351
|
Ilina O, Campanello L, Gritsenko PG, Vullings M, Wang C, Bult P, Losert W, Friedl P. Intravital microscopy of collective invasion plasticity in breast cancer. Dis Model Mech 2018; 11:dmm.034330. [PMID: 29997220 PMCID: PMC6176993 DOI: 10.1242/dmm.034330] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 07/04/2018] [Indexed: 01/15/2023] Open
Abstract
Cancer invasion programs are adaptive by switching between metastatic collective and single-cell dissemination; however, current intravital microscopy models for epithelial cancer in mice fail to reliably recreate such invasion plasticity. Using microimplantation of breast cancer spheroids into the murine mammary fat pad and live-cell monitoring, we show microenvironmental conditions and cytoskeletal adaptation during collective to single-cell transition in vivo E-cadherin-expressing 4T1 and E-cadherin-negative MMT tumors both initiated collective invasion along stromal structures, reflecting invasion patterns in 3D organotypic culture and human primary ductal and lobular carcinoma. Collectively invading cells developed weakly oscillatory actin dynamics, yet provided zones for single-cell transitions with accentuated, more chaotic actin fluctuations. This identifies collective invasion in vivo as a dynamic niche and efficient source for single-cell release.
Collapse
Affiliation(s)
- Olga Ilina
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, PO Box 9101, 6500HB, Nijmegen, The Netherlands
| | - Leonard Campanello
- Department of Physics, Institute for Physical Science and Technology, University of Maryland, College Park, MD 20742, USA
| | - Pavlo G Gritsenko
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, PO Box 9101, 6500HB, Nijmegen, The Netherlands
| | - Manon Vullings
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, PO Box 9101, 6500HB, Nijmegen, The Netherlands
| | - Chenlu Wang
- Department of Physics, Institute for Physical Science and Technology, University of Maryland, College Park, MD 20742, USA
| | - Peter Bult
- Department of Pathology, Radboud University Medical Center, PO Box 9101, 6500HB, Nijmegen, The Netherlands
| | - Wolfgang Losert
- Department of Physics, Institute for Physical Science and Technology, University of Maryland, College Park, MD 20742, USA
| | - Peter Friedl
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, PO Box 9101, 6500HB, Nijmegen, The Netherlands .,Cancer Genomic Centre, 3584CG, Utrecht, The Netherlands.,David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
352
|
Wang RX, Chen S, Huang L, Zhou Y, Shao ZM. Monitoring Serum VEGF in Neoadjuvant Chemotherapy for Patients with Triple-Negative Breast Cancer: A New Strategy for Early Prediction of Treatment Response and Patient Survival. Oncologist 2018; 24:753-761. [PMID: 30126858 DOI: 10.1634/theoncologist.2017-0602] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 06/20/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND This study aimed to investigate the clinical utility of serum biomarker changes during neoadjuvant chemotherapy (NAC) for triple-negative breast cancer (TNBC). METHODS A total of 303 patients with TNBC were included in this study. Serum samples were taken at three time points during NAC: baseline, prior to the third cycle, and prior to surgery. Luminex multibiomarker panel for 29 serum biomarkers was used to detect their correlation with NAC response. The predictive and prognostic value of each selected biomarker was then studied. RESULTS Vascular endothelial growth factor (VEGF) was the only biomarker that correlated with treatment response, with a decreasing trend in pCR patients relative to non-pCR patients (p < .001). Univariable and multivariable analyses revealed that the relative change in VEGF prior to the third cycle of NAC had a remarkable predictive value for both pCR and pathological nonresponse with high sensitivity and specificity. VEGF was also independently correlated with disease-free survival. CONCLUSION Our findings indicate that monitoring serum VEGF could help identify patients with different responses at an early time point of NAC and at varying risk of disease relapse. Serum VEGF may also serve as an alternative to traditional response-evaluating methodologies in tailoring and modifying the NAC strategy for both operable and advanced TNBCs. IMPLICATIONS FOR PRACTICE Neoadjuvant chemotherapy (NAC) followed by definitive surgery is a standard of care for locally advanced breast cancer. The identification of sensitive responders to neoadjuvant therapy is highly significant for breast cancer, especially triple-negative breast cancer (TNBC). Results of this study indicate that the monitoring of serum vascular endothelial growth factor (VEGF) could identify patients with favorable or poor responses at an early time point of NAC. Furthermore, the prediction power of VEGF was better than traditional response-evaluating methods. VEGF might serve as a complement or alternative to traditional imaging-based response-evaluating methodologies in tailoring systemic treatment strategies for both operable and advanced TNBCs.
Collapse
Affiliation(s)
- Ruo-Xi Wang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center/Cancer Institute, Shanghai, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Sheng Chen
- Department of Breast Surgery, Fudan University Shanghai Cancer Center/Cancer Institute, Shanghai, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Liang Huang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center/Cancer Institute, Shanghai, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Ying Zhou
- Department of Breast Surgery, Fudan University Shanghai Cancer Center/Cancer Institute, Shanghai, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center/Cancer Institute, Shanghai, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
- Institutes of Biomedical Science, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
353
|
Impact of the hypoxic phenotype on the uptake and efflux of nanoparticles by human breast cancer cells. Sci Rep 2018; 8:12318. [PMID: 30120388 PMCID: PMC6098061 DOI: 10.1038/s41598-018-30517-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 07/30/2018] [Indexed: 12/30/2022] Open
Abstract
Breast cancer cells adapt to the hypoxic tumoral environment by undergoing changes in metabolism, cell signalling, endo-lysosomal receptor uptake and recycling. The resulting hypoxic cell phenotype has the potential to undermine the therapeutic efficacy of nanomedicines designed for endocytic uptake and specific intracellular trafficking. The aim of this study was to examine the impact of hypoxia and simulated reperfusion on the in vitro uptake and release of nanomedicines by human breast cancer cells. Cells were exposed to a hypoxic preconditioning treatment in 1% oxygen for 6 and 24 hours to induce temporal changes in the hypoxic circuit (e.g. HIF-1α expression). The preconditioned cells were then dosed with nanoparticles for 45 or 180 minutes emulating nanomedicine access following tumor reperfusion. Hypoxic preconditioning significantly increased nanoparticle retention by up to 10% when compared to normoxic cultures, with the greatest relative difference between normoxic and hypoxic cultures occurring with a 45 minute dosing interval. Exocytosis studies indicated that the preconditioned cells had a significantly increased nanoparticle efflux (up to 9%) when compared to normoxic cells. Overall, we were able to show that hypoxic preconditioning regulates both the endocytosis and exocytosis of nanomedicines in human breast cancer cells.
Collapse
|
354
|
Pseudopterosin Inhibits Proliferation and 3D Invasion in Triple-Negative Breast Cancer by Agonizing Glucocorticoid Receptor Alpha. Molecules 2018; 23:molecules23081992. [PMID: 30103404 PMCID: PMC6222322 DOI: 10.3390/molecules23081992] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 08/07/2018] [Accepted: 08/08/2018] [Indexed: 12/18/2022] Open
Abstract
Pseudopterosin, produced by the sea whip of the genus Antillogorgia, possesses a variety of promising biological activities, including potent anti-inflammatory effects. However, few studies examined pseudopterosin in the treatment of cancer cells and, to our knowledge, the ability to inhibit triple-negative breast cancer (TNBC) proliferation or invasion has not been explored. Thus, we evaluated the as-yet unknown mechanism of action of pseudopterosin: Pseudopterosin was able to inhibit proliferation of TNBC. Interestingly, analyzing breast cancer cell proliferation after knocking down glucocorticoid receptor α (GRα) revealed that the antiproliferative effects of pseudopterosin were significantly inhibited when GRα expression was reduced. Furthermore, pseudopterosin inhibited the invasion of MDA-MB-231 3D tumor spheroids embedded in an extracellular-like matrix. Remarkably, the knockdown of GRα in 3D tumor spheroids revealed increased ability of cells to invade the surrounding matrix. In a coculture, encompassing peripheral blood mononuclear cells (PBMC) and MDA-MB-231 cells, and the production of interleukin 6 (IL-6) and interleukin 8 (IL-8) significantly increased compared to a monoculture. Notably, pseudopterosin indicated to block cytokine elevation, representing key players in tumor progression in the coculture. Thus, our results reveal pseudopterosin treatment as a potential novel approach in TNBC therapy.
Collapse
|
355
|
Chitty JL, Filipe EC, Lucas MC, Herrmann D, Cox TR, Timpson P. Recent advances in understanding the complexities of metastasis. F1000Res 2018; 7. [PMID: 30135716 DOI: 10.12688/f1000research.15064.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/24/2018] [Indexed: 12/14/2022] Open
Abstract
Tumour metastasis is a dynamic and systemic process. It is no longer seen as a tumour cell-autonomous program but as a multifaceted and complex series of events, which is influenced by the intrinsic cellular mutational burden of cancer cells and the numerous bidirectional interactions between malignant and non-malignant cells and fine-tuned by the various extrinsic cues of the extracellular matrix. In cancer biology, metastasis as a process is one of the most technically challenging aspects of cancer biology to study. As a result, new platforms and technologies are continually being developed to better understand this process. In this review, we discuss some of the recent advances in metastasis and how the information gleaned is re-shaping our understanding of metastatic dissemination.
Collapse
Affiliation(s)
- Jessica L Chitty
- Garvan Institute of Medical Research & the Kinghorn Cancer Centre, Cancer Division, Sydney, NSW, 2010, Australia
| | - Elysse C Filipe
- Garvan Institute of Medical Research & the Kinghorn Cancer Centre, Cancer Division, Sydney, NSW, 2010, Australia
| | - Morghan C Lucas
- Garvan Institute of Medical Research & the Kinghorn Cancer Centre, Cancer Division, Sydney, NSW, 2010, Australia
| | - David Herrmann
- Garvan Institute of Medical Research & the Kinghorn Cancer Centre, Cancer Division, Sydney, NSW, 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, NSW , 2010, Australia
| | - Thomas R Cox
- Garvan Institute of Medical Research & the Kinghorn Cancer Centre, Cancer Division, Sydney, NSW, 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, NSW , 2010, Australia
| | - Paul Timpson
- Garvan Institute of Medical Research & the Kinghorn Cancer Centre, Cancer Division, Sydney, NSW, 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, NSW , 2010, Australia
| |
Collapse
|
356
|
Chitty JL, Filipe EC, Lucas MC, Herrmann D, Cox TR, Timpson P. Recent advances in understanding the complexities of metastasis. F1000Res 2018; 7. [PMID: 30135716 PMCID: PMC6073095 DOI: 10.12688/f1000research.15064.2] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/05/2018] [Indexed: 12/14/2022] Open
Abstract
Tumour metastasis is a dynamic and systemic process. It is no longer seen as a tumour cell-autonomous program but as a multifaceted and complex series of events, which is influenced by the intrinsic cellular mutational burden of cancer cells and the numerous bidirectional interactions between malignant and non-malignant cells and fine-tuned by the various extrinsic cues of the extracellular matrix. In cancer biology, metastasis as a process is one of the most technically challenging aspects of cancer biology to study. As a result, new platforms and technologies are continually being developed to better understand this process. In this review, we discuss some of the recent advances in metastasis and how the information gleaned is re-shaping our understanding of metastatic dissemination.
