401
|
Miller MA, Arlauckas S, Weissleder R. Prediction of Anti-cancer Nanotherapy Efficacy by Imaging. Nanotheranostics 2017; 1:296-312. [PMID: 29071194 PMCID: PMC5646731 DOI: 10.7150/ntno.20564] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 05/27/2017] [Indexed: 12/17/2022] Open
Abstract
Anticancer nanotherapeutics have shown mixed results in clinical trials, raising the questions of whether imaging should be used to i) identify patients with a higher likelihood of nanoparticle accumulation, ii) assess nanotherapeutic efficacy before traditional measures show response, and iii) guide adjuvant treatments to enhance therapeutic nanoparticle (TNP) delivery. Here we review the use of a clinically approved MRI nanoparticle (ferumoxytol, FMX) to predict TNP delivery and efficacy. It is becoming increasingly apparent that nanoparticles used for imaging, despite clearly distinct physicochemical properties, often co-localize with TNP in tumors. This evidence offers the possibility of using FMX as a generic “companion diagnostic” nanoparticle for multiple TNP formulations, thus potentially allowing many of the complex regulatory and cost challenges of other approaches to be avoided.
Collapse
Affiliation(s)
- Miles A Miller
- Center for Systems Biology, Massachusetts General Hospital, USA.,Department of Radiology, Massachusetts General Hospital, USA
| | - Sean Arlauckas
- Center for Systems Biology, Massachusetts General Hospital, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, USA.,Department of Radiology, Massachusetts General Hospital, USA.,Department of Systems Biology, Harvard Medical School, USA
| |
Collapse
|
402
|
Karagiannis GS, Pastoriza JM, Wang Y, Harney AS, Entenberg D, Pignatelli J, Sharma VP, Xue EA, Cheng E, D'Alfonso TM, Jones JG, Anampa J, Rohan TE, Sparano JA, Condeelis JS, Oktay MH. Neoadjuvant chemotherapy induces breast cancer metastasis through a TMEM-mediated mechanism. Sci Transl Med 2017; 9:eaan0026. [PMID: 28679654 PMCID: PMC5592784 DOI: 10.1126/scitranslmed.aan0026] [Citation(s) in RCA: 359] [Impact Index Per Article: 44.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 06/13/2017] [Indexed: 12/11/2022]
Abstract
Breast cancer cells disseminate through TIE2/MENACalc/MENAINV-dependent cancer cell intravasation sites, called tumor microenvironment of metastasis (TMEM), which are clinically validated as prognostic markers of metastasis in breast cancer patients. Using fixed tissue and intravital imaging of a PyMT murine model and patient-derived xenografts, we show that chemotherapy increases the density and activity of TMEM sites and Mena expression and promotes distant metastasis. Moreover, in the residual breast cancers of patients treated with neoadjuvant paclitaxel after doxorubicin plus cyclophosphamide, TMEM score and its mechanistically connected MENAINV isoform expression pattern were both increased, suggesting that chemotherapy, despite decreasing tumor size, increases the risk of metastatic dissemination. Chemotherapy-induced TMEM activity and cancer cell dissemination were reversed by either administration of the TIE2 inhibitor rebastinib or knockdown of the MENA gene. Our results indicate that TMEM score increases and MENA isoform expression pattern changes with chemotherapy and can be used in predicting prometastatic changes in response to chemotherapy. Furthermore, inhibitors of TMEM function may improve clinical benefits of chemotherapy in the neoadjuvant setting or in metastatic disease.
Collapse
Affiliation(s)
- George S Karagiannis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jessica M Pastoriza
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10467, USA
| | - Yarong Wang
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Allison S Harney
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Radiology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - David Entenberg
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jeanine Pignatelli
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ved P Sharma
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Emily A Xue
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Esther Cheng
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Timothy M D'Alfonso
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Joan G Jones
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Pathology, Montefiore Medical Center, Bronx, NY 10467, USA
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jesus Anampa
- Department of Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10467, USA
| | - Thomas E Rohan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Joseph A Sparano
- Department of Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10467, USA
| | - John S Condeelis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Maja H Oktay
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Pathology, Montefiore Medical Center, Bronx, NY 10467, USA
| |
Collapse
|
403
|
Hirata E, Sahai E. Tumor Microenvironment and Differential Responses to Therapy. Cold Spring Harb Perspect Med 2017; 7:cshperspect.a026781. [PMID: 28213438 DOI: 10.1101/cshperspect.a026781] [Citation(s) in RCA: 283] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cancer evolution plays a key role in both the development of tumors and their response to therapy. Like all evolutionary processes, tumor evolution is shaped by the environment. In tumors, this consists of a complex mixture of nontransformed cell types and extracellular matrix. Chemotherapy or radiotherapy imposes further strong selective pressures on cancer cells during cancer treatment. Here, we review how different components of the tumor microenvironment can modulate the response to chemo- and radiotherapy. We further describe how therapeutic strategies directly alter the composition, or function, of the tumor microenvironment, thereby further altering the selective pressures to which cancer cells are exposed. Last, we explore the consequences of these interactions for therapy outcomes and how to exploit our increasing understanding of the tumor microenvironment for therapeutic benefit.
Collapse
Affiliation(s)
- Eishu Hirata
- Department of Oncologic Pathology, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Erik Sahai
- Tumor Cell Biology Laboratory, Francis Crick Institute, London WC2A 3LY, United Kingdom
| |
Collapse
|
404
|
Schmittnaegel M, Rigamonti N, Kadioglu E, Cassará A, Wyser Rmili C, Kiialainen A, Kienast Y, Mueller HJ, Ooi CH, Laoui D, De Palma M. Dual angiopoietin-2 and VEGFA inhibition elicits antitumor immunity that is enhanced by PD-1 checkpoint blockade. Sci Transl Med 2017; 9:9/385/eaak9670. [PMID: 28404865 DOI: 10.1126/scitranslmed.aak9670] [Citation(s) in RCA: 426] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 03/17/2017] [Indexed: 12/14/2022]
Abstract
Pathological angiogenesis is a hallmark of cancer and a therapeutic target. Vascular endothelial growth factor A (VEGFA) and angiopoietin-2 (ANGPT2; also known as ANG2) are proangiogenic cytokines that sustain tumor angiogenesis and limit antitumor immunity. We show that combined ANGPT2 and VEGFA blockade by a bispecific antibody (A2V) provided superior therapeutic benefits, as compared to the single agents, in both genetically engineered and transplant tumor models, including metastatic breast cancer (MMTV-PyMT), pancreatic neuroendocrine tumor (RIP1-Tag2), and melanoma. Mechanistically, A2V promoted vascular regression, tumor necrosis, and antigen presentation by intratumoral phagocytes. A2V also normalized the remaining blood vessels and facilitated the extravasation and perivascular accumulation of activated, interferon-γ (IFNγ)-expressing CD8+ cytotoxic T lymphocytes (CTLs). Whereas the antitumoral activity of A2V was, at least partly, CTL-dependent, perivascular T cells concurrently up-regulated the expression of the immune checkpoint ligand programmed cell death ligand 1 (PD-L1) in tumor endothelial cells. IFNγ neutralization blunted this adaptive response, and PD-1 blockade improved tumor control by A2V in different cancer models. These findings position immune cells as key effectors of antiangiogenic therapy and support the rationale for cotargeting angiogenesis and immune checkpoints in cancer therapy.
Collapse
Affiliation(s)
- Martina Schmittnaegel
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Nicolò Rigamonti
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Ece Kadioglu
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Antonino Cassará
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Céline Wyser Rmili
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Anna Kiialainen
- Roche Innovation Center Basel, Pharmaceutical Sciences, Pharma Research and Early Development, 4070 Basel, Switzerland
| | - Yvonne Kienast
- Roche Innovation Center Munich, Oncology Discovery, Pharma Research and Early Development, 82377 Penzberg, Germany
| | - Hans-Joachim Mueller
- Roche Innovation Center Munich, Oncology Discovery, Pharma Research and Early Development, 82377 Penzberg, Germany
| | - Chia-Huey Ooi
- Roche Innovation Center Basel, Pharmaceutical Sciences, Pharma Research and Early Development, 4070 Basel, Switzerland.,Roche Innovation Center Munich, Oncology Discovery, Pharma Research and Early Development, 82377 Penzberg, Germany
| | - Damya Laoui
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Michele De Palma
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland.
| |
Collapse
|
405
|
Abstract
Imaging is widely used in anticancer drug development, typically for whole-body tracking of labelled drugs to different organs or to assess drug efficacy through volumetric measurements. However, increasing attention has been drawn to pharmacology at the single-cell level. Diverse cell types, including cancer-associated immune cells, physicochemical features of the tumour microenvironment and heterogeneous cell behaviour all affect drug delivery, response and resistance. This Review summarizes developments in the imaging of in vivo anticancer drug action, with a focus on microscopy approaches at the single-cell level and translational lessons for the clinic.
Collapse
Affiliation(s)
- Miles A. Miller
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA
- Department of Systems Biology, Harvard Medical School, Boston, MA
| |
Collapse
|
406
|
Mesenchymal-Epithelial Transition and Circulating Tumor Cells in Small Cell Lung Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 994:229-245. [DOI: 10.1007/978-3-319-55947-6_12] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
407
|
Saharinen P, Eklund L, Alitalo K. Therapeutic targeting of the angiopoietin-TIE pathway. Nat Rev Drug Discov 2017; 16:635-661. [PMID: 28529319 DOI: 10.1038/nrd.2016.278] [Citation(s) in RCA: 400] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The endothelial angiopoietin (ANG)-TIE growth factor receptor pathway regulates vascular permeability and pathological vascular remodelling during inflammation, tumour angiogenesis and metastasis. Drugs that target the ANG-TIE pathway are in clinical development for oncological and ophthalmological applications. The aim is to complement current vascular endothelial growth factor (VEGF)-based anti-angiogenic therapies in cancer, wet age-related macular degeneration and macular oedema. The unique function of the ANG-TIE pathway in vascular stabilization also renders this pathway an attractive target in sepsis, organ transplantation, atherosclerosis and vascular complications of diabetes. This Review covers key aspects of the function of the ANG-TIE pathway in vascular disease and describes the recent development of novel therapeutics that target this pathway.
Collapse
Affiliation(s)
- Pipsa Saharinen
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, Haartmaninkatu 8, P.O. Box 63, FI-00014 Helsinki, Finland
| | - Lauri Eklund
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, Aapistie 5A, University of Oulu, 90220 Oulu, Finland
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, Haartmaninkatu 8, P.O. Box 63, FI-00014 Helsinki, Finland
| |
Collapse
|
408
|
Deryugina EI, Kiosses WB. Intratumoral Cancer Cell Intravasation Can Occur Independent of Invasion into the Adjacent Stroma. Cell Rep 2017; 19:601-616. [PMID: 28423322 DOI: 10.1016/j.celrep.2017.03.064] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 01/29/2017] [Accepted: 03/21/2017] [Indexed: 02/07/2023] Open
Abstract
Intravasation, active entry of cancer cells into the circulation, is often considered to be a relatively late event in tumor development occurring after stromal invasion. Here, we provide evidence that intravasation can be initiated early during tumor development and proceed in parallel to or independent of tumor invasion into surrounding stroma. By applying direct and unbiased intravasation-scoring methods to two histologically distinct human cancer types in live-animal models, we demonstrate that intravasation takes place almost exclusively within the tumor core, involves intratumoral vasculature, and does not involve vasculotropic cancer cells invading tumor-adjacent stroma and migrating along tumor-converging blood vessels. Highlighting an additional role for EGFR in cancer, we find that EGFR is required for the development of an intravasation-sustaining intratumoral vasculature. Intratumoral localization of intravasation supports the notion that overt metastases in cancer patients could be initiated much earlier during cancer progression than appreciated within conventional clinical tumor staging systems.
