumor progression, particularly in the process of invasion during metastasis establishment. In addition, by distinct mechanisms, a vast diversity of glycoproteins and proteoglycans, such as laminin, fibronectin, tenascin C, galectin, dermatan sulfate, and hyaluronic acid exert remarkable effects in esophageal malignant cells due to the activation of oncogenic signaling pathways mainly involved in cytoskeleton alterations during adhesion and migration processes. Finally, the wide spectrum of interactions potentially mediated by ECM may represent a singular intervention scenario in esophageal carcinogenesis natural history and, due to the scarce knowledge on the cellular and molecular mechanisms involved in EC development, the growing body of evidence on ECM’s role along esophageal carcinogenesis might provide a solid base to improve its management in the future.
Collapse
|
198
|
Rafaeva M, Erler JT. Framing cancer progression: influence of the organ- and tumour-specific matrisome. FEBS J 2020; 287:1454-1477. [PMID: 31972068 DOI: 10.1111/febs.15223] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/16/2019] [Accepted: 01/20/2020] [Indexed: 12/19/2022]
Abstract
The extracellular matrix (ECM) plays a crucial role in regulating organ homeostasis. It provides mechanical and biochemical cues directing cellular behaviour and, therefore, has control over the progression of diseases such as cancer. Recent efforts have greatly enhanced our knowledge of the protein composition of the ECM and its regulators, the so-called matrisome, in healthy and cancerous tissues; yet, an overview of the common signatures and organ-specific ECM in cancer is missing. Here, we address this by taking a detailed approach to review why cancer grows in certain organs, and focus on the influence of the matrisome at primary and metastatic tumour sites. Our in-depth and comprehensive review of the current literature and general understanding identifies important commonalities and distinctions, providing insight into the biology of metastasis, which could pave the way to improve future diagnostics and therapies.
Collapse
Affiliation(s)
- Maria Rafaeva
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen (UCPH), Denmark
| | - Janine T Erler
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen (UCPH), Denmark
| |
Collapse
|
199
|
Hebert JD, Myers SA, Naba A, Abbruzzese G, Lamar JM, Carr SA, Hynes RO. Proteomic Profiling of the ECM of Xenograft Breast Cancer Metastases in Different Organs Reveals Distinct Metastatic Niches. Cancer Res 2020; 80:1475-1485. [PMID: 32019869 DOI: 10.1158/0008-5472.can-19-2961] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/19/2019] [Accepted: 12/19/2019] [Indexed: 12/21/2022]
Abstract
Metastasis causes most cancer-related deaths, and one poorly understood aspect of metastatic cancer is the adaptability of cells from a primary tumor to create new niches and survive in multiple, different secondary sites. We used quantitative mass spectrometry to analyze the extracellular matrix (ECM), a critical component of metastatic niches, in metastases to the brain, lungs, liver, and bone marrow, all derived from parental MDA-MB-231 triple-negative breast cancer cells. Tumor and stromal cells cooperated in forming niches; stromal cells produced predominantly core, structural ECM proteins and tumor cells produced a diverse array of ECM-associated proteins, including secreted factors and modulators of the matrix. In addition, tumor and stromal cells together created distinct niches in each tissue. Downregulation of SERPINB1, a protein elevated in brain metastases, led to a reduction in brain metastasis, suggesting that some niche-specific ECM proteins may be involved in metastatic tropism. SIGNIFICANCE: Tumor and stromal cells together create distinct ECM niches in breast cancer metastases to various tissues, providing new insight into how tumor cells adapt to survive in different tissue environments.
Collapse
Affiliation(s)
- Jess D Hebert
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Samuel A Myers
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Alexandra Naba
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Genevieve Abbruzzese
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - John M Lamar
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Steven A Carr
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Richard O Hynes
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts. .,Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Howard Hughes Medical Institute, Chevy Chase, Maryland
| |
Collapse
|
200
|
Maniati E, Berlato C, Gopinathan G, Heath O, Kotantaki P, Lakhani A, McDermott J, Pegrum C, Delaine-Smith RM, Pearce OMT, Hirani P, Joy JD, Szabova L, Perets R, Sansom OJ, Drapkin R, Bailey P, Balkwill FR. Mouse Ovarian Cancer Models Recapitulate the Human Tumor Microenvironment and Patient Response to Treatment. Cell Rep 2020; 30:525-540.e7. [PMID: 31940494 PMCID: PMC6963791 DOI: 10.1016/j.celrep.2019.12.034] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 09/06/2019] [Accepted: 12/10/2019] [Indexed: 12/13/2022] Open
Abstract
Although there are many prospective targets in the tumor microenvironment (TME) of high-grade serous ovarian cancer (HGSOC), pre-clinical testing is challenging, especially as there is limited information on the murine TME. Here, we characterize the TME of six orthotopic, transplantable syngeneic murine HGSOC lines established from genetic models and compare these to patient biopsies. We identify significant correlations between the transcriptome, host cell infiltrates, matrisome, vasculature, and tissue modulus of mouse and human TMEs, with several stromal and malignant targets in common. However, each model shows distinct differences and potential vulnerabilities that enabled us to test predictions about response to chemotherapy and an anti-IL-6 antibody. Using machine learning, the transcriptional profiles of the mouse tumors that differed in chemotherapy response are able to classify chemotherapy-sensitive and -refractory patient tumors. These models provide useful pre-clinical tools and may help identify subgroups of HGSOC patients who are most likely to respond to specific therapies.
Collapse
Affiliation(s)
- Eleni Maniati
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Chiara Berlato
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Ganga Gopinathan
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Owen Heath
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Panoraia Kotantaki
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Anissa Lakhani
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Jacqueline McDermott
- University College Hospital, UCLH Cellular Pathology, 11-20 Capper Street, London WC1E 6JA, UK
| | - Colin Pegrum
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | | | - Oliver M T Pearce
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Priyanka Hirani
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Joash D Joy
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Ludmila Szabova
- Center for Advanced Preclinical Research, Frederick National Laboratory for Cancer Research at the National Cancer Institute-Frederick, Frederick, MD, USA
| | - Ruth Perets
- Rambam Health Care Campus, Technion - Israel Institute of Technology, Haifa, Israel
| | - Owen J Sansom
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
| | - Ronny Drapkin
- Penn Ovarian Cancer Research Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Peter Bailey
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK; Department for Surgical Research, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Frances R Balkwill
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK.
| |
Collapse
|