Collapse
Affiliation(s)
- Jessica L Chitty
- Garvan Institute of Medical Research & the Kinghorn Cancer Centre, Cancer Division, Sydney, NSW, 2010, Australia
| | - Elysse C Filipe
- Garvan Institute of Medical Research & the Kinghorn Cancer Centre, Cancer Division, Sydney, NSW, 2010, Australia
| | - Morghan C Lucas
- Garvan Institute of Medical Research & the Kinghorn Cancer Centre, Cancer Division, Sydney, NSW, 2010, Australia
| | - David Herrmann
- Garvan Institute of Medical Research & the Kinghorn Cancer Centre, Cancer Division, Sydney, NSW, 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, NSW , 2010, Australia
| | - Thomas R Cox
- Garvan Institute of Medical Research & the Kinghorn Cancer Centre, Cancer Division, Sydney, NSW, 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, NSW , 2010, Australia
| | - Paul Timpson
- Garvan Institute of Medical Research & the Kinghorn Cancer Centre, Cancer Division, Sydney, NSW, 2010, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, NSW , 2010, Australia
| |
Collapse
|
357
|
Nowak-Sliwinska P, Alitalo K, Allen E, Anisimov A, Aplin AC, Auerbach R, Augustin HG, Bates DO, van Beijnum JR, Bender RHF, Bergers G, Bikfalvi A, Bischoff J, Böck BC, Brooks PC, Bussolino F, Cakir B, Carmeliet P, Castranova D, Cimpean AM, Cleaver O, Coukos G, Davis GE, De Palma M, Dimberg A, Dings RPM, Djonov V, Dudley AC, Dufton NP, Fendt SM, Ferrara N, Fruttiger M, Fukumura D, Ghesquière B, Gong Y, Griffin RJ, Harris AL, Hughes CCW, Hultgren NW, Iruela-Arispe ML, Irving M, Jain RK, Kalluri R, Kalucka J, Kerbel RS, Kitajewski J, Klaassen I, Kleinmann HK, Koolwijk P, Kuczynski E, Kwak BR, Marien K, Melero-Martin JM, Munn LL, Nicosia RF, Noel A, Nurro J, Olsson AK, Petrova TV, Pietras K, Pili R, Pollard JW, Post MJ, Quax PHA, Rabinovich GA, Raica M, Randi AM, Ribatti D, Ruegg C, Schlingemann RO, Schulte-Merker S, Smith LEH, Song JW, Stacker SA, Stalin J, Stratman AN, Van de Velde M, van Hinsbergh VWM, Vermeulen PB, Waltenberger J, Weinstein BM, Xin H, Yetkin-Arik B, Yla-Herttuala S, Yoder MC, Griffioen AW. Consensus guidelines for the use and interpretation of angiogenesis assays. Angiogenesis 2018; 21:425-532. [PMID: 29766399 PMCID: PMC6237663 DOI: 10.1007/s10456-018-9613-x] [Citation(s) in RCA: 455] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The formation of new blood vessels, or angiogenesis, is a complex process that plays important roles in growth and development, tissue and organ regeneration, as well as numerous pathological conditions. Angiogenesis undergoes multiple discrete steps that can be individually evaluated and quantified by a large number of bioassays. These independent assessments hold advantages but also have limitations. This article describes in vivo, ex vivo, and in vitro bioassays that are available for the evaluation of angiogenesis and highlights critical aspects that are relevant for their execution and proper interpretation. As such, this collaborative work is the first edition of consensus guidelines on angiogenesis bioassays to serve for current and future reference.
Collapse
Affiliation(s)
- Patrycja Nowak-Sliwinska
- Molecular Pharmacology Group, School of Pharmaceutical Sciences, Faculty of Sciences, University of Geneva, University of Lausanne, Rue Michel-Servet 1, CMU, 1211, Geneva 4, Switzerland.
- Translational Research Center in Oncohaematology, University of Geneva, Geneva, Switzerland.
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Biology Program, University of Helsinki, Helsinki, Finland
| | - Elizabeth Allen
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, VIB-Center for Cancer Biology, KU Leuven, Louvain, Belgium
| | - Andrey Anisimov
- Wihuri Research Institute and Translational Cancer Biology Program, University of Helsinki, Helsinki, Finland
| | - Alfred C Aplin
- Department of Pathology, University of Washington, Seattle, WA, USA
| | | | - Hellmut G Augustin
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Division of Vascular Oncology and Metastasis Research, German Cancer Research Center, Heidelberg, Germany
- German Cancer Consortium, Heidelberg, Germany
| | - David O Bates
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| | - Judy R van Beijnum
- Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - R Hugh F Bender
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | - Gabriele Bergers
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, VIB-Center for Cancer Biology, KU Leuven, Louvain, Belgium
- Department of Neurological Surgery, Brain Tumor Research Center, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Andreas Bikfalvi
- Angiogenesis and Tumor Microenvironment Laboratory (INSERM U1029), University Bordeaux, Pessac, France
| | - Joyce Bischoff
- Vascular Biology Program and Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Barbara C Böck
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Division of Vascular Oncology and Metastasis Research, German Cancer Research Center, Heidelberg, Germany
- German Cancer Consortium, Heidelberg, Germany
| | - Peter C Brooks
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, USA
| | - Federico Bussolino
- Department of Oncology, University of Torino, Turin, Italy
- Candiolo Cancer Institute-FPO-IRCCS, 10060, Candiolo, Italy
| | - Bertan Cakir
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Daniel Castranova
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Anca M Cimpean
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Ondine Cleaver
- Department of Molecular Biology, Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - George Coukos
- Ludwig Institute for Cancer Research, Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - George E Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, School of Medicine and Dalton Cardiovascular Center, Columbia, MO, USA
| | - Michele De Palma
- School of Life Sciences, Swiss Federal Institute of Technology, Lausanne, Switzerland
| | - Anna Dimberg
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Ruud P M Dings
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | | - Andrew C Dudley
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
- Emily Couric Cancer Center, The University of Virginia, Charlottesville, VA, USA
| | - Neil P Dufton
- Vascular Sciences, Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London, UK
| | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB Center for Cancer Biology, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute, Leuven, Belgium
| | | | - Marcus Fruttiger
- Institute of Ophthalmology, University College London, London, UK
| | - Dai Fukumura
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Bart Ghesquière
- Metabolomics Expertise Center, VIB Center for Cancer Biology, VIB, Leuven, Belgium
- Department of Oncology, Metabolomics Expertise Center, KU Leuven, Leuven, Belgium
| | - Yan Gong
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Robert J Griffin
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Adrian L Harris
- Molecular Oncology Laboratories, Oxford University Department of Oncology, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK
| | - Christopher C W Hughes
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | - Nan W Hultgren
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | | | - Melita Irving
- Ludwig Institute for Cancer Research, Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joanna Kalucka
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Robert S Kerbel
- Department of Medical Biophysics, Biological Sciences Platform, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Jan Kitajewski
- Department of Physiology and Biophysics, University of Illinois, Chicago, IL, USA
| | - Ingeborg Klaassen
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Hynda K Kleinmann
- The George Washington University School of Medicine, Washington, DC, USA
| | - Pieter Koolwijk
- Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Elisabeth Kuczynski
- Department of Medical Biophysics, Biological Sciences Platform, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Brenda R Kwak
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | | | - Juan M Melero-Martin
- Department of Cardiac Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Lance L Munn
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Roberto F Nicosia
- Department of Pathology, University of Washington, Seattle, WA, USA
- Pathology and Laboratory Medicine Service, VA Puget Sound Health Care System, Seattle, WA, USA
| | - Agnes Noel
- Laboratory of Tumor and Developmental Biology, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Jussi Nurro
- Department of Biotechnology and Molecular Medicine, University of Eastern Finland, Kuopio, Finland
| | - Anna-Karin Olsson
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Tatiana V Petrova
- Department of oncology UNIL-CHUV, Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Kristian Pietras
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund, Sweden
| | - Roberto Pili
- Genitourinary Program, Indiana University-Simon Cancer Center, Indianapolis, IN, USA
| | - Jeffrey W Pollard
- Medical Research Council Centre for Reproductive Health, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - Mark J Post
- Department of Physiology, Maastricht University, Maastricht, The Netherlands
| | - Paul H A Quax
- Einthoven Laboratory for Experimental Vascular Medicine, Department Surgery, LUMC, Leiden, The Netherlands
| | - Gabriel A Rabinovich
- Laboratory of Immunopathology, Institute of Biology and Experimental Medicine, National Council of Scientific and Technical Investigations (CONICET), Buenos Aires, Argentina
| | - Marius Raica
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Anna M Randi
- Vascular Sciences, Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London, UK
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
- National Cancer Institute "Giovanni Paolo II", Bari, Italy
| | - Curzio Ruegg
- Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Reinier O Schlingemann
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Stefan Schulte-Merker
- Institute of Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU, Münster, Germany
| | - Lois E H Smith
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Jonathan W Song
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Steven A Stacker
- Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre and The Sir Peter MacCallum, Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Jimmy Stalin
- Institute of Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU, Münster, Germany
| | - Amber N Stratman
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Maureen Van de Velde
- Laboratory of Tumor and Developmental Biology, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Victor W M van Hinsbergh
- Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Peter B Vermeulen
- HistoGeneX, Antwerp, Belgium
- Translational Cancer Research Unit, GZA Hospitals, Sint-Augustinus & University of Antwerp, Antwerp, Belgium
| | - Johannes Waltenberger
- Medical Faculty, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany
| | - Brant M Weinstein
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Hong Xin
- University of California, San Diego, La Jolla, CA, USA
| | - Bahar Yetkin-Arik
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Seppo Yla-Herttuala
- Department of Biotechnology and Molecular Medicine, University of Eastern Finland, Kuopio, Finland
| | - Mervin C Yoder
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
358
|
Comen EA, Bowman RL, Kleppe M. Underlying Causes and Therapeutic Targeting of the Inflammatory Tumor Microenvironment. Front Cell Dev Biol 2018; 6:56. [PMID: 29946544 PMCID: PMC6005853 DOI: 10.3389/fcell.2018.00056] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 05/11/2018] [Indexed: 12/13/2022] Open
Abstract
Historically, the link between chronic inflammation and cancer has long been speculated. Only more recently, pre-clinical and epidemiologic data as well as clinical evidence all point to the role of the tumor microenvironment as inextricably connected to the neoplastic process. The tumor microenvironment (TME), a complex mix of vasculature, inflammatory cells, and stromal cells is the essential "soil" helping to modulate tumor potential. Increasingly, evidence suggests that chronic inflammation modifies the tumor microenvironment, via a host of mechanisms, including the production of cytokines, pro-inflammatory mediators, angiogenesis, and tissue remodeling. Inflammation can be triggered by a variety of different pressures, such as carcinogen exposure, immune dysfunction, dietary habits, and obesity, as well as genetic alterations leading to oncogene activation or loss of tumor suppressors. In this review, we examine the concept of the tumor microenvironment as related to both extrinsic and intrinsic stimuli that promote chronic inflammation and in turn tumorigenesis. Understanding the common pathways inherent in an inflammatory response and the tumor microenvironment may shed light on new therapies for both primary and metastatic disease. The concept of personalized medicine has pushed the field of oncology to drill down on the genetic changes of a cancer, in the hopes of identifying individually targeted agents. Given the complexities of the tumor microenvironment, it is clear that effective oncologic therapies will necessitate targeting not only the cancer cells, but their dynamic relationship to the tumor microenvironment as well.
Collapse
Affiliation(s)
- Elizabeth A. Comen
- Breast Cancer Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Robert L. Bowman
- Center for Hematopoietic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Maria Kleppe
- Center for Hematopoietic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
359
|
Abstract
The concept that progression of cancer is regulated by interactions of cancer cells with their microenvironment was postulated by Stephen Paget over a century ago. Contemporary tumour microenvironment (TME) research focuses on the identification of tumour-interacting microenvironmental constituents, such as resident or infiltrating non-tumour cells, soluble factors and extracellular matrix components, and the large variety of mechanisms by which these constituents regulate and shape the malignant phenotype of tumour cells. In this Timeline article, we review the developmental phases of the TME paradigm since its initial description. While illuminating controversies, we discuss the importance of interactions between various microenvironmental components and tumour cells and provide an overview and assessment of therapeutic opportunities and modalities by which the TME can be targeted.
Collapse
Affiliation(s)
- Shelly Maman
- Department of Cell Research and Immunology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Isaac P Witz
- Department of Cell Research and Immunology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
360
|
Reeves MQ, Kandyba E, Harris S, Del Rosario R, Balmain A. Multicolour lineage tracing reveals clonal dynamics of squamous carcinoma evolution from initiation to metastasis. Nat Cell Biol 2018; 20:699-709. [PMID: 29802408 PMCID: PMC6400587 DOI: 10.1038/s41556-018-0109-0] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 04/24/2018] [Indexed: 12/30/2022]
Abstract
Tumor cells are subjected to evolutionary selection pressures during progression from initiation to metastasis. We analyzed the clonal evolution of squamous skin carcinomas induced by DMBA/TPA treatment using the K5CreER-Confetti mouse and stage-specific lineage tracing. We show that benign tumors are polyclonal, but only one population contains the Hras driver mutation. Benign papillomas are therefore monoclonal in origin, but recruit neighboring epithelial cells during growth. Papillomas that never progress to malignancy retain several distinct clones, whereas progression to carcinoma is associated with a clonal sweep. Newly generated clones within carcinomas demonstrate intratumoral invasion and clonal intermixing, often giving rise to metastases containing two or more distinct clones derived from the matched primary tumor. These data demonstrate that late stage tumor progression and dissemination are governed by evolutionary selection pressures that operate at a multicellular level, and thus differ from the clonal events that drive initiation and the benign-malignant transition.