Collapse
Affiliation(s)
- Elena I Deryugina
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | - William B Kiosses
- Confocal Microscopy Core Facility, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|
409
|
Suijkerbuijk SJE, van Rheenen J. From good to bad: Intravital imaging of the hijack of physiological processes by cancer cells. Dev Biol 2017; 428:328-337. [PMID: 28473106 DOI: 10.1016/j.ydbio.2017.04.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 04/21/2017] [Accepted: 04/23/2017] [Indexed: 12/23/2022]
Abstract
Homeostasis of tissues is tightly regulated at the cellular, tissue and organismal level. Interestingly, tumor cells have found ways to hijack many of these physiological processes at all the different levels. Here we review how intravital microscopy techniques have provided new insights into our understanding of tissue homeostasis and cancer progression. In addition, we highlight the different strategies that tumor cells have adopted to use these physiological processes for their own benefit. We describe how visualization of these dynamic processes in living mice has broadened to our view on cancer initiation and progression.
Collapse
Affiliation(s)
- Saskia J E Suijkerbuijk
- Hubrecht Institute - KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands; Cancer Genomics Netherlands, 3584 CG Utrecht, The Netherlands
| | - Jacco van Rheenen
- Hubrecht Institute - KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands; Cancer Genomics Netherlands, 3584 CG Utrecht, The Netherlands.
| |
Collapse
|
410
|
Oudin MJ, Weaver VM. Physical and Chemical Gradients in the Tumor Microenvironment Regulate Tumor Cell Invasion, Migration, and Metastasis. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2017; 81:189-205. [PMID: 28424337 DOI: 10.1101/sqb.2016.81.030817] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cancer metastasis requires the invasion of tumor cells into the stroma and the directed migration of tumor cells through the stroma toward the vasculature and lymphatics where they can disseminate and colonize secondary organs. Physical and biochemical gradients that form within the primary tumor tissue promote tumor cell invasion and drive persistent migration toward blood vessels and the lymphatics to facilitate tumor cell dissemination. These microenvironment cues include hypoxia and pH gradients, gradients of soluble cues that induce chemotaxis, and ions that facilitate galvanotaxis, as well as modifications to the concentration, organization, and stiffness of the extracellular matrix that produce haptotactic, alignotactic, and durotactic gradients. These gradients form through dynamic interactions between the tumor cells and the resident fibroblasts, adipocytes, nerves, endothelial cells, infiltrating immune cells, and mesenchymal stem cells. Malignant progression results from the integrated response of the tumor to these extrinsic physical and chemical cues. Here, we first describe how these physical and chemical gradients develop, and we discuss their role in tumor progression. We then review assays to study these gradients. We conclude with a discussion of clinical strategies used to detect and inhibit these gradients in tumors and of new intervention opportunities. Clarifying the role of these gradients in tumor evolution offers a unique approach to target metastasis.
Collapse
Affiliation(s)
- Madeleine J Oudin
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Valerie M Weaver
- Department of Surgery, Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, California 94143
- UCSF Comprehensive Cancer Center, Helen Diller Family Cancer Research Center, University of California, San Francisco, San Francisco, California 94143
- Department of Anatomy, Department of Bioengineering and Therapeutic Sciences, and Department of Radiation Oncology, University of California San Francisco, San Francisco, California 94143
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research and The Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California 94143
| |
Collapse
|
411
|
Tumor Cell Invadopodia: Invasive Protrusions that Orchestrate Metastasis. Trends Cell Biol 2017; 27:595-607. [PMID: 28412099 DOI: 10.1016/j.tcb.2017.03.003] [Citation(s) in RCA: 267] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/09/2017] [Accepted: 03/13/2017] [Indexed: 12/26/2022]
Abstract
Invadopodia are a subset of invadosomes that are implicated in the integration of signals from the tumor microenvironment to support tumor cell invasion and dissemination. Recent progress has begun to define how tumor cells regulate the plasticity necessary for invadopodia to assemble and function efficiently in the different microenvironments encountered during dissemination in vivo. Exquisite mapping by many laboratories of the pathways involved in integrating diverse invadopodium initiation signals, from growth factors, to extracellular matrix (ECM) and cell-cell contact in the tumor microenvironment, has led to insight into the molecular basis of this plasticity. Here, we integrate this new information to discuss how the invadopodium is an important conductor that orchestrates tumor cell dissemination during metastasis.
Collapse
|
412
|
Holen I, Speirs V, Morrissey B, Blyth K. In vivo models in breast cancer research: progress, challenges and future directions. Dis Model Mech 2017; 10:359-371. [PMID: 28381598 PMCID: PMC5399571 DOI: 10.1242/dmm.028274] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Research using animal model systems has been instrumental in delivering improved therapies for breast cancer, as well as in generating new insights into the mechanisms that underpin development of the disease. A large number of different models are now available, reflecting different types and stages of the disease; choosing which one to use depends on the specific research question(s) to be investigated. Based on presentations and discussions from leading experts who attended a recent workshop focused on in vivo models of breast cancer, this article provides a perspective on the many varied uses of these models in breast cancer research, their strengths, associated challenges and future directions. Among the questions discussed were: how well do models represent the different stages of human disease; how can we model the involvement of the human immune system and microenvironment in breast cancer; what are the appropriate models of metastatic disease; can we use models to carry out preclinical drug trials and identify pathways responsible for drug resistance; and what are the limitations of patient-derived xenograft models? We briefly outline the areas where the existing breast cancer models require improvement in light of the increased understanding of the disease process, reflecting the drive towards more personalised therapies and identification of mechanisms of drug resistance.
Collapse
Affiliation(s)
- Ingunn Holen
- Academic Unit of Clinical Oncology, University of Sheffield, Sheffield S10 2RX, UK
| | - Valerie Speirs
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds LS9 7TF, UK
| | - Bethny Morrissey
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds LS9 7TF, UK
| | - Karen Blyth
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK
| |
Collapse
|
413
|
Miller MA, Weissleder R. Imaging the pharmacology of nanomaterials by intravital microscopy: Toward understanding their biological behavior. Adv Drug Deliv Rev 2017; 113:61-86. [PMID: 27266447 PMCID: PMC5136524 DOI: 10.1016/j.addr.2016.05.023] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 05/25/2016] [Indexed: 12/15/2022]
Abstract
Therapeutic nanoparticles (NPs) can deliver cytotoxic chemotherapeutics and other drugs more safely and efficiently to patients; furthermore, selective delivery to target tissues can theoretically be accomplished actively through coating NPs with molecular ligands, and passively through exploiting physiological "enhanced permeability and retention" features. However, clinical trial results have been mixed in showing improved efficacy with drug nanoencapsulation, largely due to heterogeneous NP accumulation at target sites across patients. Thus, a clear need exists to better understand why many NP strategies fail in vivo and not result in significantly improved tumor uptake or therapeutic response. Multicolor in vivo confocal fluorescence imaging (intravital microscopy; IVM) enables integrated pharmacokinetic and pharmacodynamic (PK/PD) measurement at the single-cell level, and has helped answer key questions regarding the biological mechanisms of in vivo NP behavior. This review summarizes progress to date and also describes useful technical strategies for successful IVM experimentation.
Collapse
Affiliation(s)
- Miles A Miller
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, Boston, MA 02114, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, Boston, MA 02114, USA; Department of Systems Biology, Harvard Medical School, 200 Longwood Ave, Boston, MA 02115, USA.
| |
Collapse
|
414
|
Brown JM, Recht L, Strober S. The Promise of Targeting Macrophages in Cancer Therapy. Clin Cancer Res 2017; 23:3241-3250. [PMID: 28341752 DOI: 10.1158/1078-0432.ccr-16-3122] [Citation(s) in RCA: 248] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 01/17/2017] [Accepted: 03/17/2017] [Indexed: 12/14/2022]
Abstract
Cancer therapy has developed around the concept of killing, or stopping the growth of, the cancer cells. Molecularly targeted therapy is the modern expression of this paradigm. Increasingly, however, the realization that the cancer has co-opted the normal cells of the stroma for its own survival has led to the concept that the tumor microenvironment (TME) could be targeted for effective therapy. In this review, we outline the importance of tumor-associated macrophages (TAM), a major component of the TME, in the response of tumors to cancer therapy. We discuss the normal role of macrophages in wound healing, the major phenotypes of TAMs, and their role in blunting the efficacy of cancer treatment by radiation and anticancer drugs, both by promoting tumor angiogenesis and by suppressing antitumor immunity. Finally, we review the many preclinical studies that have shown that the response of tumors to irradiation and anticancer drugs can be improved, sometimes markedly so, by depleting TAMs from tumors or by suppressing their polarization from an M1 to an M2 phenotype. The data clearly support the validity of clinical testing of combining targeting TAMs with conventional therapy. Clin Cancer Res; 23(13); 3241-50. ©2017 AACR.
Collapse
Affiliation(s)
- J Martin Brown
- Department of Radiation Oncology, Stanford University, Stanford, California.
| | - Lawrence Recht
- Department of Neurology, Stanford University, Stanford, California
| | - Samuel Strober
- Department of Medicine, Stanford University, Stanford, California
| |
Collapse
|
415
|
Torcellan T, Stolp J, Chtanova T. In Vivo Imaging Sheds Light on Immune Cell Migration and Function in Cancer. Front Immunol 2017; 8:309. [PMID: 28382036 PMCID: PMC5360706 DOI: 10.3389/fimmu.2017.00309] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 03/06/2017] [Indexed: 01/04/2023] Open
Abstract
There is ample evidence for both beneficial and harmful involvement of the immune system in tumor development and spread. Immune cell recruitment to tumors is essential not only for the success of anticancer immune therapies but also for tumor-induced immune suppression. Now that immune-based therapies are playing an increasingly important role in treatment of solid tumors such as metastatic melanomas, precise analysis of the in vivo contributions of different leukocyte subsets in tumor immunity has become an even greater priority. Recently, this goal has been markedly facilitated by the use of intravital microscopy, which has enabled us to visualize the dynamic interactions between cells of the immune system and tumor targets in the context of the tumor microenvironment. For example, intravital imaging techniques have shed new light on T cell infiltration of tumors, the mechanisms of cancer cell killing, and how myeloid cells contribute to tumor tolerance and spread. This mini-review summarizes the recent advances made to our understanding of the roles of innate and adaptive immune cells in cancer based on the use of these in vivo imaging approaches.
Collapse
Affiliation(s)
- Tommaso Torcellan
- Immunology Division, Garvan Institute of Medical Research, Sydney, NSW, Australia; Faculty of Medicine, St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Jessica Stolp
- Immunology Division, Garvan Institute of Medical Research, Sydney, NSW, Australia; Faculty of Medicine, St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Tatyana Chtanova
- Immunology Division, Garvan Institute of Medical Research, Sydney, NSW, Australia; Faculty of Medicine, St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
416
|
Custódio-Santos T, Videira M, Brito MA. Brain metastasization of breast cancer. Biochim Biophys Acta Rev Cancer 2017; 1868:132-147. [PMID: 28341420 DOI: 10.1016/j.bbcan.2017.03.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 03/17/2017] [Accepted: 03/18/2017] [Indexed: 01/15/2023]
Abstract
Central nervous system metastases have been reported in 15-25% of breast cancer patients, and the incidence is increasing. Moreover, the survival of these patients is generally poor, with reports of a 1-year survival rate of 20%. Therefore, a better knowledge about the determinants of brain metastasization is essential for the improvement of the clinical outcomes. Here, we summarize the current data about the metastatic cascade, ranging from the output of cancer cells from the primary tumour to their colonization in the brain, which involves the epithelial-mesenchymal transition, invasion of mammary tissue, intravasation into circulation, and homing into and extravasation towards the brain. The phenotypic change in malignant cells, and the importance of the microenvironment in the formation of brain metastases are also inspected. Finally, the importance of genetic and epigenetic changes, and the recently disclosed effects of microRNAs in brain metastasization of breast cancer are highlighted.