Collapse
Affiliation(s)
- Melissa Q Reeves
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, CA, USA.
| | - Eve Kandyba
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, CA, USA
| | - Sophie Harris
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, CA, USA
| | - Reyno Del Rosario
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, CA, USA
| | - Allan Balmain
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, CA, USA.
| |
Collapse
|
361
|
Porrello A, Leslie PL, Harrison EB, Gorentla BK, Kattula S, Ghosh SK, Azam SH, Holtzhausen A, Chao YL, Hayward MC, Waugh TA, Bae S, Godfrey V, Randell SH, Oderup C, Makowski L, Weiss J, Wilkerson MD, Hayes DN, Earp HS, Baldwin AS, Wolberg AS, Pecot CV. Factor XIIIA-expressing inflammatory monocytes promote lung squamous cancer through fibrin cross-linking. Nat Commun 2018; 9:1988. [PMID: 29777108 PMCID: PMC5959879 DOI: 10.1038/s41467-018-04355-w] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 04/25/2018] [Indexed: 12/26/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide, and lung squamous carcinomas (LUSC) represent about 30% of cases. Molecular aberrations in lung adenocarcinomas have allowed for effective targeted treatments, but corresponding therapeutic advances in LUSC have not materialized. However, immune checkpoint inhibitors in sub-populations of LUSC patients have led to exciting responses. Using computational analyses of The Cancer Genome Atlas, we identified a subset of LUSC tumors characterized by dense infiltration of inflammatory monocytes (IMs) and poor survival. With novel, immunocompetent metastasis models, we demonstrated that tumor cell derived CCL2-mediated recruitment of IMs is necessary and sufficient for LUSC metastasis. Pharmacologic inhibition of IM recruitment had substantial anti-metastatic effects. Notably, we show that IMs highly express Factor XIIIA, which promotes fibrin cross-linking to create a scaffold for LUSC cell invasion and metastases. Consistently, human LUSC samples containing extensive cross-linked fibrin in the microenvironment correlated with poor survival. Lung squamous carcinomas (LUSC) are poorly molecularly characterized, but sub-populations show promising response to immune checkpoint inhibitors. Here, the authors identify a subset of LUSC characterized by infiltration of inflammatory monocytes, where metastasis is linked to Factor XIIIA promoting fibrin cross-linking.
Collapse
Affiliation(s)
- Alessandro Porrello
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Patrick L Leslie
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Emily B Harrison
- Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Balachandra K Gorentla
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Sravya Kattula
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Subrata K Ghosh
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Salma H Azam
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Alisha Holtzhausen
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Yvonne L Chao
- Division of Hematology & Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Michele C Hayward
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Trent A Waugh
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Sanggyu Bae
- Division of Hematology & Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Virginia Godfrey
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Scott H Randell
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.,Marsico Lung Institute/Cystic Fibrosis Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Cecilia Oderup
- Cancer Immunology, Pfizer, Inc, San Francisco, CA, 94080, USA
| | - Liza Makowski
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.,Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.,Nutrition Obesity Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Jared Weiss
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.,Division of Hematology & Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.,Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Matthew D Wilkerson
- Department of Anatomy, Physiology and Genetics, The American Genome Center, Collaborative Health Initiative Research Program, Uniformed Services University, Bethesda, MD, 20814, USA
| | - D Neil Hayes
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.,Division of Hematology & Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.,Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - H Shelton Earp
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.,Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.,Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Albert S Baldwin
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.,Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Alisa S Wolberg
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Chad V Pecot
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA. .,Division of Hematology & Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA. .,Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
362
|
Meirson T, Gil-Henn H. Targeting invadopodia for blocking breast cancer metastasis. Drug Resist Updat 2018; 39:1-17. [PMID: 30075834 DOI: 10.1016/j.drup.2018.05.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 05/04/2018] [Accepted: 05/15/2018] [Indexed: 12/13/2022]
Abstract
Dissemination of cancer cells from the primary tumor and their spread to distant sites of the body is the leading cause of mortality in metastatic cancer patients. Metastatic cancer cells invade surrounding tissues and blood vessels by forming F-actin-rich protrusions known as invadopodia, which degrade the extracellular matrix and enable invasion of tumor cells through it. Invadopodia have now been observed in vivo, and recent evidence demonstrates direct molecular links between assembly of invadopodia and cancer metastasis in both mouse models and in human patients. While significant progress has been achieved in the last decade in understanding the molecular mechanisms and signaling pathways regulating invadopodia formation and function, the application of this knowledge to development of prognostic and therapeutic approaches for cancer metastasis has not been discussed before. Here, we provide a detailed overview of current prognostic markers and tests for cancer metastasis and discuss their advantages, disadvantages, and their predicted efficiency. Using bioinformatic patient database analysis, we demonstrate, for the first time, a significant correlation between invadopodia-associated genes to breast cancer metastasis, suggesting that invadopodia could be used as both a prognostic marker and as a therapeutic target for blocking cancer metastasis. We include here a novel network interaction map of invadopodia-associated proteins with currently available inhibitors, demonstrating a central role for the recently identified EGFR-Pyk2-Src-Arg-cortactin invadopodial pathway, to which re-purposing of existent inhibitors could be used to block breast cancer metastasis. We then present an updated overview of current cancer-related clinical trials, demonstrating the negligible number of trials focusing on cancer metastasis. We also discuss the difficulties and complexity of performing cancer metastasis clinical trials, and the possible development of anti-metastasis drug resistance when using a prolonged preventive treatment with invadopodia inhibitors. This review presents a new perspective on invadopodia-mediated tumor invasiveness and may lead to the development of novel prognostic and therapeutic approaches for cancer metastasis.
Collapse
Affiliation(s)
- Tomer Meirson
- Laboratory of Cell Migration and Invasion, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel; Drug Discovery Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Hava Gil-Henn
- Laboratory of Cell Migration and Invasion, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel.
| |
Collapse
|
363
|
Norton KA, Jin K, Popel AS. Modeling triple-negative breast cancer heterogeneity: Effects of stromal macrophages, fibroblasts and tumor vasculature. J Theor Biol 2018; 452:56-68. [PMID: 29750999 DOI: 10.1016/j.jtbi.2018.05.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 04/13/2018] [Accepted: 05/03/2018] [Indexed: 12/20/2022]
Abstract
A hallmark of breast tumors is its spatial heterogeneity that includes its distribution of cancer stem cells and progenitor cells, but also heterogeneity in the tumor microenvironment. In this study we focus on the contributions of stromal cells, specifically macrophages, fibroblasts, and endothelial cells on tumor progression. We develop a computational model of triple-negative breast cancer based on our previous work and expand it to include macrophage infiltration, fibroblasts, and angiogenesis. In vitro studies have shown that the secretomes of tumor-educated macrophages and fibroblasts increase both the migration and proliferation rates of triple-negative breast cancer cells. In vivo studies also demonstrated that blocking signaling of selected secreted factors inhibits tumor growth and metastasis in mouse xenograft models. We investigate the influences of increased migration and proliferation rates on tumor growth, the effect of the presence on fibroblasts or macrophages on growth and morphology, and the contributions of macrophage infiltration on tumor growth. We find that while the presence of macrophages increases overall tumor growth, the increase in macrophage infiltration does not substantially increase tumor growth and can even stifle tumor growth at excessive rates.
Collapse
Affiliation(s)
| | - Kideok Jin
- Department of Biomedical Engineering; Department of Pharmaceutical Science, Albany College of Pharmacy and Health Science, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering; Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, USA
| |
Collapse
|
364
|
Alieva M, van Rheenen J, Broekman MLD. Potential impact of invasive surgical procedures on primary tumor growth and metastasis. Clin Exp Metastasis 2018; 35:319-331. [PMID: 29728948 PMCID: PMC6063335 DOI: 10.1007/s10585-018-9896-8] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 04/30/2018] [Indexed: 02/08/2023]
Abstract
Surgical procedures such as tumor resection and biopsy are still the gold standard for diagnosis and (determination of) treatment of solid tumors, and are prognostically beneficial for patients. However, growing evidence suggests that even a minor surgical trauma can influence several (patho) physiological processes that might promote postoperative metastatic spread and tumor recurrence. Local effects include tumor seeding and a wound healing response that can promote tumor cell migration, proliferation, differentiation, extracellular matrix remodeling, angiogenesis and extravasation. In addition, local and systemic immunosuppression impairs antitumor immunity and contributes to tumor cell survival. Surgical manipulation of the tumor can result in cancer cell release into the circulation, thus increasing the chance of tumor cell dissemination. To prevent these undesired effects of surgical interventions, therapeutic strategies targeting immune response exacerbation or alteration have been proposed. This review summarizes the current literature regarding these local, systemic and secondary site effects of surgical interventions on tumor progression and dissemination, and discusses studies that aimed to identify potential therapeutic approaches to prevent these effects in order to further increase the clinical benefit from surgical procedures.
Collapse
Affiliation(s)
- Maria Alieva
- Princess Máxima Center for Pediatric Oncology, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands.
| | - Jacco van Rheenen
- Department of Molecular Pathology, Oncode Institute, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Marike L D Broekman
- Department of Neurology & Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| |
Collapse
|
365
|
Stuelten CH, Parent CA, Montell DJ. Cell motility in cancer invasion and metastasis: insights from simple model organisms. Nat Rev Cancer 2018; 18:296-312. [PMID: 29546880 PMCID: PMC6790333 DOI: 10.1038/nrc.2018.15] [Citation(s) in RCA: 345] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Metastasis remains the greatest challenge in the clinical management of cancer. Cell motility is a fundamental and ancient cellular behaviour that contributes to metastasis and is conserved in simple organisms. In this Review, we evaluate insights relevant to human cancer that are derived from the study of cell motility in non-mammalian model organisms. Dictyostelium discoideum, Caenorhabditis elegans, Drosophila melanogaster and Danio rerio permit direct observation of cells moving in complex native environments and lend themselves to large-scale genetic and pharmacological screening. We highlight insights derived from each of these organisms, including the detailed signalling network that governs chemotaxis towards chemokines; a novel mechanism of basement membrane invasion; the positive role of E-cadherin in collective direction-sensing; the identification and optimization of kinase inhibitors for metastatic thyroid cancer on the basis of work in flies; and the value of zebrafish for live imaging, especially of vascular remodelling and interactions between tumour cells and host tissues. While the motility of tumour cells and certain host cells promotes metastatic spread, the motility of tumour-reactive T cells likely increases their antitumour effects. Therefore, it is important to elucidate the mechanisms underlying all types of cell motility, with the ultimate goal of identifying combination therapies that will increase the motility of beneficial cells and block the spread of harmful cells.
Collapse
Affiliation(s)
- Christina H. Stuelten
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA
| | - Carole A. Parent
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA
- Department of Pharmacology, Michigan Medicine, Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- ;
| | - Denise J. Montell
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA, USA
- ;
| |
Collapse
|
366
|
Arwert EN, Harney AS, Entenberg D, Wang Y, Sahai E, Pollard JW, Condeelis JS. A Unidirectional Transition from Migratory to Perivascular Macrophage Is Required for Tumor Cell Intravasation. Cell Rep 2018; 23:1239-1248. [PMID: 29719241 PMCID: PMC5946803 DOI: 10.1016/j.celrep.2018.04.007] [Citation(s) in RCA: 185] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 02/12/2018] [Accepted: 03/30/2018] [Indexed: 12/30/2022] Open
Abstract
Tumor-associated macrophages (TAMs) are critical for tumor metastasis. Two TAM subsets support cancer cell intravasation: migratory macrophages guide cancer cells toward blood vessels, where sessile perivascular macrophages assist their entry into the blood. However, little is known about the inter-relationship between these functionally distinct TAMs or their possible inter-conversion. We show that motile, streaming TAMs are newly arrived monocytes, recruited via CCR2 signaling, that then differentiate into the sessile perivascular macrophages. This unidirectional process is regulated by CXCL12 and CXCR4. Cancer cells induce TGF-β-dependent upregulation of CXCR4 in monocytes, while CXCL12 expressed by perivascular fibroblasts attracts these motile TAMs toward the blood vessels, bringing motile cancer cells with them. Once on the blood vessel, the migratory TAMs differentiate into perivascular macrophages, promoting vascular leakiness and intravasation.
Collapse
Affiliation(s)
- Esther N Arwert
- Tumour Cell Biology Laboratory, Francis Crick Institute, London, UK; Gruss-Lipper Biophotonics Center and the Integrated Imaging Program, Albert Einstein College of Medicine, New York, NY, USA
| | - Allison S Harney
- Gruss-Lipper Biophotonics Center and the Integrated Imaging Program, Albert Einstein College of Medicine, New York, NY, USA
| | - David Entenberg
- Gruss-Lipper Biophotonics Center and the Integrated Imaging Program, Albert Einstein College of Medicine, New York, NY, USA
| | - Yarong Wang
- Gruss-Lipper Biophotonics Center and the Integrated Imaging Program, Albert Einstein College of Medicine, New York, NY, USA
| | - Erik Sahai
- Tumour Cell Biology Laboratory, Francis Crick Institute, London, UK.
| | - Jeffrey W Pollard
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK; Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, NY 10461, USA.
| | - John S Condeelis
- Gruss-Lipper Biophotonics Center and the Integrated Imaging Program, Albert Einstein College of Medicine, New York, NY, USA.
| |
Collapse
|
367
|
Cabrera RM, Mao SPH, Surve CR, Condeelis JS, Segall JE. A novel neuregulin - jagged1 paracrine loop in breast cancer transendothelial migration. Breast Cancer Res 2018; 20:24. [PMID: 29636067 PMCID: PMC5894135 DOI: 10.1186/s13058-018-0960-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 03/21/2018] [Indexed: 01/01/2023] Open
Abstract
Background The interaction of breast cancer cells with other cells in the tumor microenvironment plays an important role in metastasis. Invasion and intravasation, two critical steps in the metastatic process, are influenced by these interactions. Macrophages are of particular interest when it comes to studying tumor cell invasiveness. Previous studies have shown that there is paracrine loop signaling between breast cancer cells and macrophages involving colony stimulating factor 1 (CSF-1) produced by tumor cells and epidermal growth factor (EGF) production by macrophages. In this paper, we identify a novel paracrine loop between tumor cells and macrophages involving neuregulin (NRG1) and notch signaling. Methods The aim of this study was to determine the role of NRG1, a ligand of the ErbB3 receptor, in macrophage stimulation of tumor cell transendothelial migration and intravasation. We used fluorescence-activated cell sorting (FACS) and western blot to determine ErbB3 and NRG1 expression, respectively. An in vitro transendothelial migration (iTEM) assay was used to examine the effects of short hairpin (sh)RNA targeting NRG1 in tumor cells and clustered regularly interspaced short palindromic repeats (CRISPR) knockout of jagged 1 (JAG1) in macrophages. Orthotopic xenograft injections in mice were used to confirm results in vivo. Results In our system, macrophages were the primary cells showing expression of ErbB3, and a blocking antibody against ErbB3 resulted in a significant decrease in macrophage-induced transendothelial migration of breast cancer cells. Stimulation of macrophages with NRG1 upregulated mRNA and protein expression of JAG1, a ligand of the Notch receptor, and JAG1 production by macrophages was important for transendothelial migration of tumor cells. Conclusions This study demonstrates that stimulation of macrophages by tumor cell NRG1 can enhance transendothelial migration and intravasation. We also demonstrate that this effect is due to induction of macrophage JAG1, an important ligand of the Notch signaling pathway.
Collapse
Affiliation(s)
- Ramon M Cabrera
- Department of Anatomy and Structural Biology, Price 201, Albert Einstein College of Medicine, 1301 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Serena P H Mao
- Department of Anatomy and Structural Biology, Price 201, Albert Einstein College of Medicine, 1301 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Chinmay R Surve
- Department of Anatomy and Structural Biology, Price 201, Albert Einstein College of Medicine, 1301 Morris Park Avenue, Bronx, NY, 10461, USA
| | - John S Condeelis
- Department of Anatomy and Structural Biology, Price 201, Albert Einstein College of Medicine, 1301 Morris Park Avenue, Bronx, NY, 10461, USA.,Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.,Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Jeffrey E Segall
- Department of Anatomy and Structural Biology, Price 201, Albert Einstein College of Medicine, 1301 Morris Park Avenue, Bronx, NY, 10461, USA. .,Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| |
Collapse
|
368
|
Bogorad MI, DeStefano J, Wong AD, Searson PC. Tissue-engineered 3D microvessel and capillary network models for the study of vascular phenomena. Microcirculation 2018; 24. [PMID: 28164421 DOI: 10.1111/micc.12360] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 01/29/2017] [Indexed: 12/13/2022]
Abstract
Advances in tissue engineering, cell biology, microfabrication, and microfluidics have led to the development of a wide range of vascular models. Here, we review platforms based on templated microvessel fabrication to generate increasingly complex vascular models of (i) the tumor microenvironment, (ii) occluded microvessels, and (iii) perfused capillary networks. We outline fabrication guidelines and demonstrate a number of experimental methods for probing vascular function such as permeability measurements, tumor cell intravasation, flow characterization, and endothelial cell morphology and proliferation.
Collapse
Affiliation(s)
- Max I Bogorad
- Institute for Nanobiotechnology (INBT), Johns Hopkins University, Baltimore, MD, USA.,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jackson DeStefano
- Institute for Nanobiotechnology (INBT), Johns Hopkins University, Baltimore, MD, USA.,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Andrew D Wong
- Institute for Nanobiotechnology (INBT), Johns Hopkins University, Baltimore, MD, USA.,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Peter C Searson
- Institute for Nanobiotechnology (INBT), Johns Hopkins University, Baltimore, MD, USA.,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
369
|
Pan Y, Ma P, Liu Y, Li W, Shu Y. Multiple functions of m 6A RNA methylation in cancer. J Hematol Oncol 2018; 11:48. [PMID: 29587823 PMCID: PMC5870302 DOI: 10.1186/s13045-018-0590-8] [Citation(s) in RCA: 239] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Accepted: 03/11/2018] [Indexed: 02/07/2023] Open
Abstract
First identified in 1974, m6A RNA methylation, which serves as a predominant internal modification of RNA in higher eukaryotes, has gained prodigious interest in recent years. Modifications of m6A are dynamic and reversible in mammalian cells, which have been proposed as another layer of epigenetic regulation similar to DNA and histone modifications. m6A RNA methylation is involved in all stages in the life cycle of RNA, ranging from RNA processing, through nuclear export, translation modulation to RNA degradation, which suggests its potential of influencing a plurality of aspects of RNA metabolism. All of the recent studies have pointed to a complicated regulation network of m6A modification in different tissues, cell lines, and space–time models. m6A methylation has been found to have an impact on tumor initiation and progression through various mechanisms. Furthermore, m6A RNA methylation has provided new opportunities for early stage diagnosis and treatment of cancers. Herein, we review the chemical basis of m6A RNA methylation, its multiple functions and potential significance in cancer.
Collapse
Affiliation(s)
- Yutian Pan
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Pei Ma
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Yu Liu
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Wei Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China. .,Department of Oncology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, People's Republic of China.
| | - Yongqian Shu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China. .,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, People's Republic of China. .,Department of Oncology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, People's Republic of China.
| |
Collapse
|
370
|
Miller MA, Chandra R, Cuccarese MF, Pfirschke C, Engblom C, Stapleton S, Adhikary U, Kohler RH, Mohan JF, Pittet MJ, Weissleder R. Radiation therapy primes tumors for nanotherapeutic delivery via macrophage-mediated vascular bursts. Sci Transl Med 2018; 9:9/392/eaal0225. [PMID: 28566423 DOI: 10.1126/scitranslmed.aal0225] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 01/23/2017] [Accepted: 04/24/2017] [Indexed: 12/13/2022]
Abstract
Efficient delivery of therapeutic nanoparticles (TNPs) to tumors is critical in improving efficacy, yet strategies that universally maximize tumoral targeting by TNP modification have been difficult to achieve in the clinic. Instead of focusing on TNP optimization, we show that the tumor microenvironment itself can be therapeutically primed to facilitate accumulation of multiple clinically relevant TNPs. Building on the recent finding that tumor-associated macrophages (TAM) can serve as nanoparticle drug depots, we demonstrate that local tumor irradiation substantially increases TAM relative to tumor cells and, thus, TNP delivery. High-resolution intravital imaging reveals that after radiation, TAM primarily accumulate adjacent to microvasculature, elicit dynamic bursts of extravasation, and subsequently enhance drug uptake in neighboring tumor cells. TAM depletion eliminates otherwise beneficial radiation effects on TNP accumulation and efficacy, and controls with unencapsulated drug show that radiation effects are more pronounced with TNPs. Priming with combined radiation and cyclophosphamide enhances vascular bursting and tumoral TNP concentration, in some cases leading to a sixfold increase of TNP accumulation in the tumor, reaching 6% of the injected dose per gram of tissue. Radiation therapy alters tumors for enhanced TNP delivery in a TAM-dependent fashion, and these observations have implications for the design of next-generation tumor-targeted nanomaterials and clinical trials for adjuvant strategies.
Collapse
Affiliation(s)
- Miles A Miller
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, MA 02114, USA.,Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
| | - Ravi Chandra
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, MA 02114, USA.,Harvard Radiation Oncology Program, 55 Fruit Street, Boston, MA 02114, USA
| | - Michael F Cuccarese
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, MA 02114, USA
| | - Christina Pfirschke
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, MA 02114, USA
| | - Camilla Engblom
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, MA 02114, USA
| | - Shawn Stapleton
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, MA 02114, USA
| | - Utsarga Adhikary
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, MA 02114, USA
| | - Rainer H Kohler
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, MA 02114, USA
| | - James F Mohan
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, MA 02114, USA
| | - Mikael J Pittet
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, MA 02114, USA.,Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, MA 02114, USA. .,Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA.,Department of Systems Biology, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
371
|
Li Y, Liu TM. Discovering Macrophage Functions Using In Vivo Optical Imaging Techniques. Front Immunol 2018; 9:502. [PMID: 29599778 PMCID: PMC5863475 DOI: 10.3389/fimmu.2018.00502] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 02/26/2018] [Indexed: 12/27/2022] Open
Abstract
Macrophages are an important component of host defense and inflammation and play a pivotal role in immune regulation, tissue remodeling, and metabolic regulation. Since macrophages are ubiquitous in human bodies and have versatile physiological functions, they are involved in virtually every disease, including cancer, diabetes, multiple sclerosis, and atherosclerosis. Molecular biological and histological methods have provided critical information on macrophage biology. However, many in vivo dynamic behaviors of macrophages are poorly understood and yet to be discovered. A better understanding of macrophage functions and dynamics in pathogenesis will open new opportunities for better diagnosis, prognostic assessment, and therapeutic intervention. In this article, we will review the advances in macrophage tracking and analysis with in vivo optical imaging in the context of different diseases. Moreover, this review will cover the challenges and solutions for optical imaging techniques during macrophage intravital imaging.
Collapse
Affiliation(s)
- Yue Li
- Faculty of Health Sciences, University of Macau, Macao, China
| | - Tzu-Ming Liu
- Faculty of Health Sciences, University of Macau, Macao, China
| |
Collapse
|
372
|
Ewald AJ. Metastasis inside-out: dissemination of cancer cell clusters with inverted polarity. EMBO J 2018. [PMID: 29523585 DOI: 10.15252/embj.201899144] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Andrew J Ewald
- Departments of Cell Biology and Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
373
|
Nobis M, Warren SC, Lucas MC, Murphy KJ, Herrmann D, Timpson P. Molecular mobility and activity in an intravital imaging setting - implications for cancer progression and targeting. J Cell Sci 2018; 131:131/5/jcs206995. [PMID: 29511095 DOI: 10.1242/jcs.206995] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Molecular mobility, localisation and spatiotemporal activity are at the core of cell biological processes and deregulation of these dynamic events can underpin disease development and progression. Recent advances in intravital imaging techniques in mice are providing new avenues to study real-time molecular behaviour in intact tissues within a live organism and to gain exciting insights into the intricate regulation of live cell biology at the microscale level. The monitoring of fluorescently labelled proteins and agents can be combined with autofluorescent properties of the microenvironment to provide a comprehensive snapshot of in vivo cell biology. In this Review, we summarise recent intravital microscopy approaches in mice, in processes ranging from normal development and homeostasis to disease progression and treatment in cancer, where we emphasise the utility of intravital imaging to observe dynamic and transient events in vivo We also highlight the recent integration of advanced subcellular imaging techniques into the intravital imaging pipeline, which can provide in-depth biological information beyond the single-cell level. We conclude with an outlook of ongoing developments in intravital microscopy towards imaging in humans, as well as provide an overview of the challenges the intravital imaging community currently faces and outline potential ways for overcoming these hurdles.
Collapse
Affiliation(s)
- Max Nobis
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2010, Australia
| | - Sean C Warren
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2010, Australia
| | - Morghan C Lucas
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2010, Australia
| | - Kendelle J Murphy
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2010, Australia
| | - David Herrmann
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2010, Australia
| | - Paul Timpson
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2010, Australia
| |
Collapse
|
374
|
Kiss M, Van Gassen S, Movahedi K, Saeys Y, Laoui D. Myeloid cell heterogeneity in cancer: not a single cell alike. Cell Immunol 2018; 330:188-201. [PMID: 29482836 DOI: 10.1016/j.cellimm.2018.02.008] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 02/10/2018] [Accepted: 02/11/2018] [Indexed: 12/14/2022]
Abstract
Tumors of various histological origins show abundant infiltration of myeloid cells from early stages of disease progression. These cells have a profound impact on antitumor immunity and influence fundamental processes that underlie malignancy, including neoangiogenesis, sustained cancer cell proliferation, metastasis and therapy resistance. For these reasons, development of therapeutic approaches to deplete or reprogram myeloid cells in cancer is an emerging field of interest. However, knowledge about the heterogeneity of myeloid cells in tumors and their variability between patients and disease stages is still limited. In this review, we summarize the most recent advances in our understanding about how the phenotype of tumor-associated macrophages, monocytes, neutrophils, myeloid-derived suppressor cells and dendritic cells is dictated by their ontogeny, activation status and localization. We also outline major open questions that will only be resolved by applying high-dimensional single-cell technologies and systems biology approaches in the analysis of the tumor microenvironment.