Collapse
Affiliation(s)
- Tânia Custódio-Santos
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Mafalda Videira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; Department of Galenic Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Maria Alexandra Brito
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| |
Collapse
|
417
|
Turrini R, Pabois A, Xenarios I, Coukos G, Delaloye JF, Doucey MA. TIE-2 expressing monocytes in human cancers. Oncoimmunology 2017; 6:e1303585. [PMID: 28507810 PMCID: PMC5414874 DOI: 10.1080/2162402x.2017.1303585] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 02/28/2017] [Accepted: 03/01/2017] [Indexed: 12/13/2022] Open
Abstract
Tumor-associated macrophages (TAM) are well known as a key player in the tumor microenvironment, which support cancer progression. More recently, a lineage of monocytes characterized by the expression of the TIE-2/Tek angiopoietin receptor identified a subset of circulating and tumor-associated monocytes endowed with proangiogenic activity. TIE-2 expressing monocytes (TEM) were found both in humans and mice. Here, we review the phenotypes and functions of TEM reported so far in human cancer and their potential use as markers of cancer progression and metastasis. Finally, we discuss the therapeutic approaches currently used or proposed to target TEM.
Collapse
Affiliation(s)
- Riccardo Turrini
- Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Angélique Pabois
- Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Ioannis Xenarios
- Vital-IT, Swiss Institute of Bioinformatics, University of Lausanne, Lausanne, Switzerland
| | - George Coukos
- Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | | | - Marie-Agnès Doucey
- Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
418
|
Labernadie A, Kato T, Brugués A, Serra-Picamal X, Derzsi S, Arwert E, Weston A, González-Tarragó V, Elosegui-Artola A, Albertazzi L, Alcaraz J, Roca-Cusachs P, Sahai E, Trepat X. A mechanically active heterotypic E-cadherin/N-cadherin adhesion enables fibroblasts to drive cancer cell invasion. Nat Cell Biol 2017; 19:224-237. [PMID: 28218910 PMCID: PMC5831988 DOI: 10.1038/ncb3478] [Citation(s) in RCA: 547] [Impact Index Per Article: 68.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 01/18/2017] [Indexed: 02/08/2023]
Abstract
Cancer-associated fibroblasts (CAFs) promote tumour invasion and metastasis. We show that CAFs exert a physical force on cancer cells that enables their collective invasion. Force transmission is mediated by a heterophilic adhesion involving N-cadherin at the CAF membrane and E-cadherin at the cancer cell membrane. This adhesion is mechanically active; when subjected to force it triggers β-catenin recruitment and adhesion reinforcement dependent on α-catenin/vinculin interaction. Impairment of E-cadherin/N-cadherin adhesion abrogates the ability of CAFs to guide collective cell migration and blocks cancer cell invasion. N-cadherin also mediates repolarization of the CAFs away from the cancer cells. In parallel, nectins and afadin are recruited to the cancer cell/CAF interface and CAF repolarization is afadin dependent. Heterotypic junctions between CAFs and cancer cells are observed in patient-derived material. Together, our findings show that a mechanically active heterophilic adhesion between CAFs and cancer cells enables cooperative tumour invasion.
Collapse
Affiliation(s)
- Anna Labernadie
- Institute for Bioengineering of Catalonia, Barcelona 08028,
Spain
| | - Takuya Kato
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT,
UK
| | - Agustí Brugués
- Institute for Bioengineering of Catalonia, Barcelona 08028,
Spain
| | - Xavier Serra-Picamal
- Institute for Bioengineering of Catalonia, Barcelona 08028,
Spain
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina,
Universitat de Barcelona, Barcelona 08036, Spain
| | - Stefanie Derzsi
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT,
UK
| | - Esther Arwert
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT,
UK
| | - Anne Weston
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT,
UK
| | | | | | | | - Jordi Alcaraz
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina,
Universitat de Barcelona, Barcelona 08036, Spain
| | - Pere Roca-Cusachs
- Institute for Bioengineering of Catalonia, Barcelona 08028,
Spain
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina,
Universitat de Barcelona, Barcelona 08036, Spain
| | - Erik Sahai
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT,
UK
| | - Xavier Trepat
- Institute for Bioengineering of Catalonia, Barcelona 08028,
Spain
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina,
Universitat de Barcelona, Barcelona 08036, Spain
- Institució Catalana de Recerca i Estudis Avançats
(ICREA), Barcelona 08010, Spain
- Centro de Investigación Biomédica en Red en
Bioingeniería, Biomateriales y Nanomedicina, Barcelona 08028, Spain
| |
Collapse
|
419
|
Heterogeneity of macrophage infiltration and therapeutic response in lung carcinoma revealed by 3D organ imaging. Nat Commun 2017; 8:14293. [PMID: 28176769 PMCID: PMC5309815 DOI: 10.1038/ncomms14293] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 12/14/2016] [Indexed: 12/19/2022] Open
Abstract
Involvement of the immune system in tumour progression is at the forefront of cancer research. Analysis of the tumour immune microenvironment has yielded a wealth of information on tumour biology, and alterations in some immune subtypes, such as tumour-associated macrophages (TAM), can be strong prognostic indicators. Here, we use optical tissue clearing and a TAM-targeting injectable fluorescent nanoparticle (NP) to examine three-dimensional TAM composition, tumour-to-tumour heterogeneity, response to colony-stimulating factor 1 receptor (CSF-1R) blockade and nanoparticle-based drug delivery in murine pulmonary carcinoma. The method allows for rapid tumour volume assessment and spatial information on TAM infiltration at the cellular level in entire lungs. This method reveals that TAM density was heterogeneous across tumours in the same animal, overall TAM density is different among separate pulmonary tumour models, nanotherapeutic drug delivery correlated with TAM heterogeneity, and successful response to CSF-1R blockade is characterized by enhanced TAM penetration throughout and within tumours. Tumour-associated macrophages (TAM) can be used as prognostic indicators in cancer. Here, the authors establish a platform for high-throughput 3D microscopy in murine lung carcinoma that allows to visualize TAMs infiltration throughout the entire lung, response to CSF-1R blockade and nanoparticle drug delivery.
Collapse
|
420
|
Macrophages as Key Drivers of Cancer Progression and Metastasis. Mediators Inflamm 2017; 2017:9624760. [PMID: 28210073 PMCID: PMC5292164 DOI: 10.1155/2017/9624760] [Citation(s) in RCA: 212] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 12/08/2016] [Indexed: 12/12/2022] Open
Abstract
Macrophages are one of the most abundant immune cells in the tumour microenvironment of solid tumours and their presence correlates with reduced survival in most cancers. Macrophages are present at all stages of tumour progression and stimulate angiogenesis, tumour cell invasion, and intravasation at the primary site. At the metastatic site, macrophages and monocytes prepare for the arrival of disseminated tumour cells and promote their extravasation and survival by inhibiting immune-mediated clearance or by directly engaging with tumour cells to activate prosurvival signalling pathways. In addition, macrophages promote the growth of disseminated tumour cells at the metastatic site by organising the formation of a supportive metastatic niche. The development of agents inhibiting the recruitment or the protumorigenic effector functions of macrophages in both the primary tumour and at the metastatic site is a promising strategy to improve cancer survival in the future.
Collapse
|
421
|
Abstract
The significant parallels between cell plasticity during embryonic development and carcinoma progression have helped us understand the importance of the epithelial-mesenchymal transition (EMT) in human disease. Our expanding knowledge of EMT has led to a clarification of the EMT program as a set of multiple and dynamic transitional states between the epithelial and mesenchymal phenotypes, as opposed to a process involving a single binary decision. EMT and its intermediate states have recently been identified as crucial drivers of organ fibrosis and tumor progression, although there is some need for caution when interpreting its contribution to metastatic colonization. Here, we discuss the current state-of-the-art and latest findings regarding the concept of cellular plasticity and heterogeneity in EMT. We raise some of the questions pending and identify the challenges faced in this fast-moving field.
Collapse
|
422
|
Tumor Associated Macrophages as Therapeutic Targets for Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1026:331-370. [PMID: 29282692 DOI: 10.1007/978-981-10-6020-5_16] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Tumor-associated macrophages (TAMs) are the most abundant inflammatory infiltrates in the tumor stroma. TAMs promote tumor growth by suppressing immunocompetent cells, including neovascularization and supporting cancer stem cells. In the chapter, we discuss recent efforts in reprogramming or inhibiting tumor-protecting properties of TAMs, and developing potential strategies to increase the efficacy of breast cancer treatment.
Collapse
|
423
|
Kohrman AQ, Matus DQ. Divide or Conquer: Cell Cycle Regulation of Invasive Behavior. Trends Cell Biol 2017; 27:12-25. [PMID: 27634432 PMCID: PMC5186408 DOI: 10.1016/j.tcb.2016.08.003] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 07/30/2016] [Accepted: 08/10/2016] [Indexed: 12/18/2022]
Abstract
Cell invasion through the basement membrane (BM) occurs during normal embryonic development and is a fundamental feature of cancer metastasis. The underlying cellular and genetic machinery required for invasion has been difficult to identify, due to a lack of adequate in vivo models to accurately examine invasion in single cells at subcellular resolution. Recent evidence has documented a functional link between cell cycle arrest and invasive activity. While cancer progression is traditionally thought of as a disease of uncontrolled cell proliferation, cancer cell dissemination, a critical aspect of metastasis, may require a switch from a proliferative to an invasive state. In this work, we review evidence that BM invasion requires cell cycle arrest and discuss the implications of this concept with regard to limiting the lethality associated with cancer metastasis.
Collapse
Affiliation(s)
- Abraham Q Kohrman
- 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
|
424
|
Novel insights in the regulation and function of macrophages in the tumor microenvironment. Curr Opin Oncol 2017; 29:55-61. [DOI: 10.1097/cco.0000000000000344] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
425
|
Oudin MJ, Gertler FB. Signatures of Breast Cancer Metastasis: aMENAble to Interpretation? Trends Cancer 2017; 3:7-9. [PMID: 28718427 DOI: 10.1016/j.trecan.2016.11.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 11/03/2016] [Accepted: 11/07/2016] [Indexed: 11/18/2022]
Abstract
MenaINV, an isoform of the motility regulator protein Mena, contributes to prometastatic phenotypes. Tumor microenvironment of metastasis (TMEM), a three-cell structure associated with intravasation, contains a stationary Mena-expressing tumor cell. TMEM density and MenaINV expression both correlate with poor clinical outcome in breast cancer patients. However, is MenaINV involved in TMEM assembly and function?
Collapse
Affiliation(s)
- Madeleine J Oudin
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA.
| | - Frank B Gertler
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA; Department of Biology, MIT, Cambridge, MA, USA.
| |
Collapse
|
426
|
Sparano JA, Gray R, Oktay MH, Entenberg D, Rohan T, Xue X, Donovan M, Peterson M, Shuber A, Hamilton DA, D’Alfonso T, Goldstein LJ, Gertler F, Davidson NE, Condeelis J, Jones J. A metastasis biomarker (MetaSite Breast™ Score) is associated with distant recurrence in hormone receptor-positive, HER2-negative early-stage breast cancer. NPJ Breast Cancer 2017; 3:42. [PMID: 29138761 PMCID: PMC5678158 DOI: 10.1038/s41523-017-0043-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 08/24/2017] [Accepted: 09/22/2017] [Indexed: 12/16/2022] Open
Abstract
Metastasis is the primary cause of death in early-stage breast cancer. We evaluated the association between a metastasis biomarker, which we call "Tumor Microenviroment of Metastasis" (TMEM), and risk of recurrence. TMEM are microanatomic structures where invasive tumor cells are in direct contact with endothelial cells and macrophages, and which serve as intravasation sites for tumor cells into the circulation. We evaluated primary tumors from 600 patients with Stage I-III breast cancer treated with adjuvant chemotherapy in trial E2197 (NCT00003519), plus endocrine therapy for hormone receptor (HR)+ disease. TMEM were identified and enumerated using an analytically validated, fully automated digital pathology/image analysis method (MetaSite Breast™), hereafter referred to as MetaSite Score (MS). The objectives were to determine the association between MS and distant relapse free interval (DRFI) and relapse free interval (RFI). MS was not associated with tumor size or nodal status, and correlated poorly with Oncotype DX Recurrence Score (r = 0.29) in 297 patients with HR+/HER2- disease. Proportional hazards models revealed a significant positive association between continuous MS and DRFI (p = 0.001) and RFI (p = 0.00006) in HR+/HER2- disease in years 0-5, and by MS tertiles for DRFI (p = 0.04) and RFI (p = 0.01), but not after year 5 or in triple negative or HER2+ disease. Multivariate models in HR+/HER- disease including continuous MS, clinical covariates, and categorical Recurrence Score (<18, 18-30, > 30) showed MS is an independent predictor for 5-year RFI (p = 0.05). MetaSite Score provides prognostic information for early recurrence complementary to clinicopathologic features and Recurrence Score.
Collapse
Affiliation(s)
- Joseph A. Sparano
- 0000 0001 2152 0791grid.240283.fMontefiore Medical Center, Albert Einstein College of Medicine, 1695 Eastchester Road, 10461 Bronx, NY USA
| | | | - Maja H. Oktay
- 0000 0001 2152 0791grid.240283.fMontefiore Medical Center, Albert Einstein College of Medicine, 1695 Eastchester Road, 10461 Bronx, NY USA
| | - David Entenberg
- 0000 0001 2152 0791grid.240283.fAlbert Einstein College of Medicine, Bronx, NY USA
| | - Thomas Rohan
- 0000 0001 2152 0791grid.240283.fAlbert Einstein College of Medicine, Bronx, NY USA
| | - Xiaonan Xue
- 0000 0001 2152 0791grid.240283.fAlbert Einstein College of Medicine, Bronx, NY USA
| | - Michael Donovan
- 0000 0001 0670 2351grid.59734.3cMt. Sinai School of Medicine, New York, NY USA
| | | | | | | | | | - Lori J. Goldstein
- 0000 0004 0456 6466grid.412530.1Fox Chase Cancer Center, Philadelphia, PA USA
| | - Frank Gertler
- 0000 0001 2341 2786grid.116068.8Massachusetts Institute of Technology, Boston, MA USA
| | - Nancy E. Davidson
- 0000 0004 0456 9819grid.478063.eUniversity of Pittsburgh Cancer Institute, Pittsburgh, PA USA
| | - John Condeelis
- 0000 0001 2152 0791grid.240283.fAlbert Einstein College of Medicine, Bronx, NY USA
| | - Joan Jones
- 0000 0001 2152 0791grid.240283.fAlbert Einstein College of Medicine, Bronx, NY USA
| |
Collapse
|
427
|
He C, Zhou Z, Jiang H, Yin Z, Meng S, Zhang J, Huang P, Xu K, Bian L, Xiao Z, Wang J. Epithelial-Mesenchymal Transition is Superior to Vessels-Encapsulate Tumor Cluster in Promoting Metastasis of Hepatocellular Carcinoma: a Morphological Evidence. J Cancer 2017; 8:39-47. [PMID: 28123596 PMCID: PMC5264038 DOI: 10.7150/jca.16736] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/18/2016] [Indexed: 12/12/2022] Open
Abstract
Purpose Vessels-encapsulate tumor cluster (VETC) is a vascular pattern distinct from classical capillary-like pattern. It is reported that VETC structure is common in hepatocellular carcinoma (HCC) and can promote HCC metastasis in an epithelial-mesenchymal transition (EMT)-independent but VETC-dependent manner. However, the main metastatic manner of HCC containing both VETC and classical vascular structure (we called VETC±) is unknown. Methods Vascular pattern types and E-cadherin expression were evaluated by immunohistochemical staining in 168 HCC tissues, 50 pairs of primary HCC tissues and intrahepatic metastatic lesions, as well as 12 pairs of primary HCC tissues and major portal vein tumor thrombus. Survival and recurrence rates were evaluated using Kaplan-Meier analysis. The multivariate Cox proportional hazards model was used to determine the independent prognostic factors of HCC. Results VETC± cases were more common than VETC+ cases (HCC tissues with a VETC pattern fully distributed in the HCC section) in HCC. Statistical analysis showed that VETC± was an independent predictor of survival and recurrence. Furthermore, E-cadherin was positively correlated with the presence of VETC structure. In the case of HCCs with VETC±, their metastases (both intrahepatic and major vascular) were more likely to be VETC negative. Conclusions Our findings suggest that EMT may be superior to VETC in promoting HCC metastasis. Thus, both anti-EMT and anti-VETC agents should be considered in the case of HCC with VETC±.
Collapse
Affiliation(s)
- Chuanchao He
- Guangdong Province Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Research Center of Medicine, Sun Yat-Sen Memorial Hospital, No. 107 Yanjiang Western Road, Guangzhou 510120, China
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, No. 33 Yingfeng Road, Guangzhou 510289, China
| | - Zhenyu Zhou
- Guangdong Province Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Research Center of Medicine, Sun Yat-Sen Memorial Hospital, No. 107 Yanjiang Western Road, Guangzhou 510120, China
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, No. 33 Yingfeng Road, Guangzhou 510289, China
| | - Hai Jiang
- Guangdong Province Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Research Center of Medicine, Sun Yat-Sen Memorial Hospital, No. 107 Yanjiang Western Road, Guangzhou 510120, China
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, No. 33 Yingfeng Road, Guangzhou 510289, China
| | - Zi Yin
- General Surgery Department, Guangdong General Hospital, Guangdong Academy of Medical Sciences, No. 106 Zhongshan Er Road., Guangzhou, 510080, China
| | - Shiyu Meng
- Guangdong Province Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Research Center of Medicine, Sun Yat-Sen Memorial Hospital, No. 107 Yanjiang Western Road, Guangzhou 510120, China
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, No. 33 Yingfeng Road, Guangzhou 510289, China
| | - Jianlong Zhang
- Guangdong Province Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Research Center of Medicine, Sun Yat-Sen Memorial Hospital, No. 107 Yanjiang Western Road, Guangzhou 510120, China
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, No. 33 Yingfeng Road, Guangzhou 510289, China
| | - Pinbo Huang
- Guangdong Province Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Research Center of Medicine, Sun Yat-Sen Memorial Hospital, No. 107 Yanjiang Western Road, Guangzhou 510120, China
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, No. 33 Yingfeng Road, Guangzhou 510289, China
| | - Kang Xu
- Guangdong Province Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Research Center of Medicine, Sun Yat-Sen Memorial Hospital, No. 107 Yanjiang Western Road, Guangzhou 510120, China
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, No. 33 Yingfeng Road, Guangzhou 510289, China
| | - Lijuan Bian
- Guangdong Province Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Research Center of Medicine, Sun Yat-Sen Memorial Hospital, No. 107 Yanjiang Western Road, Guangzhou 510120, China
- Department of Pathology, Sun Yat-Sen Memorial Hospital, No. 107 Yanjiang Western Road, Guangzhou 510120, China
| | - Zhiyu Xiao
- Guangdong Province Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Research Center of Medicine, Sun Yat-Sen Memorial Hospital, No. 107 Yanjiang Western Road, Guangzhou 510120, China
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, No. 33 Yingfeng Road, Guangzhou 510289, China
| | - Jie Wang
- Guangdong Province Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Research Center of Medicine, Sun Yat-Sen Memorial Hospital, No. 107 Yanjiang Western Road, Guangzhou 510120, China
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, No. 33 Yingfeng Road, Guangzhou 510289, China
| |
Collapse
|
428
|
Cortes J, Talpaz M, Smith HP, Snyder DS, Khoury J, Bhalla KN, Pinilla-Ibarz J, Larson R, Mitchell D, Wise SC, Rutkoski TJ, Smith BD, Flynn DL, Kantarjian HM, Rosen O, Van Etten RA. Phase 1 dose-finding study of rebastinib (DCC-2036) in patients with relapsed chronic myeloid leukemia and acute myeloid leukemia. Haematologica 2016; 102:519-528. [PMID: 27927766 PMCID: PMC5394958 DOI: 10.3324/haematol.2016.152710] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 11/29/2016] [Indexed: 01/03/2023] Open
Abstract
A vailable tyrosine kinase inhibitors for chronic myeloid leukemia bind in an adenosine 5′-triphosphate-binding pocket and are affected by evolving mutations that confer resistance. Rebastinib was identified as a switch control inhibitor of BCR-ABL1 and FLT3 and may be active against resistant mutations. A Phase 1, first-in-human, single-agent study investigated rebastinib in relapsed or refractory chronic or acute myeloid leukemia. The primary objectives were to investigate the safety of rebastinib and establish the maximum tolerated dose and recommended Phase 2 dose. Fifty-seven patients received treatment with rebastinib. Sixteen patients were treated using powder-in-capsule preparations at doses from 57 mg to 1200 mg daily, and 41 received tablet preparations at doses of 100 mg to 400 mg daily. Dose-limiting toxicities were dysarthria, muscle weakness, and peripheral neuropathy. The maximum tolerated dose was 150 mg tablets administered twice daily. Rebastinib was rapidly absorbed. Bioavailability was 3- to 4-fold greater with formulated tablets compared to unformulated capsules. Eight complete hematologic responses were achieved in 40 evaluable chronic myeloid leukemia patients, 4 of which had a T315I mutation. None of the 5 patients with acute myeloid leukemia responded. Pharmacodynamic analysis showed inhibition of phosphorylation of substrates of BCR-ABL1 or FLT3 by rebastinib. Although clinical activity was observed, clinical benefit was insufficient to justify continued development in chronic or acute myeloid leukemia. Pharmacodynamic analyses suggest that other kinases inhibited by rebastinib, such as TIE2, may be more relevant targets for the clinical development of rebastinib (clinicaltrials.gov Identifier:00827138).
Collapse
Affiliation(s)
- Jorge Cortes
- University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | - Jean Khoury
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Kapil N Bhalla
- University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
429
|
Pignatelli J, Bravo-Cordero JJ, Roh-Johnson M, Gandhi SJ, Wang Y, Chen X, Eddy RJ, Xue A, Singer RH, Hodgson L, Oktay MH, Condeelis JS. Macrophage-dependent tumor cell transendothelial migration is mediated by Notch1/Mena INV-initiated invadopodium formation. Sci Rep 2016; 6:37874. [PMID: 27901093 PMCID: PMC5129016 DOI: 10.1038/srep37874] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 11/02/2016] [Indexed: 12/27/2022] Open
Abstract
The process of intravasation involving transendothelial migration is a key step in metastatic spread. How the triple cell complex composed of a macrophage, Mena over-expressing tumor cell and endothelial cell, called the tumor microenvironment of metastasis (TMEM), facilitates tumor cell transendothelial migration is not completely understood. Previous work has shown that the physical contact between a macrophage and tumor cell results in the formation of invadopodia, actin-rich matrix degrading protrusions, important for tumor cell invasion and transendothelial migration and tumor cell dissemination. Herein, we show that the macrophage-induced invadopodium is formed through a Notch1/MenaINV signaling pathway in the tumor cell upon macrophage contact. This heterotypic tumor cell – macrophage interaction results in the upregulation of MenaINV through the activation of MENA transcription. Notch1 and MenaINV expression are required for tumor cell transendothelial migration, a necessary step during intravasation. Inhibition of the Notch signaling pathway blocked macrophage-induced invadopodium formation in vitro and the dissemination of tumor cells from the primary tumor in vivo. Our findings indicate a novel role for Notch1 signaling in the regulation of MenaINV expression and transendothelial migration and provide mechanistic information essential to the use of therapeutic inhibitors of metastasis.