Collapse
Affiliation(s)
- Mate Kiss
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, Brussels, Belgium.
| | - Sofie Van Gassen
- IDLab, Department of Information Technology, Ghent University - IMEC, Ghent, Belgium; Data Mining and Modeling for Biomedicine, VIB Center for Inflammation Research, Ghent, Belgium
| | - Kiavash Movahedi
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, Brussels, Belgium
| | - Yvan Saeys
- Data Mining and Modeling for Biomedicine, VIB Center for Inflammation Research, Ghent, Belgium; Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Damya Laoui
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, Brussels, Belgium.
| |
Collapse
|
375
|
Jungwirth U, van Weverwijk A, Melake MJ, Chambers AF, Gao Q, Fivaz M, Isacke CM. Generation and characterisation of two D2A1 mammary cancer sublines to model spontaneous and experimental metastasis in a syngeneic BALB/c host. Dis Model Mech 2018; 11:dmm.031740. [PMID: 29208627 PMCID: PMC5818081 DOI: 10.1242/dmm.031740] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 11/17/2017] [Indexed: 12/20/2022] Open
Abstract
Studying the complex mechanisms underlying breast cancer metastasis and therapy response necessitates relevant in vivo models, particularly syngeneic models with an intact immune system. Two syngeneic spontaneously metastatic sublines, D2A1-m1 and D2A1-m2, were generated from the poorly metastasising BALB/c-derived D2A1 cell line by serial in vivo passaging. In vivo and in vitro analyses revealed distinct and shared characteristics of the metastatic D2A1-m1 and D2A1-m2 sublines. In particular, D2A1-m1 cells are more aggressive in experimental metastasis assays, while D2A1-m2 cells are more efficient at disseminating from the primary tumour in spontaneous metastasis assays. Surprisingly, classical metastasis-associated in vitro phenotypes, such as enhanced proliferation, migration and invasion, are reduced in the sublines compared to the parental cell line. Further, evasion of immune control cannot fully explain their enhanced metastatic properties. By contrast, both sublines show increased resistance to apoptosis when cultured in non-adherent conditions and, for the D2A1-m2 subline, increased 3D tumour spheroid growth. Moreover, the enhanced spontaneous metastatic phenotype of the D2A1-m2 subline is associated with an increased ability to recruit an activated tumour stroma. The metastatic D2A1-m1 and D2A1-m2 cell lines provide additional syngeneic models for investigating the different steps of the metastatic cascade and thereby represent valuable tools for breast cancer researchers. Finally, this study highlights that morphology and cell behaviour in 2D cell-based assays cannot be used as a reliable predictor of metastatic behaviour in vivo. Summary: We describe two D2A1 mouse mammary cancer sublines with enhanced spontaneous metastasis in a syngeneic host, and highlight the limitations of in vitro assays to predict in vivo metastatic behaviour.
Collapse
Affiliation(s)
- Ute Jungwirth
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK
| | - Antoinette van Weverwijk
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK
| | - Miriam J Melake
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK.,German Cancer Research Center, DKFZ, 69120, Heidelberg, Germany
| | - Ann F Chambers
- Department of Oncology, University of Western Ontario, London, Ontario, N6A 4L6, Canada
| | - Qiong Gao
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK
| | - Marc Fivaz
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK
| | - Clare M Isacke
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK
| |
Collapse
|
376
|
Nordgård O, Tjensvoll K, Gilje B, Søreide K. Circulating tumour cells and DNA as liquid biopsies in gastrointestinal cancer. Br J Surg 2018; 105:e110-e120. [DOI: 10.1002/bjs.10782] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 10/02/2017] [Accepted: 11/09/2017] [Indexed: 12/15/2022]
Abstract
Abstract
Background
Blood is the most extensively studied body fluid and, because it contains circulating tumour cells (CTCs) and circulating tumour-derived cell-free DNA (ctDNA), it may represent a liquid biopsy for cancer. Methods for enrichment and detection of CTCs and ctDNA, their clinical applications and future opportunities in gastrointestinal cancers were the focus of this review.
Methods
The PubMed database was searched for literature up to 24 June 2017, with a focus on the past 10 years. Identified articles were further scrutinized for relevant references. Articles were those in English relating to colorectal, gastric and pancreatic cancer.
Results
Both CTCs and ctDNA are in low abundance compared with other cellular components of blood, but effective enrichment and highly sensitive techniques are available for their detection. Potential clinical applications of these liquid biopsies include screening, prognostic stratification, therapy administration, monitoring of treatment effect or resistance, and surveillance. Liquid biopsies provide opportunities to reduce the need for invasive tissue sampling, especially in the context of intratumoral heterogeneity and the need for tumour genotyping.
Conclusion
Liquid biopsies have applications in gastrointestinal cancers to improve clinical decision-making.
Collapse
Affiliation(s)
- O Nordgård
- Department of Haematology and Oncology, Stavanger University Hospital, Stavanger, Norway
- Department of Mathematics and Natural Science, University of Stavanger, Stavanger, Norway
| | - K Tjensvoll
- Department of Haematology and Oncology, Stavanger University Hospital, Stavanger, Norway
| | - B Gilje
- Department of Haematology and Oncology, Stavanger University Hospital, Stavanger, Norway
| | - K Søreide
- Department of Gastrointestinal Surgery, Stavanger University Hospital, Stavanger, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Clinical Surgery, Royal Infirmary of Edinburgh and University of Edinburgh, Edinburgh, UK
| |
Collapse
|
377
|
Chemotherapy-induced metastasis: mechanisms and translational opportunities. Clin Exp Metastasis 2018; 35:269-284. [PMID: 29307118 DOI: 10.1007/s10585-017-9870-x] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 12/20/2017] [Indexed: 12/13/2022]
Abstract
Tumors often overcome the cytotoxic effects of chemotherapy through either acquired or environment-mediated drug resistance. In addition, signals from the microenvironment obfuscate the beneficial effects of chemotherapy and may facilitate progression and metastatic dissemination. Seminal mediators in chemotherapy-induced metastasis appear to be a wide range of hematopoietic, mesenchymal and immune progenitor cells, originating from the bone marrow. The actual purpose of these cells is to orchestrate the repair response to the cytotoxic damage of chemotherapy. However, these repair responses are exploited by tumor cells at every step of the metastatic cascade, ranging from tumor cell invasion, intravasation and hematogenous dissemination to extravasation and effective colonization at the metastatic site. A better understanding of the mechanistic underpinnings of chemotherapy-induced metastasis will allow us to better predict which patients are more likely to exhibit pro-metastatic responses to chemotherapy and will help develop new therapeutic strategies to neutralize chemotherapy-driven prometastatic changes.
Collapse
|
378
|
Linde N, Casanova-Acebes M, Sosa MS, Mortha A, Rahman A, Farias E, Harper K, Tardio E, Reyes Torres I, Jones J, Condeelis J, Merad M, Aguirre-Ghiso JA. Macrophages orchestrate breast cancer early dissemination and metastasis. Nat Commun 2018; 9:21. [PMID: 29295986 PMCID: PMC5750231 DOI: 10.1038/s41467-017-02481-5] [Citation(s) in RCA: 313] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 12/04/2017] [Indexed: 02/07/2023] Open
Abstract
Cancer cell dissemination during very early stages of breast cancer proceeds through poorly understood mechanisms. Here we show, in a mouse model of HER2+ breast cancer, that a previously described sub-population of early-evolved cancer cells requires macrophages for early dissemination. Depletion of macrophages specifically during pre-malignant stages reduces early dissemination and also results in reduced metastatic burden at end stages of cancer progression. Mechanistically, we show that, in pre-malignant lesions, CCL2 produced by cancer cells and myeloid cells attracts CD206+/Tie2+ macrophages and induces Wnt-1 upregulation that in turn downregulates E-cadherin junctions in the HER2+ early cancer cells. We also observe macrophage-containing tumor microenvironments of metastasis structures in the pre-malignant lesions that can operate as portals for intravasation. These data support a causal role for macrophages in early dissemination that affects long-term metastasis development much later in cancer progression. A pilot analysis on human specimens revealed intra-epithelial macrophages and loss of E-cadherin junctions in ductal carcinoma in situ, supporting a potential clinical relevance.
Collapse
Affiliation(s)
- Nina Linde
- Division of Hematology and Oncology, Department of Medicine, Tisch Cancer Institute, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Department of Otolaryngology, Tisch Cancer Institute, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Merck KGaA, Frankfurter Str. 250, Postcode: A025/301, Darmstadt, 64293, Germany
| | - Maria Casanova-Acebes
- Department of Oncological Sciences, The Immunology Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Maria Soledad Sosa
- Division of Hematology and Oncology, Department of Medicine, Tisch Cancer Institute, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Department of Otolaryngology, Tisch Cancer Institute, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Arthur Mortha
- Department of Oncological Sciences, The Immunology Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8, USA
| | - Adeeb Rahman
- Human Immune Monitoring Core, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Eduardo Farias
- Division of Hematology and Oncology, Department of Medicine, Tisch Cancer Institute, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Department of Otolaryngology, Tisch Cancer Institute, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Kathryn Harper
- Division of Hematology and Oncology, Department of Medicine, Tisch Cancer Institute, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Department of Otolaryngology, Tisch Cancer Institute, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ethan Tardio
- Division of Hematology and Oncology, Department of Medicine, Tisch Cancer Institute, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Department of Otolaryngology, Tisch Cancer Institute, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ivan Reyes Torres
- Department of Oncological Sciences, The Immunology Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Joan Jones
- Department of Anatomy and Structural Biology, Integrated Imaging Program, Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - John Condeelis
- Department of Anatomy and Structural Biology, Integrated Imaging Program, Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Miriam Merad
- Department of Oncological Sciences, The Immunology Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Human Immune Monitoring Core, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Julio A Aguirre-Ghiso
- Division of Hematology and Oncology, Department of Medicine, Tisch Cancer Institute, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA. .,Department of Otolaryngology, Tisch Cancer Institute, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
379
|
Paracrine interactions of cancer-associated fibroblasts, macrophages and endothelial cells: tumor allies and foes. Curr Opin Oncol 2018; 30:45-53. [DOI: 10.1097/cco.0000000000000420] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
380
|
Katt ME, Wong AD, Searson PC. Dissemination from a Solid Tumor: Examining the Multiple Parallel Pathways. Trends Cancer 2018; 4:20-37. [PMID: 29413419 PMCID: PMC5806201 DOI: 10.1016/j.trecan.2017.12.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 12/04/2017] [Accepted: 12/05/2017] [Indexed: 12/23/2022]
Abstract
Metastasis can be generalized as a linear sequence of events whereby halting one or more steps in the cascade may reduce tumor cell dissemination and ultimately improve patient outcomes. However, metastasis is a complex process with multiple parallel mechanisms of dissemination. Clinical strategies focus on removing the primary tumor and/or treating distant metastases through chemo- or immunotherapies. Successful strategies for blocking metastasis will need to address the parallel mechanisms of dissemination and identify common bottlenecks. Here, we review the current understanding of common dissemination pathways for tumors. Understanding the complexities of metastasis will guide the design of new therapies that halt dissemination.
Collapse
Affiliation(s)
- Moriah E Katt
- Institute for Nanobiotechnology, 100 Croft Hall, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; These authors contributed equally
| | - Andrew D Wong
- Institute for Nanobiotechnology, 100 Croft Hall, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; These authors contributed equally
| | - Peter C Searson
- Institute for Nanobiotechnology, 100 Croft Hall, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| |
Collapse
|
381
|
Abstract
Macrophages are a heterogeneous population of innate immune cells and are distributed in most adult tissues. Certain tissue-resident macrophages with a prenatal origin, together with postnatal monocyte-derived macrophages, serve as the host scavenger system to eliminate invading pathogens, malignant cells, senescent cells, dead cells, cellular debris, and other foreign substances. As a key member of the mononuclear phagocyte system, macrophages play essential roles in regulation of prenatal development, tissue homeostasis, and disease progression. Over the past two decades, considerable efforts have been made to generate genetic models of macrophage ablation in mice. These models support investigations of the precise functions of tissue-specific macrophages under physiological and pathological conditions. Herein, we overview the currently available mouse strains for in vivo genetic ablation of macrophages and discuss their respective advantages and limitations.
Collapse
Affiliation(s)
- Li Hua
- The Jackson Laboratory, Bar Harbor, ME, USA
| | | | | | | |
Collapse
|
382
|
Abstract
The epithelial-mesenchymal transition (EMT) plays an important role in development and cancer progression. Upon EMT, epithelial cells lose stable cell-cell adhesions and reorganize their cytoskeleton to acquire migratory activity. Recent data demonstrated that EMT drives cancer cells from the epithelial state to a hybrid epithelial/mesenchymal phenotype with retention of some epithelial markers (in particular, E-cadherin), which is important for cancer cell dissemination. In vitro studies of the effect of growth factors (in particular, epidermal growth factor (EGF)) on cultured cells can be highly advantageous for understanding the details of the early stages of EMT. The methods described in this chapter are intended for studying intermediate phenotypes of EMT. Time-lapse DIC microscopy is used for visualization of changes in morphology and motility of the cells stimulated with EGF. The transwell migration assay allows the evaluation of the migratory activity of the cells. Studying of dynamics of a fluorescently labeled actin-binding protein F-tractin-tdTomato using confocal microscopy allows detection of EGF-induced changes in the organization of the actin cytoskeleton. Live-cell imaging of cells stably expressing GFP-E-cadherin visualizes reorganization of stable tangential E-cadherin-based adherens junctions (AJs) into unstable radial AJs during the early stages of EMT.
Collapse
|
383
|
Bayarmagnai B, Perrin L, Esmaeili Pourfarhangi K, Gligorijevic B. Intravital Imaging of Tumor Cell Motility in the Tumor Microenvironment Context. Methods Mol Biol 2018; 1749:175-193. [PMID: 29525998 PMCID: PMC5996994 DOI: 10.1007/978-1-4939-7701-7_14] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cancer cell motility and invasion are key features of metastatic tumors. Both are highly linked to tumor microenvironmental parameters, such as collagen architecture or macrophage density. However, due to the genetic, epigenetic and microenvironmental heterogeneities, only a small portion of tumor cells in the primary tumor are motile and furthermore, only a small portion of those will metastasize. This creates a challenge in predicting metastatic fate of single cells based on the phenotype they exhibit in the primary tumor. To overcome this challenge, tumor cell subpopulations need to be monitored at several timescales, mapping their phenotype in primary tumor as well as their potential homing to the secondary tumor site. Additionally, to address the spatial heterogeneity of the tumor microenvironment and how it relates to tumor cell phenotypes, large numbers of images need to be obtained from the same tumor. Finally, as the microenvironment complexity results in nonlinear relationships between tumor cell phenotype and its surroundings, advanced statistical models are required to interpret the imaging data. Toward improving our understanding of the relationship between cancer cell motility, the tumor microenvironment context and successful metastasis, we have developed several intravital approaches for continuous and longitudinal imaging, as well as data classification via support vector machine (SVM) algorithm. We also describe methods that extend the capabilities of intravital imaging by postsacrificial microscopy of the lung as well as correlative immunofluorescence in the primary tumor.
Collapse
Affiliation(s)
| | - Louisiane Perrin
- Department of Bioengineering, Temple University, Philadelphia, PA, USA
| | | | - Bojana Gligorijevic
- Department of Bioengineering, Temple University, Philadelphia, PA, USA.
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
384
|
Cortes-Santiago N, Hossain MB, Gabrusiewicz K, Fan X, Gumin J, Marini FC, Alonso MM, Lang F, Yung WK, Fueyo J, Gomez-Manzano C. Soluble Tie2 overrides the heightened invasion induced by anti-angiogenesis therapies in gliomas. Oncotarget 2017; 7:16146-57. [PMID: 26910374 PMCID: PMC4941303 DOI: 10.18632/oncotarget.7550] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 01/29/2016] [Indexed: 11/25/2022] Open
Abstract
Glioblastoma recurrence after treatment with the anti–vascular endothelial growth factor (VEGF) agent bevacizumab is characterized by a highly infiltrative and malignant behavior that renders surgical excision and chemotherapy ineffective. Our group has previously reported that Tie2-expressing monocytes (TEMs) are aberrantly present at the tumor/normal brain interface after anti-VEGF therapies and their significant role in the invasive outgrowth of these tumors. Here, we aimed to further understand the mechanisms leading to this pro-invasive tumor microenvironment. Examination of a U87MG xenogeneic glioma model and a GL261 murine syngeneic model showed increased tumor expression of angiopoietin 2 (Ang2), a natural ligand of Tie2, after anti-angiogenesis therapies targeting VEGF or VEGF receptor (VEGFR), as assessed by immunohistochemical analysis, immunofluorescence analysis, and enzyme-linked immunosorbent assays of tumor lysates. Migration and gelatinolytic assays showed that Ang2 acts as both a chemoattractant of TEMs and an enhancing signal for their tumor-remodeling properties. Accordingly, in vivo transduction of Ang2 into intracranial gliomas increased recruitment of TEMs into the tumor. To reduce invasive tumor outgrowth after anti-angiogenesis therapy, we targeted the Ang-Tie2 axis using a Tie2 decoy receptor. Using syngeneic models, we observed that overexpression of soluble Tie2 within the tumor prevented the recruitment of TEMs to the tumor and the development of invasion after anti-angiogenesis treatment. Taken together, these data indicate an active role for the Ang2-Tie2 pathway in invasive glioma recurrence after anti-angiogenesis treatment and provide a rationale for testing the combined targeting of VEGF and Ang-Tie2 pathways in patients with glioblastoma.
Collapse
Affiliation(s)
- Nahir Cortes-Santiago
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Cancer Biology Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA
| | - Mohammad B Hossain
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Konrad Gabrusiewicz
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xuejun Fan
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joy Gumin
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Frank C Marini
- Comprehensive Cancer Center, Wake Forest University, Winston-Salem, NC, USA
| | - Marta M Alonso
- Department of Medical Oncology, University Hospital of Navarra, Pamplona, Spain
| | - Frederick Lang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - W K Yung
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Juan Fueyo
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Candelaria Gomez-Manzano
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Cancer Biology Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA
| |
Collapse
|
385
|
Qian BZ. Inflammation fires up cancer metastasis. Semin Cancer Biol 2017; 47:170-176. [PMID: 28838845 DOI: 10.1016/j.semcancer.2017.08.006] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 08/08/2017] [Accepted: 08/09/2017] [Indexed: 12/16/2022]
Abstract
Metastatic disease is the major challenge of cancer that accounts for over 90% of total cancer lethality. Mounting clinical and preclinical data now indicate that inflammation, a potent immune and repair response, is indispensable for metastasis. In this review we describe our current understanding of how major inflammatory cells contribute to metastatic cascade with a focus on the primary tumour. We also discuss exciting new directions for future research and novel therapeutic approaches to tackle metastatic disease through targeting inflammation.
Collapse
Affiliation(s)
- Bin-Zhi Qian
- University of Edinburgh and MRC Centre for Reproductive Health, EH16 4TJ, Edinburgh, United Kingdom; Edinburgh Cancer Research UK Centre, EH16 4TJ, Edinburgh, United Kingdom.
| |
Collapse
|
386
|
Medwig TN, Matus DQ. Breaking down barriers: the evolution of cell invasion. Curr Opin Genet Dev 2017; 47:33-40. [PMID: 28881331 PMCID: PMC5716887 DOI: 10.1016/j.gde.2017.08.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 08/02/2017] [Accepted: 08/11/2017] [Indexed: 12/12/2022]
Abstract
Cell invasion is a specialized cell behavior that likely co-evolved with the emergence of basement membranes in metazoans as a mechanism to break down the barriers that separate tissues. A variety of conserved and lineage-specific biological processes that occur during development and homeostasis rely on cell invasive behavior. Recent innovations in genome editing and live-cell imaging have shed some light on the programs that mediate acquisition of an invasive phenotype; however, comparative approaches among species are necessary to understand how this cell behavior evolved. Here, we discuss the contexts of cell invasion, highlighting both established and emerging model systems, and underscore gaps in our understanding of the evolution of this key cellular behavior.
Collapse
Affiliation(s)
- Taylor N Medwig
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA
| | - David Q Matus
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA.
| |
Collapse
|
387
|
Entenberg D, Voiculescu S, Guo P, Borriello L, Wang Y, Karagiannis GS, Jones J, Baccay F, Oktay M, Condeelis J. A permanent window for the murine lung enables high-resolution imaging of cancer metastasis. Nat Methods 2017; 15:73-80. [PMID: 29176592 DOI: 10.1038/nmeth.4511] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Accepted: 10/25/2017] [Indexed: 12/26/2022]
Abstract
Stable, high-resolution intravital imaging of the lung has become possible through the utilization of vacuum-stabilized imaging windows. However, this technique is extremely invasive and limited to only hours in duration. Here we describe a minimally invasive, permanently implantable window for high-resolution intravital imaging of the murine lung that allows the mouse to survive surgery, recover from anesthesia, and breathe independently. Compared to vacuum-stabilized windows, this window produces the same high-quality images without vacuum-induced artifacts; it is also less invasive, which allows imaging of the same lung tissue over a period of weeks. We further adapt the technique of microcartography for reliable relocalization of the same cells longitudinally. Using commonly employed experimental, as well as more clinically relevant, spontaneous metastasis models, we visualize all stages of metastatic seeding, including: tumor cell arrival; extravasation; growth and progression to micrometastases; as well as tumor microenvironment of metastasis function, the hallmark of hematogenous dissemination of tumor cells.
Collapse
Affiliation(s)
- David Entenberg
- Anatomy and Structural Biology, Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA.,Gruss-Lipper Biophotonics Center, Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA.,Integrated Imaging Program, Einstein College of Medicine, Montefiore Medical Center, Bronx,New York, USA
| | - Sonia Voiculescu
- Anatomy and Structural Biology, Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA.,Department of Surgery, Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA
| | - Peng Guo
- Anatomy and Structural Biology, Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA.,Gruss-Lipper Biophotonics Center, Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA.,Analytical Imaging Facility, Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA
| | - Lucia Borriello
- Anatomy and Structural Biology, Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA.,Gruss-Lipper Biophotonics Center, Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA
| | - Yarong Wang
- Anatomy and Structural Biology, Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA.,Gruss-Lipper Biophotonics Center, Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA.,Integrated Imaging Program, Einstein College of Medicine, Montefiore Medical Center, Bronx,New York, USA
| | - George S Karagiannis
- Anatomy and Structural Biology, Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA.,Gruss-Lipper Biophotonics Center, Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA.,Integrated Imaging Program, Einstein College of Medicine, Montefiore Medical Center, Bronx,New York, USA
| | - Joan Jones
- Anatomy and Structural Biology, Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA.,Gruss-Lipper Biophotonics Center, Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA.,Integrated Imaging Program, Einstein College of Medicine, Montefiore Medical Center, Bronx,New York, USA.,Department of Pathology, Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA
| | - Francis Baccay
- Department of Surgery, Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA
| | - Maja Oktay
- Anatomy and Structural Biology, Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA.,Gruss-Lipper Biophotonics Center, Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA.,Integrated Imaging Program, Einstein College of Medicine, Montefiore Medical Center, Bronx,New York, USA.,Department of Pathology, Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA
| | - John Condeelis
- Anatomy and Structural Biology, Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA.,Gruss-Lipper Biophotonics Center, Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA.,Integrated Imaging Program, Einstein College of Medicine, Montefiore Medical Center, Bronx,New York, USA
| |
Collapse
|
388
|
Abstract
This review by Micalizzi et al. discusses advances in analysis and characterization of circulating tumor cells (CTCs) from patients with cancer. It focuses on the current knowledge of CTC biology and the potential clinical implications. Advances in the enrichment and analysis of rare cells from the bloodstream have allowed for detection and characterization of circulating tumor cells (CTCs) from patients with cancer. The analysis of CTCs has provided significant insight into the metastatic process. Studies on the biology of CTCs have begun to elucidate the molecular mechanisms of CTC generation, intravasation, survival, interactions with components of the blood, extravasation, and colonization of distant organs. Additionally, the study of CTCs has exposed dramatic intrapatient and interpatient heterogeneity and their evolution over time. In this review, we focus on the current knowledge of CTC biology and the potential clinical implications.