Collapse
Affiliation(s)
- Jeanine Pignatelli
- Department of Anatomy and Structural Biology Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States.,Gruss Lipper Biophotonics Center Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States
| | - Jose Javier Bravo-Cordero
- Department of Anatomy and Structural Biology Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States.,Gruss Lipper Biophotonics Center Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States
| | - Minna Roh-Johnson
- Department of Anatomy and Structural Biology Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States.,Gruss Lipper Biophotonics Center Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States
| | - Saumil J Gandhi
- Department of Anatomy and Structural Biology Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States
| | - Yarong Wang
- Department of Anatomy and Structural Biology Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States.,Gruss Lipper Biophotonics Center Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States
| | - Xiaoming Chen
- Department of Anatomy and Structural Biology Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States
| | - Robert J Eddy
- Department of Anatomy and Structural Biology Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States
| | - Alice Xue
- Department of Anatomy and Structural Biology Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States
| | - Robert H Singer
- Department of Anatomy and Structural Biology Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States
| | - Louis Hodgson
- Department of Anatomy and Structural Biology Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States.,Gruss Lipper Biophotonics Center Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States
| | - Maja H Oktay
- Department of Anatomy and Structural Biology Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States.,Department of Pathology Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States
| | - John S Condeelis
- Department of Anatomy and Structural Biology Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States.,Gruss Lipper Biophotonics Center Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States
| |
Collapse
|
430
|
Blood vessel endothelium-directed tumor cell streaming in breast tumors requires the HGF/C-Met signaling pathway. Oncogene 2016; 36:2680-2692. [PMID: 27893712 PMCID: PMC5426963 DOI: 10.1038/onc.2016.421] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 09/13/2016] [Accepted: 09/30/2016] [Indexed: 01/14/2023]
Abstract
During metastasis to distant sites, tumor cells migrate to blood vessels. In vivo, breast tumor cells utilize a specialized mode of migration known as streaming, where a linear assembly of tumor cells migrate directionally towards blood vessels on fibronectin-collagen I-containing extracellular matrix (ECM) fibers in response to chemotactic signals. We have successfully reconstructed tumor cell streaming in vitro by co-plating tumors cells, macrophages and endothelial cells on 2.5 μm thick ECM-coated micro-patterned substrates. We found that tumor cells and macrophages, when plated together on the micro-patterned substrates, do not demonstrate sustained directional migration in only one direction (sustained directionality) but show random bi-directional walking. Sustained directionality of tumor cells as seen in vivo was established in vitro when beads coated with human umbilical vein endothelial cells were placed at one end of the micro-patterned 'ECM fibers' within the assay. We demonstrated that these endothelial cells supply the hepatocyte growth factor (HGF) required for the chemotactic gradient responsible for sustained directionality. Using this in vitro reconstituted streaming system, we found that directional streaming is dependent on, and most effectively blocked, by inhibiting the HGF/C-Met signaling pathway between endothelial cells and tumor cells. Key observations made with the in vitro reconstituted system implicating C-Met signaling were confirmed in vivo in mammary tumors using the in vivo invasion assay and intravital multiphoton imaging of tumor cell streaming. These results establish HGF/C-Met as a central organizing signal in blood vessel-directed tumor cell migration in vivo and highlight a promising role for C-Met inhibitors in blocking tumor cell streaming and metastasis in vivo, and for use in human trials.
Collapse
|
431
|
Mena INV dysregulates cortactin phosphorylation to promote invadopodium maturation. Sci Rep 2016; 6:36142. [PMID: 27824079 PMCID: PMC5099927 DOI: 10.1038/srep36142] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 10/11/2016] [Indexed: 01/12/2023] Open
Abstract
Invadopodia, actin-based protrusions of invasive carcinoma cells that focally activate extracellular matrix-degrading proteases, are essential for the migration and intravasation of tumor cells during dissemination from the primary tumor. We have previously shown that cortactin phosphorylation at tyrosine residues, in particular tyrosine 421, promotes actin polymerization at newly-forming invadopodia, promoting their maturation to matrix-degrading structures. However, the mechanism by which cells regulate the cortactin tyrosine phosphorylation-dephosphorylation cycle at invadopodia is unknown. Mena, an actin barbed-end capping protein antagonist, is expressed as various splice-isoforms. The MenaINV isoform is upregulated in migratory and invasive sub-populations of breast carcinoma cells, and is involved in tumor cell intravasation. Here we show that forced MenaINV expression increases invadopodium maturation to a far greater extent than equivalent expression of other Mena isoforms. MenaINV is recruited to invadopodium precursors just after their initial assembly at the plasma membrane, and promotes the phosphorylation of cortactin tyrosine 421 at invadopodia. In addition, we show that cortactin phosphorylation at tyrosine 421 is suppressed by the phosphatase PTP1B, and that PTP1B localization to the invadopodium is reduced by MenaINV expression. We conclude that MenaINV promotes invadopodium maturation by inhibiting normal dephosphorylation of cortactin at tyrosine 421 by the phosphatase PTP1B.
Collapse
|
432
|
Rodriguez-Tirado C, Kitamura T, Kato Y, Pollard JW, Condeelis JS, Entenberg D. Long-term High-Resolution Intravital Microscopy in the Lung with a Vacuum Stabilized Imaging Window. J Vis Exp 2016. [PMID: 27768066 DOI: 10.3791/54603] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Metastasis to secondary sites such as the lung, liver and bone is a traumatic event with a mortality rate of approximately 90% 1. Of these sites, the lung is the most difficult to assess using intravital optical imaging due to its enclosed position within the body, delicate nature and vital role in sustaining proper physiology. While clinical modalities (positron emission tomography (PET), magnetic resonance imaging (MRI) and computed tomography (CT)) are capable of providing noninvasive images of this tissue, they lack the resolution necessary to visualize the earliest seeding events, with a single pixel consisting of nearly a thousand cells. Current models of metastatic lung seeding postulate that events just after a tumor cell's arrival are deterministic for survival and subsequent growth. This means that real-time intravital imaging tools with single cell resolution 2 are required in order to define the phenotypes of the seeding cells and test these models. While high resolution optical imaging of the lung has been performed using various ex vivo preparations, these experiments are typically single time-point assays and are susceptible to artifacts and possible erroneous conclusions due to the dramatically altered environment (temperature, profusion, cytokines, etc.) resulting from removal from the chest cavity and circulatory system 3. Recent work has shown that time-lapse intravital optical imaging of the intact lung is possible using a vacuum stabilized imaging window 2,4,5 however, typical imaging times have been limited to approximately 6 hr. Here we describe a protocol for performing long-term intravital time-lapse imaging of the lung utilizing such a window over a period of 12 hr. The time-lapse image sequences obtained using this method enable visualization and quantitation of cell-cell interactions, membrane dynamics and vascular perfusion in the lung. We further describe an image processing technique that gives an unprecedentedly clear view of the lung microvasculature.
Collapse
Affiliation(s)
| | - Takanori Kitamura
- Medical Research Council Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh
| | - Yu Kato
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine; Department of Obstetrics/Gynecology and Woman's Health, Albert Einstein College of Medicine
| | - Jeffery W Pollard
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine; Department of Obstetrics/Gynecology and Woman's Health, Albert Einstein College of Medicine; Medical Research Council Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh
| | - John S Condeelis
- Department of Anatomy & Structural Biology, Albert Einstein College of Medicine; Gruss-Lipper Biophotonics Center Integrated Imaging Program, Albert Einstein College of Medicine
| | - David Entenberg
- Department of Anatomy & Structural Biology, Albert Einstein College of Medicine; Gruss-Lipper Biophotonics Center Integrated Imaging Program, Albert Einstein College of Medicine;
| |
Collapse
|
433
|
Velaei K, Samadi N, Barazvan B, Soleimani Rad J. Tumor microenvironment-mediated chemoresistance in breast cancer. Breast 2016; 30:92-100. [PMID: 27668856 DOI: 10.1016/j.breast.2016.09.002] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 09/02/2016] [Accepted: 09/02/2016] [Indexed: 12/20/2022] Open
Abstract
Therapy resistance or tumor relapse in cancer is common. Tumors develop resistance to chemotherapeutic through a variety of mechanisms, with tumor microenvironment (TM) serving pivotal roles. Using breast cancer as a paradigm, we propose that responses of cancer cells to drugs are not exclusively determined by their intrinsic characteristics but are also controlled by deriving signals from TM. Affected microenvironment by chemotherapy is an avenue to promote phenotype which tends to resist on to be ruined. Therefore, exclusively targeting cancer cells does not demolish tumor recurrence after chemotherapy. Regardless of tumor-microenvironment pathways and their profound influence on the responsiveness of treatment, diversity of molecular properties of breast cancer also behave differently in terms of response to chemotherapy. And also it is assumed that there is cross-talk between phenotypic diversity and TM. Collectively, raising complex signal from TM in chemotherapy condition often encourages cancer cells are not killed but strengthen. Here, we summarized how TM modifies responses to chemotherapy in breast cancer. We also discussed successful treatment strategies have been considered TM in breast cancer treatment.
Collapse
Affiliation(s)
- Kobra Velaei
- Department of Anatomical Science, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nasser Samadi
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Balal Barazvan
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jafar Soleimani Rad
- Department of Anatomical Science, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
434
|
He H, Mack JJ, Güç E, Warren CM, Squadrito ML, Kilarski WW, Baer C, Freshman RD, McDonald AI, Ziyad S, Swartz MA, De Palma M, Iruela-Arispe ML. Perivascular Macrophages Limit Permeability. Arterioscler Thromb Vasc Biol 2016; 36:2203-2212. [PMID: 27634833 DOI: 10.1161/atvbaha.116.307592] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 08/31/2016] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Perivascular cells, including pericytes, macrophages, smooth muscle cells, and other specialized cell types, like podocytes, participate in various aspects of vascular function. However, aside from the well-established roles of smooth muscle cells and pericytes, the contributions of other vascular-associated cells are poorly understood. Our goal was to ascertain the function of perivascular macrophages in adult tissues under nonpathological conditions. APPROACH AND RESULTS We combined confocal microscopy, in vivo cell depletion, and in vitro assays to investigate the contribution of perivascular macrophages to vascular function. We found that resident perivascular macrophages are associated with capillaries at a frequency similar to that of pericytes. Macrophage depletion using either clodronate liposomes or antibodies unexpectedly resulted in hyperpermeability. This effect could be rescued when M2-like macrophages, but not M1-like macrophages or dendritic cells, were reconstituted in vivo, suggesting subtype-specific roles for macrophages in the regulation of vascular permeability. Furthermore, we found that permeability-promoting agents elicit motility and eventual dissociation of macrophages from the vasculature. Finally, in vitro assays showed that M2-like macrophages attenuate the phosphorylation of VE-cadherin upon exposure to permeability-promoting agents. CONCLUSIONS This study points to a direct contribution of macrophages to vessel barrier integrity and provides evidence that heterotypic cell interactions with the endothelium, in addition to those of pericytes, control vascular permeability.
Collapse
Affiliation(s)
- Huanhuan He
- From the Department of Human Genetics (H.H.), Department of Molecular, Cell and Developmental Biology (J.J.M., C.M.W., R.D.F., A.I.M., S.Z., M.L.I.-A.), Molecular Biology Institute (M.L.I.-A.), and Jonsson Comprehensive Cancer Center (M.L.I.-A.), University of California, Los Angeles; Institute for Bioengineering (IBI) (E.G., M.A.S.) and The Swiss Institute for Experimental Cancer Research (ISREC) (M.L.S., C.B., M.A.S., M.D.P., M.L.I.-A.), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Switzerland; and Institute for Molecular Engineering and Ben May Department of Cancer Research, University of Chicago, IL (W.W.K., M.A.S.)
| | - Julia J Mack
- From the Department of Human Genetics (H.H.), Department of Molecular, Cell and Developmental Biology (J.J.M., C.M.W., R.D.F., A.I.M., S.Z., M.L.I.-A.), Molecular Biology Institute (M.L.I.-A.), and Jonsson Comprehensive Cancer Center (M.L.I.-A.), University of California, Los Angeles; Institute for Bioengineering (IBI) (E.G., M.A.S.) and The Swiss Institute for Experimental Cancer Research (ISREC) (M.L.S., C.B., M.A.S., M.D.P., M.L.I.-A.), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Switzerland; and Institute for Molecular Engineering and Ben May Department of Cancer Research, University of Chicago, IL (W.W.K., M.A.S.)
| | - Esra Güç
- From the Department of Human Genetics (H.H.), Department of Molecular, Cell and Developmental Biology (J.J.M., C.M.W., R.D.F., A.I.M., S.Z., M.L.I.-A.), Molecular Biology Institute (M.L.I.-A.), and Jonsson Comprehensive Cancer Center (M.L.I.-A.), University of California, Los Angeles; Institute for Bioengineering (IBI) (E.G., M.A.S.) and The Swiss Institute for Experimental Cancer Research (ISREC) (M.L.S., C.B., M.A.S., M.D.P., M.L.I.-A.), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Switzerland; and Institute for Molecular Engineering and Ben May Department of Cancer Research, University of Chicago, IL (W.W.K., M.A.S.)
| | - Carmen M Warren
- From the Department of Human Genetics (H.H.), Department of Molecular, Cell and Developmental Biology (J.J.M., C.M.W., R.D.F., A.I.M., S.Z., M.L.I.-A.), Molecular Biology Institute (M.L.I.-A.), and Jonsson Comprehensive Cancer Center (M.L.I.-A.), University of California, Los Angeles; Institute for Bioengineering (IBI) (E.G., M.A.S.) and The Swiss Institute for Experimental Cancer Research (ISREC) (M.L.S., C.B., M.A.S., M.D.P., M.L.I.-A.), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Switzerland; and Institute for Molecular Engineering and Ben May Department of Cancer Research, University of Chicago, IL (W.W.K., M.A.S.)
| | - Mario Leonardo Squadrito
- From the Department of Human Genetics (H.H.), Department of Molecular, Cell and Developmental Biology (J.J.M., C.M.W., R.D.F., A.I.M., S.Z., M.L.I.-A.), Molecular Biology Institute (M.L.I.-A.), and Jonsson Comprehensive Cancer Center (M.L.I.-A.), University of California, Los Angeles; Institute for Bioengineering (IBI) (E.G., M.A.S.) and The Swiss Institute for Experimental Cancer Research (ISREC) (M.L.S., C.B., M.A.S., M.D.P., M.L.I.-A.), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Switzerland; and Institute for Molecular Engineering and Ben May Department of Cancer Research, University of Chicago, IL (W.W.K., M.A.S.)
| | - Witold W Kilarski
- From the Department of Human Genetics (H.H.), Department of Molecular, Cell and Developmental Biology (J.J.M., C.M.W., R.D.F., A.I.M., S.Z., M.L.I.-A.), Molecular Biology Institute (M.L.I.-A.), and Jonsson Comprehensive Cancer Center (M.L.I.-A.), University of California, Los Angeles; Institute for Bioengineering (IBI) (E.G., M.A.S.) and The Swiss Institute for Experimental Cancer Research (ISREC) (M.L.S., C.B., M.A.S., M.D.P., M.L.I.-A.), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Switzerland; and Institute for Molecular Engineering and Ben May Department of Cancer Research, University of Chicago, IL (W.W.K., M.A.S.)
| | - Caroline Baer
- From the Department of Human Genetics (H.H.), Department of Molecular, Cell and Developmental Biology (J.J.M., C.M.W., R.D.F., A.I.M., S.Z., M.L.I.-A.), Molecular Biology Institute (M.L.I.-A.), and Jonsson Comprehensive Cancer Center (M.L.I.-A.), University of California, Los Angeles; Institute for Bioengineering (IBI) (E.G., M.A.S.) and The Swiss Institute for Experimental Cancer Research (ISREC) (M.L.S., C.B., M.A.S., M.D.P., M.L.I.-A.), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Switzerland; and Institute for Molecular Engineering and Ben May Department of Cancer Research, University of Chicago, IL (W.W.K., M.A.S.)
| | - Ryan D Freshman
- From the Department of Human Genetics (H.H.), Department of Molecular, Cell and Developmental Biology (J.J.M., C.M.W., R.D.F., A.I.M., S.Z., M.L.I.-A.), Molecular Biology Institute (M.L.I.-A.), and Jonsson Comprehensive Cancer Center (M.L.I.-A.), University of California, Los Angeles; Institute for Bioengineering (IBI) (E.G., M.A.S.) and The Swiss Institute for Experimental Cancer Research (ISREC) (M.L.S., C.B., M.A.S., M.D.P., M.L.I.-A.), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Switzerland; and Institute for Molecular Engineering and Ben May Department of Cancer Research, University of Chicago, IL (W.W.K., M.A.S.)
| | - Austin I McDonald
- From the Department of Human Genetics (H.H.), Department of Molecular, Cell and Developmental Biology (J.J.M., C.M.W., R.D.F., A.I.M., S.Z., M.L.I.-A.), Molecular Biology Institute (M.L.I.-A.), and Jonsson Comprehensive Cancer Center (M.L.I.-A.), University of California, Los Angeles; Institute for Bioengineering (IBI) (E.G., M.A.S.) and The Swiss Institute for Experimental Cancer Research (ISREC) (M.L.S., C.B., M.A.S., M.D.P., M.L.I.-A.), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Switzerland; and Institute for Molecular Engineering and Ben May Department of Cancer Research, University of Chicago, IL (W.W.K., M.A.S.)
| | - Safiyyah Ziyad
- From the Department of Human Genetics (H.H.), Department of Molecular, Cell and Developmental Biology (J.J.M., C.M.W., R.D.F., A.I.M., S.Z., M.L.I.-A.), Molecular Biology Institute (M.L.I.-A.), and Jonsson Comprehensive Cancer Center (M.L.I.-A.), University of California, Los Angeles; Institute for Bioengineering (IBI) (E.G., M.A.S.) and The Swiss Institute for Experimental Cancer Research (ISREC) (M.L.S., C.B., M.A.S., M.D.P., M.L.I.-A.), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Switzerland; and Institute for Molecular Engineering and Ben May Department of Cancer Research, University of Chicago, IL (W.W.K., M.A.S.)
| | - Melody A Swartz
- From the Department of Human Genetics (H.H.), Department of Molecular, Cell and Developmental Biology (J.J.M., C.M.W., R.D.F., A.I.M., S.Z., M.L.I.-A.), Molecular Biology Institute (M.L.I.-A.), and Jonsson Comprehensive Cancer Center (M.L.I.-A.), University of California, Los Angeles; Institute for Bioengineering (IBI) (E.G., M.A.S.) and The Swiss Institute for Experimental Cancer Research (ISREC) (M.L.S., C.B., M.A.S., M.D.P., M.L.I.-A.), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Switzerland; and Institute for Molecular Engineering and Ben May Department of Cancer Research, University of Chicago, IL (W.W.K., M.A.S.)
| | - Michele De Palma
- From the Department of Human Genetics (H.H.), Department of Molecular, Cell and Developmental Biology (J.J.M., C.M.W., R.D.F., A.I.M., S.Z., M.L.I.-A.), Molecular Biology Institute (M.L.I.-A.), and Jonsson Comprehensive Cancer Center (M.L.I.-A.), University of California, Los Angeles; Institute for Bioengineering (IBI) (E.G., M.A.S.) and The Swiss Institute for Experimental Cancer Research (ISREC) (M.L.S., C.B., M.A.S., M.D.P., M.L.I.-A.), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Switzerland; and Institute for Molecular Engineering and Ben May Department of Cancer Research, University of Chicago, IL (W.W.K., M.A.S.)
| | - M Luisa Iruela-Arispe
- From the Department of Human Genetics (H.H.), Department of Molecular, Cell and Developmental Biology (J.J.M., C.M.W., R.D.F., A.I.M., S.Z., M.L.I.-A.), Molecular Biology Institute (M.L.I.-A.), and Jonsson Comprehensive Cancer Center (M.L.I.-A.), University of California, Los Angeles; Institute for Bioengineering (IBI) (E.G., M.A.S.) and The Swiss Institute for Experimental Cancer Research (ISREC) (M.L.S., C.B., M.A.S., M.D.P., M.L.I.-A.), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Switzerland; and Institute for Molecular Engineering and Ben May Department of Cancer Research, University of Chicago, IL (W.W.K., M.A.S.).
| |
Collapse
|
435
|
Cui W, Li Y, Xu K, Chen G, Lu X, Duan Q, Kang Z. miR-361-5p inhibits hepatocellular carcinoma cell proliferation and invasion by targeting VEGFA. Biochem Biophys Res Commun 2016; 479:901-906. [PMID: 27641667 DOI: 10.1016/j.bbrc.2016.09.076] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 09/15/2016] [Indexed: 12/18/2022]
Abstract
MicroRNAs (miRNAs) play important roles in the pathogenesis of many types of cancers by negatively regulating gene expression at posttranscriptional level. Here, we found that miR-361-5p is down-regulated in 135 patients with HCV-related hepatocellular carcinoma (HCC). Moreover, the expressions of miR-361-5p were highly correlated with VEGFA in these HCC patients. Further, CCK-8 proliferation assay indicated that miR-361-5p mimics inhibited the cell proliferation of HepG2 and SNU-398 HCC cells. Transwell assay showed that miR-361-5p mimics inhibited the invasion and migration of HepG2 and SNU-398 HCC cells. Luciferase assays revealed that miR-361-5p directly bound to the 3'untranslated region of VEGFA, and western blotting showed that miR-361-5p inhibited the expression of VEGFA. Generally, this study indicated that miR-361-5p is down-regulated in HCC and inhibits proliferation and invasion of HCC cell lines via VEGFA. In future, miR-361-5p will be a potential therapeutic agent for HCC.