Collapse
Affiliation(s)
- Douglas S Micalizzi
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts, 02129, USA.,Department of Medicine, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Shyamala Maheswaran
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts, 02129, USA.,Department of Surgery, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Daniel A Haber
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts, 02129, USA.,Department of Medicine, Harvard Medical School, Charlestown, Massachusetts 02129, USA.,Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
| |
Collapse
|
389
|
Chen J, Sun Y, Xu X, Wang D, He J, Zhou H, Lu Y, Zeng J, Du F, Gong A, Xu M. YTH domain family 2 orchestrates epithelial-mesenchymal transition/proliferation dichotomy in pancreatic cancer cells. Cell Cycle 2017; 16:2259-2271. [PMID: 29135329 DOI: 10.1080/15384101.2017.1380125] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Recent studies show that YTH domain family 2 (YTHDF2) preferentially binds to m6A-containing mRNA regulates localization and stability of the bound mRNA. However, the role of YTHDF2 in pancreatic cancers remains to be elucidated. Here, we find that YTHDF2 expression is up-regulated in pancreatic cancer tissues compared with normal tissues at both mRNA and protein levels, and is higher in clinical patients with later stages of pancreatic cancer, indicating that YTHDF2 possesses potential clinical significance for diagnosis and prognosis of pancreatic cancers. Furthermore, we find that YTHDF2 orchestrates two cellular processes: promotes proliferation and inhibits migration and invasion in pancreatic cancer cells, a phenomenon called "migration-proliferation dichotomy", as well as epithelial-mesenchymal transition (EMT) in pancreatic cancer cells. Furthermore, YTHDF2 knockdown significantly increases the total YAP expression, but inhibits TGF-β/Smad signaling, indicating that YTHDF2 regulates EMT probably via YAP signaling. In summary, all these findings suggest that YTHDF2 may be a new predictive biomarker of development of pancreatic cancer, but a serious consideration is needed to treat YTHDF2 as a target for pancreatic cancer.
Collapse
Affiliation(s)
- Jixiang Chen
- a Department of General Surgery , Affiliated Hospital of Jiangsu University, Jiangsu University , Zhenjiang , Jiangsu , China
| | - Yaocheng Sun
- a Department of General Surgery , Affiliated Hospital of Jiangsu University, Jiangsu University , Zhenjiang , Jiangsu , China
| | - Xiao Xu
- b Department of Cell Biology, School of Medicine , Jiangsu University , Zhenjiang , Jiangsu , China
| | - Dawei Wang
- c Department of Gastroenterology , Affiliated Hospital of Jiangsu University, Jiangsu University , Zhenjiang , Jiangsu , China
| | - Junbo He
- c Department of Gastroenterology , Affiliated Hospital of Jiangsu University, Jiangsu University , Zhenjiang , Jiangsu , China
| | - Hailang Zhou
- c Department of Gastroenterology , Affiliated Hospital of Jiangsu University, Jiangsu University , Zhenjiang , Jiangsu , China
| | - Ying Lu
- c Department of Gastroenterology , Affiliated Hospital of Jiangsu University, Jiangsu University , Zhenjiang , Jiangsu , China
| | - Jian Zeng
- b Department of Cell Biology, School of Medicine , Jiangsu University , Zhenjiang , Jiangsu , China
| | - Fengyi Du
- b Department of Cell Biology, School of Medicine , Jiangsu University , Zhenjiang , Jiangsu , China
| | - Aihua Gong
- b Department of Cell Biology, School of Medicine , Jiangsu University , Zhenjiang , Jiangsu , China
| | - Min Xu
- c Department of Gastroenterology , Affiliated Hospital of Jiangsu University, Jiangsu University , Zhenjiang , Jiangsu , China
| |
Collapse
|
390
|
Abstract
Human cancers exhibit formidable molecular heterogeneity, to a large extent accounting for the incomplete and transitory efficacy of current anti-cancer therapies. However, neoplastic cells alone do not manifest the disease, but conscript a battery of non-tumor cells to enable and sustain hallmark capabilities of cancer. Escaping immunosurveillance is one of such capabilities. Tumors evolve immunosuppressive microenvironment to subvert anti-tumor immunity. In this review, we will focus on tumor-associated myeloid cells, which constitute an essential part of the immune microenvironment and reciprocally interact with cancer cells to establish malignancy toward metastasis. The diversity and plasticity of these cells constitute another layer of heterogeneity, beyond the heterogeneity of cancer cells themselves. We envision that immune microenvironment co-evolves with the genetic heterogeneity of tumor. Addressing the question of how genetically distinct tumors shape and are shaped by unique immune microenvironment will provide an attractive rationale to develop novel immunotherapeutic modalities. Here, we discuss the complex nature of tumor microenvironment, with an emphasis on the cellular and functional heterogeneity among tumor-associated myeloid cells as well as immune environment heterogeneity in the context of a full spectrum of human breast cancers.
Collapse
|
391
|
Runa F, Hamalian S, Meade K, Shisgal P, Gray PC, Kelber JA. Tumor microenvironment heterogeneity: challenges and opportunities. ACTA ACUST UNITED AC 2017; 3:218-229. [PMID: 29430386 DOI: 10.1007/s40610-017-0073-7] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The tumor microenvironment (TME) has been recognized as an integral component of malignancies in breast and prostate tissues, contributing in confounding ways to tumor progression, metastasis, therapy resistance and disease recurrence. Major components of the TME are immune cells, fibroblasts, pericytes, endothelial cells, mesenchymal stroma/stem cells (MSCs), and extracellular matrix (ECM) components. Herein, we discuss the molecular and cellular heterogeneity within the TME and how this presents unique challenges and opportunities for treating breast and prostate cancers.
Collapse
Affiliation(s)
- F Runa
- Department of Biology, California State University, Northridge, CA
| | - S Hamalian
- Department of Biology, California State University, Northridge, CA
| | - K Meade
- Department of Biology, California State University, Northridge, CA
| | - P Shisgal
- Department of Biology, California State University, Northridge, CA
| | - P C Gray
- The Salk Institute for Biological Studies, La Jolla, CA
| | - J A Kelber
- Department of Biology, California State University, Northridge, CA
| |
Collapse
|
392
|
Ngan E, Kiepas A, Brown CM, Siegel PM. Emerging roles for LPP in metastatic cancer progression. J Cell Commun Signal 2017; 12:143-156. [PMID: 29027626 DOI: 10.1007/s12079-017-0415-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 10/03/2017] [Indexed: 01/21/2023] Open
Abstract
LIM domain containing proteins are important regulators of diverse cellular processes, and play pivotal roles in regulating the actin cytoskeleton. Lipoma Preferred Partner (LPP) is a member of the zyxin family of LIM proteins that has long been characterized as a promoter of mesenchymal/fibroblast cell migration. More recently, LPP has emerged as a critical inducer of tumor cell migration, invasion and metastasis. LPP is thought to contribute to these malignant phenotypes by virtue of its ability to shuttle into the nucleus, localize to adhesions and, most recently, to promote invadopodia formation. In this review, we will examine the mechanisms through which LPP regulates the functions of adhesions and invadopodia, and discuss potential roles of LPP in mediating cellular responses to mechanical cues within these mechanosensory structures.
Collapse
Affiliation(s)
- Elaine Ngan
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue West, Room 508, Montréal, Québec, H3A 1A3, Canada.,Department of Medicine, McGill University, Montréal, Québec, Canada
| | - Alex Kiepas
- Department of Physiology, McGill University, Montréal, Québec, Canada
| | - Claire M Brown
- Department of Physiology, McGill University, Montréal, Québec, Canada
| | - Peter M Siegel
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue West, Room 508, Montréal, Québec, H3A 1A3, Canada. .,Department of Medicine, McGill University, Montréal, Québec, Canada.
| |
Collapse
|
393
|
Wong AD, Searson PC. Mitosis-Mediated Intravasation in a Tissue-Engineered Tumor-Microvessel Platform. Cancer Res 2017; 77:6453-6461. [PMID: 28923855 DOI: 10.1158/0008-5472.can-16-3279] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 07/09/2017] [Accepted: 09/07/2017] [Indexed: 12/20/2022]
Abstract
Intravasation involves the migration of tumor cells across the local endothelium and escape into vessel flow. Although tumor cell invasiveness has been correlated to increased intravasation, the details of transendothelial migration and detachment into circulation are still unclear. Here, we analyzed the intravasation of invasive human breast cancer cells within a tissue-engineered microvessel model of the tumor microenvironment. Using live-cell fluorescence microscopy, we captured 2,330 hours of tumor cell interactions with functional microvessels and provide evidence for a mitosis-mediated mechanism where tumor cells located along the vessel periphery are able to disrupt the vessel endothelium through cell division and detach into circulation. This model provides a framework for understanding the physical and biological parameters of the tumor microenvironment that mediate intravasation of tumor cells across an intact endothelium. Cancer Res; 77(22); 6453-61. ©2017 AACR.
Collapse
Affiliation(s)
- Andrew D Wong
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, Maryland.,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Peter C Searson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, Maryland. .,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
394
|
Song W, Mazzieri R, Yang T, Gobe GC. Translational Significance for Tumor Metastasis of Tumor-Associated Macrophages and Epithelial-Mesenchymal Transition. Front Immunol 2017; 8:1106. [PMID: 28955335 PMCID: PMC5601389 DOI: 10.3389/fimmu.2017.01106] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 08/23/2017] [Indexed: 12/18/2022] Open
Abstract
The tumor microenvironment determines development and progression of many cancers. Epithelial–mesenchymal transition (EMT) is fundamental to tumor progression and metastasis not only by increasing invasiveness but also by increasing resistance to cell death, senescence, and various cancer therapies; determining inflammation and immune surveillance; and conferring stem cell properties. It does this by enabling polarized epithelial cells to transform into cells with a mesenchymal, and therefore motile, phenotype. Tumor-associated macrophages (TAMs) are key cells of the tumor microenvironment that orchestrate the connection between inflammation and cancer. Activation of EMT often requires crosstalk between cancer cells and components of the local tumor microenvironment, including TAMs. In this review, clinical and experimental evidence is presented for control of TAMs in promoting cancer cell invasion and migration and their interaction with the EMT process in the metastatic cascade. The translational significance of these findings is that the signaling pathways that interconnect TAMs and EMT-modified cancer cells may represent promising therapeutic targets for the treatment of tumor metastasis.
Collapse
Affiliation(s)
- Wenzhe Song
- Faculty of Medicine, Translational Research Institute, The University of Queensland Diamantina Institute, Brisbane, QLD, Australia.,Department of General Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Roberta Mazzieri
- Faculty of Medicine, Translational Research Institute, The University of Queensland Diamantina Institute, Brisbane, QLD, Australia
| | - Tao Yang
- Discipline of Pathology, The Western Sydney University, Sydney, NSW, Australia.,SydPath, St Vincent's Hospital, Sydney, NSW, Australia
| | - Glenda C Gobe
- Faculty of Medicine, Translational Research Institute, The University of Queensland Diamantina Institute, Brisbane, QLD, Australia
| |
Collapse
|
395
|
Entenberg D, Pastoriza JM, Oktay MH, Voiculescu S, Wang Y, Sosa MS, Aguirre-Ghiso J, Condeelis J. Time-lapsed, large-volume, high-resolution intravital imaging for tissue-wide analysis of single cell dynamics. Methods 2017; 128:65-77. [PMID: 28911733 PMCID: PMC5659295 DOI: 10.1016/j.ymeth.2017.07.019] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 06/15/2017] [Accepted: 07/20/2017] [Indexed: 01/06/2023] Open
Abstract
Pathologists rely on microscopy to diagnose disease states in tissues and organs. They utilize both high-resolution, high-magnification images to interpret the staining and morphology of individual cells, as well as low-magnification overviews to give context and location to these cells. Intravital imaging is a powerful technique for studying cells and tissues in their native, live environment and can yield sub-cellular resolution images similar to those used by pathologists. However, technical limitations prevent the straightforward acquisition of low-magnification images during intravital imaging, and they are hence not typically captured. The serial acquisition, mosaicking, and stitching together of many high-resolution, high-magnification fields of view is a technique that overcomes these limitations in fixed and ex vivo tissues. The technique however, has not to date been widely applied to intravital imaging as movements caused by the living animal induce image distortions that are difficult to compensate for computationally. To address this, we have developed techniques for the stabilization of numerous tissues, including extremely compliant tissues, that have traditionally been extremely difficult to image. We present a novel combination of these stabilization techniques with mosaicked and stitched intravital imaging, resulting in a process we call Large-Volume High-Resolution Intravital Imaging (LVHR-IVI). The techniques we present are validated and make large volume intravital imaging accessible to any lab with a multiphoton microscope.