Collapse
Affiliation(s)
- Wenxian Cui
- Clinical Laboratory, Changzhou No.7 People's Hospital, Changzhou, China
| | - Yuanguo Li
- Clinical Laboratory, Changzhou No.7 People's Hospital, Changzhou, China
| | - Keqing Xu
- Clinical Laboratory, Changzhou No.7 People's Hospital, Changzhou, China
| | - Guijin Chen
- Clinical Laboratory, Changzhou No.7 People's Hospital, Changzhou, China
| | - Xiaoqun Lu
- Clinical Laboratory, Changzhou No.7 People's Hospital, Changzhou, China
| | - Qiuqin Duan
- Clinical Laboratory, Changzhou No.7 People's Hospital, Changzhou, China
| | - Zhirong Kang
- Clinical Laboratory, Changzhou No.7 People's Hospital, Changzhou, China
| |
Collapse
|
436
|
Wu J, Yuan P, Mao Q, Lu P, Xie T, Yang H, Wang C. RETRACTED: miR-613 inhibits proliferation and invasion of breast cancer cell via VEGFA. Biochem Biophys Res Commun 2016; 478:274-278. [DOI: 10.1016/j.bbrc.2016.07.031] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 06/29/2016] [Accepted: 07/06/2016] [Indexed: 12/23/2022]
|
437
|
Chen MM, Xiao X, Lao XM, Wei Y, Liu RX, Zeng QH, Wang JC, Ouyang FZ, Chen DP, Chan KW, Shi DC, Zheng L, Kuang DM. Polarization of Tissue-Resident TFH-Like Cells in Human Hepatoma Bridges Innate Monocyte Inflammation and M2b Macrophage Polarization. Cancer Discov 2016; 6:1182-1195. [PMID: 27531854 DOI: 10.1158/2159-8290.cd-16-0329] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 08/09/2016] [Indexed: 11/16/2022]
Abstract
The existence, regulation, and functions of IL21+ immune cells are poorly defined in human cancers. Here, we identified a subset of protumorigenic IL21+ TFH-like cells in human hepatocellular carcinoma. These cells were the major source of IL21 in tumors and represented about 10% of the CD4+ T-cell population at levels comparable with the TFH cells present in lymph nodes. However, these TFH-like cells displayed a unique CXCR5-PD-1lo/-BTLA-CD69hi tissue-resident phenotype with substantial IFNγ production, which differed from the phenotype of TFH cells. Toll-like receptor 4 (TLR4)-elicited innate monocyte inflammation was important for IL21+ TFH-like cell induction in tumors, and activation of STAT1 and STAT3 was critical for TFH-like cell polarization in this process. Importantly, the TFH-like cells operated in IL21-IFNγ-dependent pathways to induce plasma cell differentiation and thereby create conditions for protumorigenic M2b macrophage polarization and cancer progression. Thus, induction of TFH-like cells links innate inflammation to immune privilege in tumors. SIGNIFICANCE We identified a novel protumorigenic IL21+ TFH-like cell subset with a CXCR5-PD-1- BTLA-CD69hi tissue-resident phenotype in hepatoma. TLR4-mediated monocyte inflammation and subsequent T-cell STAT1 and STAT3 activation are critical for TFH-like cell induction. TFH-like cells operate via IL21-IFNγ pathways to induce plasma cells and create conditions for M2b macrophage polarization. Cancer Discov; 6(10); 1182-95. ©2016 AACR.This article is highlighted in the In This Issue feature, p. 1069.
Collapse
Affiliation(s)
- Min-Min Chen
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China. State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiao Xiao
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xiang-Ming Lao
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yuan Wei
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Rui-Xian Liu
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Qiu-Hui Zeng
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jun-Cheng Wang
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Fang-Zhu Ouyang
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Dong-Ping Chen
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Ka-Wo Chan
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Dai-Chao Shi
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Limin Zheng
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Dong-Ming Kuang
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
438
|
Lund AW, Wagner M, Fankhauser M, Steinskog ES, Broggi MA, Spranger S, Gajewski TF, Alitalo K, Eikesdal HP, Wiig H, Swartz MA. Lymphatic vessels regulate immune microenvironments in human and murine melanoma. J Clin Invest 2016; 126:3389-402. [PMID: 27525437 DOI: 10.1172/jci79434] [Citation(s) in RCA: 151] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 07/07/2016] [Indexed: 12/28/2022] Open
Abstract
Lymphatic remodeling in tumor microenvironments correlates with progression and metastasis, and local lymphatic vessels play complex and poorly understood roles in tumor immunity. Tumor lymphangiogenesis is associated with increased immune suppression, yet lymphatic vessels are required for fluid drainage and immune cell trafficking to lymph nodes, where adaptive immune responses are mounted. Here, we examined the contribution of lymphatic drainage to tumor inflammation and immunity using a mouse model that lacks dermal lymphatic vessels (K14-VEGFR3-Ig mice). Melanomas implanted in these mice grew robustly, but exhibited drastically reduced cytokine expression and leukocyte infiltration compared with those implanted in control animals. In the absence of local immune suppression, transferred cytotoxic T cells more effectively controlled tumors in K14-VEGFR3-Ig mice than in control mice. Furthermore, gene expression analysis of human melanoma samples revealed that patient immune parameters are markedly stratified by levels of lymphatic markers. This work suggests that the establishment of tumor-associated inflammation and immunity critically depends on lymphatic vessel remodeling and drainage. Moreover, these results have implications for immunotherapies, the efficacies of which are regulated by the tumor immune microenvironment.
Collapse
|
439
|
Yamauchi F, Kamioka Y, Yano T, Matsuda M. In Vivo FRET Imaging of Tumor Endothelial Cells Highlights a Role of Low PKA Activity in Vascular Hyperpermeability. Cancer Res 2016; 76:5266-76. [PMID: 27488524 DOI: 10.1158/0008-5472.can-15-3534] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2015] [Accepted: 07/04/2016] [Indexed: 11/16/2022]
Abstract
Vascular hyperpermeability is a pathological hallmark of cancer. Previous in vitro studies have elucidated roles of various signaling molecules in vascular hyperpermeability; however, the activities of such signaling molecules have not been examined in live tumor tissues for technical reasons. Here, by in vivo two-photon excitation microscopy with transgenic mice expressing biosensors based on Förster resonance energy transfer, we examined the activity of protein kinase A (PKA), which maintains endothelial barrier function. The level of PKA activity was significantly lower in the intratumoral endothelial cells than the subcutaneous endothelial cells. PKA activation with a cAMP analogue alleviated the tumor vascular hyperpermeability, suggesting that the low PKA activity in the endothelial cells may be responsible for the tumor-tissue hyperpermeability. Because the vascular endothelial growth factor (VEGF) receptor is a canonical inducer of vascular hyperpermeability and a molecular target of anticancer drugs, we examined the causality between VEGF receptor activity and the PKA activity. Motesanib, a kinase inhibitor for VEGF receptor, activated tumor endothelial PKA and reduced the vascular permeability in the tumor. Conversely, subcutaneous injection of VEGF decreased endothelial PKA activity and induced hyperpermeability of subcutaneous blood vessels. Notably, in cultured human umbilical vascular endothelial cells, VEGF activated PKA rather than decreasing its activity, highlighting the remarkable difference between its actions in vitro and in vivo These data suggested that the VEGF receptor signaling pathway increases vascular permeability, at least in part, by reducing endothelial PKA activity in the live tumor tissue. Cancer Res; 76(18); 5266-76. ©2016 AACR.
Collapse
Affiliation(s)
- Fumio Yamauchi
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan. Medical Imaging System Development Center, R&D Headquarters, Canon Inc., Japan
| | - Yuji Kamioka
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan. Innovative Techno-Hub for Integrated Medical Bio-Imaging, Kyoto University, Kyoto, Japan
| | - Tetsuya Yano
- Medical Imaging System Development Center, R&D Headquarters, Canon Inc., Japan
| | - Michiyuki Matsuda
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| |
Collapse
|
440
|
Lewis CE, Harney AS, Pollard JW. The Multifaceted Role of Perivascular Macrophages in Tumors. Cancer Cell 2016; 30:18-25. [PMID: 27411586 PMCID: PMC5024543 DOI: 10.1016/j.ccell.2016.05.017] [Citation(s) in RCA: 164] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 05/18/2016] [Accepted: 05/28/2016] [Indexed: 12/12/2022]
Abstract
Evidence has emerged for macrophages in the perivascular niche of tumors regulating important processes like angiogenesis, various steps in the metastatic cascade, the recruitment and activity of other tumor-promoting leukocytes, and tumor responses to frontline therapies like irradiation and chemotherapy. Understanding the mechanisms controlling the recruitment, retention, and function of these cells could identify important targets for anti-cancer therapeutics.
Collapse
Affiliation(s)
- Claire E Lewis
- Department of Oncology and Metabolism, University of Sheffield Medical School, Sheffield S10 2RX, UK.
| | - Allison S Harney
- Departments of Anatomy and Structural Biology, Developmental and Molecular Biology, Albert Einstein College of Medicine, New York 10461, USA
| | - Jeffrey W Pollard
- Departments of Anatomy and Structural Biology, Developmental and Molecular Biology, Albert Einstein College of Medicine, New York 10461, USA; MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
441
|
Abstract
Recent clinical trials have demonstrated the ability to durably control cancer in some patients by manipulating T lymphocytes. These immunotherapies are revolutionizing cancer treatment but benefit only a minority of patients. It is thus a crucial time for clinicians, cancer scientists and immunologists to determine the next steps in shifting cancer treatment towards better cancer control. This Review describes recent advances in our understanding of tumour-associated myeloid cells. These cells remain less studied than T lymphocytes but have attracted particular attention because their presence in tumours is often linked to altered patient survival. Also, experimental studies indicate that myeloid cells modulate key cancer-associated activities, including immune evasion, and affect virtually all types of cancer therapy. Consequently, targeting myeloid cells could overcome limitations of current treatment options.
Collapse
Affiliation(s)
- Camilla Engblom
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, Massachusetts 02114, USA
- Graduate Program in Immunology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Christina Pfirschke
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Mikael J Pittet
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, Massachusetts 02114, USA
| |
Collapse
|
442
|
Diepenbruck M, Christofori G. Epithelial-mesenchymal transition (EMT) and metastasis: yes, no, maybe? Curr Opin Cell Biol 2016; 43:7-13. [PMID: 27371787 DOI: 10.1016/j.ceb.2016.06.002] [Citation(s) in RCA: 369] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 06/06/2016] [Accepted: 06/08/2016] [Indexed: 12/17/2022]
Abstract
An epithelial to mesenchymal transition (EMT) is a process of cell remodeling critical during embryonic development and organogenesis. During an EMT, epithelial cells lose their polarized organization and acquire migratory and invasive capabilities. While a plethora of experimental results have indicated that manipulating an EMT also affects cancer metastasis, its reverse process, a mesenchymal to epithelial transition (MET), seems to support metastatic outgrowth in distant organs. Moreover, recent reports investigating cancer cells circulating in the blood stream or employing genetic lineage-tracing have questioned a critical role of an EMT in metastasis formation. Hence, we need to better understand the molecular networks underlying the cell plasticity conferred by an EMT or a MET and its functional contribution to malignant tumor progression.
Collapse
Affiliation(s)
- Maren Diepenbruck
- Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| | | |
Collapse
|
443
|
de Mingo Pulido A, Ruffell B. Immune Regulation of the Metastatic Process: Implications for Therapy. Adv Cancer Res 2016; 132:139-63. [PMID: 27613132 DOI: 10.1016/bs.acr.2016.05.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Metastatic disease is the major cause of fatalities in cancer patients, but few therapies are designed to target the metastatic process. Cancer cells must perform a number of steps to successfully establish metastatic foci, including local invasion, intravasation, survival, extravasation, and growth in ectopic tissue. Due to the nonrandom distribution of metastasis, it has long been recognized that the tissue microenvironment must be an important determinant of colonization. More recently it has been established in animal models that immune cells regulate the metastatic process, including a dominant role for monocytes and macrophages, and emerging roles for neutrophils and various lymphocyte populations. While most research has focused on the early dissemination process, patients usually present clinically with disseminated, if not macroscopic, disease. Identifying pathways by which immune cells regulate growth and therapeutic resistance within metastatic sites is therefore key to the development of pharmacological agents that will significantly extend patient survival.
Collapse
Affiliation(s)
- A de Mingo Pulido
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - B Ruffell
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States.
| |
Collapse
|
444
|
Harney AS, Wang Y, Condeelis JS, Entenberg D. Extended Time-lapse Intravital Imaging of Real-time Multicellular Dynamics in the Tumor Microenvironment. J Vis Exp 2016. [PMID: 27341448 DOI: 10.3791/54042] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
In the tumor microenvironment, host stromal cells interact with tumor cells to promote tumor progression, angiogenesis, tumor cell dissemination and metastasis. Multicellular interactions in the tumor microenvironment can lead to transient events including directional tumor cell motility and vascular permeability. Quantification of tumor vascular permeability has frequently used end-point experiments to measure extravasation of vascular dyes. However, due to the transient nature of multicellular interactions and vascular permeability, the kinetics of these dynamic events cannot be discerned. By labeling cells and vasculature with injectable dyes or fluorescent proteins, high-resolution time-lapse intravital microscopy has allowed the direct, real-time visualization of transient events in the tumor microenvironment. Here we describe a method for using multiphoton microscopy to perform extended intravital imaging in live mice to directly visualize multicellular dynamics in the tumor microenvironment. This method details cellular labeling strategies, the surgical preparation of a mammary skin flap, the administration of injectable dyes or proteins by tail vein catheter and the acquisition of time-lapse images. The time-lapse sequences obtained from this method facilitate the visualization and quantitation of the kinetics of cellular events of motility and vascular permeability in the tumor microenvironment.
Collapse
Affiliation(s)
- Allison S Harney
- Department of Anatomy & Structural Biology, Albert Einstein College of Medicine; Department of Radiology, Albert Einstein College of Medicine; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine; Integrated Imaging Program, Albert Einstein College of Medicine;
| | - Yarong Wang
- Department of Anatomy & Structural Biology, Albert Einstein College of Medicine; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine
| | - John S Condeelis
- Department of Anatomy & Structural Biology, Albert Einstein College of Medicine; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine; Integrated Imaging Program, Albert Einstein College of Medicine
| | - David Entenberg
- Department of Anatomy & Structural Biology, Albert Einstein College of Medicine; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine; Integrated Imaging Program, Albert Einstein College of Medicine
| |
Collapse
|
445
|
In Vivo Visualization of Stromal Macrophages via label-free FLIM-based metabolite imaging. Sci Rep 2016; 6:25086. [PMID: 27220760 PMCID: PMC4879594 DOI: 10.1038/srep25086] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 04/07/2016] [Indexed: 01/02/2023] Open
Abstract
Macrophage infiltration and recruitment in breast tumors has been correlated with poor prognosis in breast cancer patients and has been linked to tumor cell dissemination. Much of our understanding comes from animal models in which macrophages are labeled by expression of an extrinsic fluorophore. However, conventional extrinsic fluorescence labeling approaches are not readily applied to human tissue and clinical use. We report a novel strategy that exploits endogenous fluorescence from the metabolic co-factors NADH and FAD with quantitation from Fluorescence Lifetime Imaging Microscopy (FLIM) as a means to non-invasively identify tumor-associated macrophages in the intact mammary tumor microenvironment. Macrophages were FADHI and demonstrated a glycolytic-like NADH-FLIM signature that was readily separated from the intrinsic fluorescence signature of tumor cells. This non-invasive quantitative technique provides a unique ability to discern specific cell types based upon their metabolic signatures without the use of exogenous fluorescent labels. Not only does this provide high resolution temporal and spatial views of macrophages in live animal breast cancer models, this approach can be extended to other animal disease models where macrophages are implicated and has potential for clinical applications.
Collapse
|
446
|
Wang Y, Wang H, Li J, Entenberg D, Xue A, Wang W, Condeelis J. Direct visualization of the phenotype of hypoxic tumor cells at single cell resolution in vivo using a new hypoxia probe. INTRAVITAL 2016; 5. [PMID: 27790387 DOI: 10.1080/21659087.2016.1187803] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tumor hypoxia is linked to tumor progression, metastasis, and therapy resistance. However, the underlying mechanisms behind this linkage are not fully understood. Here we present a novel fluorescent mCherry hypoxia-responsive marker that can be used in real time imaging to specifically and sensitively identify hypoxic cells in vivo at single cell resolution. Tumors derived from triple negative tumor cells expressing the hypoxia marker reveal that the hypoxic tumor cells congregate near flowing blood vessels. Using multiphoton microscopy, hypoxic MDA-MB-231 cells were directly visualized and showed a more persistent slow migration phenotype as compared to normoxic cells in the same field in vivo. Hypoxic tumor cells are enriched in the cell population that migrates toward human epithelial growth factor gradients in vivo, and has increased collagen degradation and intravasation activity, characteristics of dissemination and metastasis competent tumor cells. The hypoxia probe introduced in this study provides a specific reporter of hypoxic cell phenotypes in vivo which reveals new insights into the mechanisms by which hypoxia is linked to metastasis.
Collapse
Affiliation(s)
- Yarong Wang
- Department of Anatomy and Structural Biology; Albert Einstein College of Medicine; Bronx, NY USA; Integrated Imaging Program; Albert Einstein College of Medicine,Bronx, New York, USA
| | - Haoxuan Wang
- Department of Anatomy and Structural Biology; Albert Einstein College of Medicine; Bronx, NY USA
| | - Jiufeng Li
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - David Entenberg
- Department of Anatomy and Structural Biology; Albert Einstein College of Medicine; Bronx, NY USA; Gruss Lipper Biophotonics Center; Albert Einstein College of Medicine; Bronx, NY USA; Integrated Imaging Program; Albert Einstein College of Medicine,Bronx, New York, USA
| | - Alice Xue
- Department of Anatomy and Structural Biology; Albert Einstein College of Medicine; Bronx, NY USA
| | - Weigang Wang
- Department of Anatomy and Structural Biology; Albert Einstein College of Medicine; Bronx, NY USA
| | - John Condeelis
- Department of Anatomy and Structural Biology; Albert Einstein College of Medicine; Bronx, NY USA; Gruss Lipper Biophotonics Center; Albert Einstein College of Medicine; Bronx, NY USA; Integrated Imaging Program; Albert Einstein College of Medicine,Bronx, New York, USA
| |
Collapse
|
447
|
Vennin C, Herrmann D, Lucas MC, Timpson P. Intravital imaging reveals new ancillary mechanisms co-opted by cancer cells to drive tumor progression. F1000Res 2016; 5. [PMID: 27239290 PMCID: PMC4870995 DOI: 10.12688/f1000research.8090.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/11/2016] [Indexed: 12/15/2022] Open
Abstract
Intravital imaging is providing new insights into the dynamics of tumor progression in native tissues and has started to reveal the layers of complexity found in cancer. Recent advances in intravital imaging have allowed us to look deeper into cancer behavior and to dissect the interactions between tumor cells and the ancillary host niche that promote cancer development. In this review, we provide an insight into the latest advances in cancer biology achieved by intravital imaging, focusing on recently discovered mechanisms by which tumor cells manipulate normal tissue to facilitate disease progression.
Collapse
Affiliation(s)
- Claire Vennin
- The Kinghorn Cancer Centre, Cancer Division, The Garvan Institute of Medical Research, Sydney, NSW, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - David Herrmann
- The Kinghorn Cancer Centre, Cancer Division, The Garvan Institute of Medical Research, Sydney, NSW, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Morghan C Lucas
- The Kinghorn Cancer Centre, Cancer Division, The Garvan Institute of Medical Research, Sydney, NSW, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Paul Timpson
- The Kinghorn Cancer Centre, Cancer Division, The Garvan Institute of Medical Research, Sydney, NSW, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
448
|
Williams JK, Entenberg D, Wang Y, Avivar-Valderas A, Padgen M, Clark A, Aguirre-Ghiso JA, Castracane J, Condeelis JS. Validation of a device for the active manipulation of the tumor microenvironment during intravital imaging. INTRAVITAL 2016; 5. [PMID: 27790386 DOI: 10.1080/21659087.2016.1182271] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The tumor microenvironment is recognized as playing a significant role in the behavior of tumor cells and their progression to metastasis. However, tools to manipulate the tumor microenvironment directly, and image the consequences of this manipulation with single cell resolution in real time in vivo, are lacking. We describe here a method for the direct, local manipulation of microenvironmental parameters through the use of an implantable Induction Nano Intravital Device (iNANIVID) and simultaneous in vivo visualization of the results at single-cell resolution. As a proof of concept, we deliver both a sustained dose of EGF to tumor cells while intravital imaging their chemotactic response as well as locally induce hypoxia in defined microenvironments in solid tumors.
Collapse
Affiliation(s)
- James K Williams
- Colleges of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY, USA
| | - David Entenberg
- Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Yeshiva University, Bronx, NY, USA; Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Yeshiva University, Bronx, NY, USA; Integrated Imaging Program, Albert Einstein College of Medicine, Yeshiva University, Bronx, NY, USA
| | - Yarong Wang
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Yeshiva University, Bronx, NY, USA; Integrated Imaging Program, Albert Einstein College of Medicine, Yeshiva University, Bronx, NY, USA
| | - Alvaro Avivar-Valderas
- Department of Medicine and Department Otolaryngology, Tisch Cancer Institute, Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, NY, USA
| | - Michael Padgen
- Colleges of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY, USA
| | - Ashley Clark
- Colleges of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY, USA
| | - Julio A Aguirre-Ghiso
- Department of Medicine and Department Otolaryngology, Tisch Cancer Institute, Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, NY, USA
| | - James Castracane
- Colleges of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY, USA
| | - John S Condeelis
- Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Yeshiva University, Bronx, NY, USA; Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Yeshiva University, Bronx, NY, USA; Integrated Imaging Program, Albert Einstein College of Medicine, Yeshiva University, Bronx, NY, USA
| |
Collapse
|
449
|
Rodriguez-Hernandez I, Cantelli G, Bruce F, Sanz-Moreno V. Rho, ROCK and actomyosin contractility in metastasis as drug targets. F1000Res 2016; 5. [PMID: 27158478 PMCID: PMC4856114 DOI: 10.12688/f1000research.7909.1] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/26/2016] [Indexed: 12/17/2022] Open
Abstract
Metastasis is the spread of cancer cells around the body and the cause of the majority of cancer deaths. Metastasis is a very complex process in which cancer cells need to dramatically modify their cytoskeleton and cope with different environments to successfully colonize a secondary organ. In this review, we discuss recent findings pointing at Rho-ROCK or actomyosin force (or both) as major drivers of many of the steps required for metastatic success. We propose that these are important drug targets that need to be considered in the clinic to palliate metastatic disease.
Collapse
Affiliation(s)
- Irene Rodriguez-Hernandez
- Tumour Plasticity Laboratory, Randall Division of Cell and Molecular Biophysics, Guy's Campus, King's College London, London, SE1 1UL, UK
| | - Gaia Cantelli
- Tumour Plasticity Laboratory, Randall Division of Cell and Molecular Biophysics, Guy's Campus, King's College London, London, SE1 1UL, UK
| | - Fanshawe Bruce
- Tumour Plasticity Laboratory, Randall Division of Cell and Molecular Biophysics, Guy's Campus, King's College London, London, SE1 1UL, UK.,Department of Imaging Chemistry and Biology, Division of Imaging Sciences and Biomedical Engineering, St. Thomas Hospital, King's College London, London, SE1 7EH, UK
| | - Victoria Sanz-Moreno
- Tumour Plasticity Laboratory, Randall Division of Cell and Molecular Biophysics, Guy's Campus, King's College London, London, SE1 1UL, UK
| |
Collapse
|
450
|
Zhang Y, Li JQ, Jiang ZZ, Li L, Wu Y, Zheng L. CD169 identifies an anti-tumour macrophage subpopulation in human hepatocellular carcinoma. J Pathol 2016; 239:231-41. [DOI: 10.1002/path.4720] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 02/21/2016] [Accepted: 03/15/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Yi Zhang
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences; Sun Yat-sen University; Guangzhou PR China
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China; Sun Yat-sen University Cancer Center, Sun Yat-sen University; Guangzhou PR China
| | - Jin-Qing Li
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China; Sun Yat-sen University Cancer Center, Sun Yat-sen University; Guangzhou PR China
| | - Ze-Zhou Jiang
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences; Sun Yat-sen University; Guangzhou PR China
| | - Lian Li
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences; Sun Yat-sen University; Guangzhou PR China
| | - Yan Wu
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences; Sun Yat-sen University; Guangzhou PR China
| | - Limin Zheng
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences; Sun Yat-sen University; Guangzhou PR China
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China; Sun Yat-sen University Cancer Center, Sun Yat-sen University; Guangzhou PR China
| |
Collapse
|