Collapse
Affiliation(s)
- David Entenberg
- Anatomy and Structural Biology, Integrated Imaging Program, Gruss-Lipper Biophotonics Center, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States.
| | - Jessica M Pastoriza
- Department of Surgery, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
| | - Maja H Oktay
- Anatomy and Structural Biology, Integrated Imaging Program, Gruss-Lipper Biophotonics Center, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States; Department of Pathology, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
| | - Sonia Voiculescu
- Department of Surgery, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
| | - Yarong Wang
- Anatomy and Structural Biology, Integrated Imaging Program, Gruss-Lipper Biophotonics Center, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
| | - Maria Soledad Sosa
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Julio Aguirre-Ghiso
- Division of Hematology and Oncology, Department of Medicine, Department of Otolaryngology, Department of Oncological Sciences, Tisch Cancer Institute, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - John Condeelis
- Anatomy and Structural Biology, Integrated Imaging Program, Gruss-Lipper Biophotonics Center, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
| |
Collapse
|
396
|
Gómez-Cuadrado L, Tracey N, Ma R, Qian B, Brunton VG. Mouse models of metastasis: progress and prospects. Dis Model Mech 2017; 10:1061-1074. [PMID: 28883015 PMCID: PMC5611969 DOI: 10.1242/dmm.030403] [Citation(s) in RCA: 211] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Metastasis is the spread of cancer cells from a primary tumor to distant sites within the body to establish secondary tumors. Although this is an inefficient process, the consequences are devastating as metastatic disease accounts for >90% of cancer-related deaths. The formation of metastases is the result of a series of events that allow cancer cells to escape from the primary site, survive in the lymphatic system or blood vessels, extravasate and grow at distant sites. The metastatic capacity of a tumor is determined by genetic and epigenetic changes within the cancer cells as well as contributions from cells in the tumor microenvironment. Mouse models have proven to be an important tool for unraveling the complex interactions involved in the metastatic cascade and delineating its many stages. Here, we critically appraise the strengths and weaknesses of the current mouse models and highlight the recent advances that have been made using these models in our understanding of metastasis. We also discuss the use of these models for testing potential therapies and the challenges associated with the translation of these findings into the provision of new and effective treatments for cancer patients.
Collapse
Affiliation(s)
- Laura Gómez-Cuadrado
- Edinburgh Cancer Research Centre, Institute for Genetics and Molecular Medicine, Edinburgh, EH4 2XR, UK
| | - Natasha Tracey
- Edinburgh Cancer Research Centre, Institute for Genetics and Molecular Medicine, Edinburgh, EH4 2XR, UK
| | - Ruoyu Ma
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Binzhi Qian
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Edinburgh Cancer Research UK Centre, Queen's Medical Research Institute, Edinburgh, EH16 4TJ, UK
| | - Valerie G Brunton
- Edinburgh Cancer Research Centre, Institute for Genetics and Molecular Medicine, Edinburgh, EH4 2XR, UK
| |
Collapse
|
397
|
Harney AS, Karagiannis GS, Pignatelli J, Smith BD, Kadioglu E, Wise SC, Hood MM, Kaufman MD, Leary CB, Lu WP, Al-Ani G, Chen X, Entenberg D, Oktay MH, Wang Y, Chun L, De Palma M, Jones JG, Flynn DL, Condeelis JS. The Selective Tie2 Inhibitor Rebastinib Blocks Recruitment and Function of Tie2 Hi Macrophages in Breast Cancer and Pancreatic Neuroendocrine Tumors. Mol Cancer Ther 2017; 16:2486-2501. [PMID: 28838996 DOI: 10.1158/1535-7163.mct-17-0241] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 07/12/2017] [Accepted: 08/10/2017] [Indexed: 01/22/2023]
Abstract
Tumor-infiltrating myeloid cells promote tumor progression by mediating angiogenesis, tumor cell intravasation, and metastasis, which can offset the effects of chemotherapy, radiation, and antiangiogenic therapy. Here, we show that the kinase switch control inhibitor rebastinib inhibits Tie2, a tyrosine kinase receptor expressed on endothelial cells and protumoral Tie2-expressing macrophages in mouse models of metastatic cancer. Rebastinib reduces tumor growth and metastasis in an orthotopic mouse model of metastatic mammary carcinoma through reduction of Tie2+ myeloid cell infiltration, antiangiogenic effects, and blockade of tumor cell intravasation mediated by perivascular Tie2Hi/Vegf-AHi macrophages in the tumor microenvironment of metastasis (TMEM). The antitumor effects of rebastinib enhance the efficacy of microtubule inhibiting chemotherapeutic agents, either eribulin or paclitaxel, by reducing tumor volume, metastasis, and improving overall survival. Rebastinib inhibition of angiopoietin/Tie2 signaling impairs multiple pathways in tumor progression mediated by protumoral Tie2+ macrophages, including TMEM-dependent dissemination and angiopoietin/Tie2-dependent angiogenesis. Rebastinib is a promising therapy for achieving Tie2 inhibition in cancer patients. Mol Cancer Ther; 16(11); 2486-501. ©2017 AACR.
Collapse
Affiliation(s)
- Allison S Harney
- Department of Anatomy & Structural Biology, Albert Einstein College of Medicine, New York, New York.,Department of Radiology, Albert Einstein College of Medicine, New York, New York.,Integrated Imaging Program, Albert Einstein College of Medicine, New York, New York.,Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, New York, New York
| | - George S Karagiannis
- Department of Anatomy & Structural Biology, Albert Einstein College of Medicine, New York, New York.,Integrated Imaging Program, Albert Einstein College of Medicine, New York, New York.,Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, New York, New York
| | - Jeanine Pignatelli
- Department of Anatomy & Structural Biology, Albert Einstein College of Medicine, New York, New York.,Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, New York, New York
| | - Bryan D Smith
- Deciphera Pharmaceuticals, LLC, Waltham, Massachusetts
| | - Ece Kadioglu
- ISREC, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Scott C Wise
- Deciphera Pharmaceuticals, LLC, Waltham, Massachusetts
| | - Molly M Hood
- Deciphera Pharmaceuticals, LLC, Waltham, Massachusetts
| | | | | | - Wei-Ping Lu
- Deciphera Pharmaceuticals, LLC, Waltham, Massachusetts
| | - Gada Al-Ani
- Deciphera Pharmaceuticals, LLC, Waltham, Massachusetts
| | - Xiaoming Chen
- Department of Anatomy & Structural Biology, Albert Einstein College of Medicine, New York, New York.,Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, New York, New York
| | - David Entenberg
- Department of Anatomy & Structural Biology, Albert Einstein College of Medicine, New York, New York.,Integrated Imaging Program, Albert Einstein College of Medicine, New York, New York.,Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, New York, New York
| | - Maja H Oktay
- Department of Anatomy & Structural Biology, Albert Einstein College of Medicine, New York, New York.,Integrated Imaging Program, Albert Einstein College of Medicine, New York, New York.,Department of Pathology Albert Einstein College of Medicine, New York, New York
| | - Yarong Wang
- Department of Anatomy & Structural Biology, Albert Einstein College of Medicine, New York, New York.,Integrated Imaging Program, Albert Einstein College of Medicine, New York, New York.,Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, New York, New York
| | | | - Michele De Palma
- ISREC, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Joan G Jones
- Department of Anatomy & Structural Biology, Albert Einstein College of Medicine, New York, New York.,Integrated Imaging Program, Albert Einstein College of Medicine, New York, New York.,Department of Pathology Albert Einstein College of Medicine, New York, New York.,Department of Epidemiology & Population Health, Albert Einstein College of Medicine, New York, New York
| | | | - John S Condeelis
- Department of Anatomy & Structural Biology, Albert Einstein College of Medicine, New York, New York. .,Integrated Imaging Program, Albert Einstein College of Medicine, New York, New York.,Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, New York, New York
| |
Collapse
|
398
|
Chang YS, Jalgaonkar SP, Middleton JD, Hai T. Stress-inducible gene Atf3 in the noncancer host cells contributes to chemotherapy-exacerbated breast cancer metastasis. Proc Natl Acad Sci U S A 2017; 114:E7159-E7168. [PMID: 28784776 PMCID: PMC5576783 DOI: 10.1073/pnas.1700455114] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Chemotherapy is a double-edged sword. It is anticancer because of its cytotoxicity. Paradoxically, by increasing chemoresistance and cancer metastasis, it is also procancer. However, the underlying mechanisms for chemotherapy-induced procancer activities are not well understood. Here we describe the ability of paclitaxel (PTX), a frontline chemotherapeutic agent, to exacerbate metastasis in mouse models of breast cancer. We demonstrate that, despite the apparent benefit of reducing tumor size, PTX increased the circulating tumor cells in the blood and enhanced the metastatic burden at the lung. At the primary tumor, PTX increased the abundance of the tumor microenvironment of metastasis, a landmark microanatomical structure at the microvasculature where cancer cells enter the blood stream. At the metastatic lung, PTX improved the tissue microenvironment (the "soil") for cancer cells (the "seeds") to thrive; these changes include increased inflammatory monocytes and reduced cytotoxicity. Importantly, these changes in the primary tumor and the metastatic lung were all dependent on Atf3, a stress-inducible gene, in the noncancer host cells. Together, our data provide mechanistic insights into the procancer effect of chemotherapy, explaining its paradox in the context of the seed-and-soil theory. Analyses of public datasets suggest that our data may have relevance to human cancers. Thus, ATF3 in the host cells links a chemotherapeutic agent-a stressor-to immune modulation and cancer metastasis. Dampening the effect of ATF3 may improve the efficacy of chemotherapy.
Collapse
Affiliation(s)
- Yi Seok Chang
- Molecular, Cellular, and Developmental Biology Program, Ohio State University, Columbus, OH 43210
- Department of Biological Chemistry and Pharmacology, Ohio State University, Columbus, OH 43210
| | - Swati P Jalgaonkar
- Molecular, Cellular, and Developmental Biology Program, Ohio State University, Columbus, OH 43210
- Department of Biological Chemistry and Pharmacology, Ohio State University, Columbus, OH 43210
| | - Justin D Middleton
- Molecular, Cellular, and Developmental Biology Program, Ohio State University, Columbus, OH 43210
- Department of Biological Chemistry and Pharmacology, Ohio State University, Columbus, OH 43210
| | - Tsonwin Hai
- Molecular, Cellular, and Developmental Biology Program, Ohio State University, Columbus, OH 43210;
- Department of Biological Chemistry and Pharmacology, Ohio State University, Columbus, OH 43210
| |
Collapse
|
399
|
Abstract
Tumours display considerable variation in the patterning and properties of angiogenic blood vessels, as well as in their responses to anti-angiogenic therapy. Angiogenic programming of neoplastic tissue is a multidimensional process regulated by cancer cells in concert with a variety of tumour-associated stromal cells and their bioactive products, which encompass cytokines and growth factors, the extracellular matrix and secreted microvesicles. In this Review, we discuss the extrinsic regulation of angiogenesis by the tumour microenvironment, highlighting potential vulnerabilities that could be targeted to improve the applicability and reach of anti-angiogenic cancer therapies.
Collapse
Affiliation(s)
- Michele De Palma
- The Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Daniela Biziato
- The Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Tatiana V Petrova
- Department of Fundamental Oncology, Ludwig Institute for Cancer Research and Division of Experimental Pathology, University of Lausanne and University of Lausanne Hospital, 1066 Lausanne, Switzerland
| |
Collapse
|
400
|
Viski C, König C, Kijewska M, Mogler C, Isacke CM, Augustin HG. Endosialin-Expressing Pericytes Promote Metastatic Dissemination. Cancer Res 2017; 76:5313-25. [PMID: 27635044 DOI: 10.1158/0008-5472.can-16-0932] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 07/18/2016] [Indexed: 11/16/2022]
Abstract
Metastasis is a multistep process that is critically dependent on the interaction of metastasizing tumor cells with cells in the local microenvironment. Within this tumor stroma, vessel-associated pericytes and myofibroblasts share a number of traits, including the upregulated expression of the transmembrane receptor endosialin (CD248). Comparative experiments in wild-type and endosialin-deficient mice revealed that stromal endosialin does not affect primary tumor growth but strongly promotes spontaneous metastasis. Mechanistically, endosialin-expressing pericytes in the primary tumor facilitate distant site metastasis by promoting tumor cell intravasation in a cell contact-dependent manner, resulting in elevated numbers of circulating tumor cells. Corresponding to these preclinical experiments, in independent cohorts of primary human breast cancers, upregulated endosialin expression significantly correlates with increased metastasis and poorer patient survival. Together, the data demonstrate a critical role for endosialin-expressing primary tumor pericytes in mediating metastatic dissemination and identify endosialin as a promising therapeutic target in breast cancer. Cancer Res; 76(18); 5313-25. ©2016 AACR.
Collapse
Affiliation(s)
- Carmen Viski
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Courtney König
- Division of Vascular Oncology and Metastasis (DKFZ-ZMBH Alliance), German Cancer Research Center, Heidelberg, Germany. Department of Vascular Biology and Tumor Angiogenesis (CBTM), Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Magdalena Kijewska
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Carolin Mogler
- Division of Vascular Oncology and Metastasis (DKFZ-ZMBH Alliance), German Cancer Research Center, Heidelberg, Germany. Institute of Pathology, Heidelberg University, Heidelberg, Germany
| | - Clare M Isacke
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom.
| | - Hellmut G Augustin
- Division of Vascular Oncology and Metastasis (DKFZ-ZMBH Alliance), German Cancer Research Center, Heidelberg, Germany. Department of Vascular Biology and Tumor Angiogenesis (CBTM), Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany. German Cancer Consortium, Heidelberg, Germany.
| |
Collapse
|