10
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Targeting PDGF‐mediated recruitment of pericytes blocks vascular mimicry and tumor growth

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Aggressive tumor cells can adopt an endothelial cell‐like phenotype and contribute to the formation of a tumor vasculature, independent of tumor angiogenesis. This adoptive mechanism is referred to as vascular mimicry and it is associated with poor survival in cancer patients. To what extent tumor cells capable of vascular mimicry phenocopy the angiogenic cascade is still poorly explored. Here, we identify pericytes as important players in vascular mimicry. We found that pericytes are recruited by vascular mimicry‐positive tumor cells in order to facilitate sprouting and to provide structural support of the vascular‐like networks. The pericyte recruitment is mediated through platelet‐derived growth factor (PDGF)‐B. Consequently, preventing PDGF‐B signaling by blocking the PDGF receptors with either the small tyrosine kinase inhibitor imatinib or blocking antibodies inhibits vascular mimicry and tumor growth. Collectively, the current study identifies an important role for pericytes in the formation of vascular‐like structures by tumor cells. Moreover, the mechanism that controls the pericyte recruitment provides therapeutic opportunities for patients with aggressive vascular mimicry‐positive cancer types. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.

          Related collections

          Most cited references50

          • Record: found
          • Abstract: found
          • Article: not found

          Vascular channel formation by human melanoma cells in vivo and in vitro: vasculogenic mimicry.

          Tissue sections from aggressive human intraocular (uveal) and metastatic cutaneous melanomas generally lack evidence of significant necrosis and contain patterned networks of interconnected loops of extracellular matrix. The matrix that forms these loops or networks may be solid or hollow. Red blood cells have been detected within the hollow channel components of this patterned matrix histologically, and these vascular channel networks have been detected in human tumors angiographically. Endothelial cells were not identified within these matrix-embedded channels by light microscopy, by transmission electron microscopy, or by using an immunohistochemical panel of endothelial cell markers (Factor VIII-related antigen, Ulex, CD31, CD34, and KDR[Flk-1]). Highly invasive primary and metastatic human melanoma cells formed patterned solid and hollow matrix channels (seen in tissue sections of aggressive primary and metastatic human melanomas) in three-dimensional cultures containing Matrigel or dilute Type I collagen, without endothelial cells or fibroblasts. These tumor cell-generated patterned channels conducted dye, highlighting looping patterns visualized angiographically in human tumors. Neither normal melanocytes nor poorly invasive melanoma cells generated these patterned channels in vitro under identical culture conditions, even after the addition of conditioned medium from metastatic pattern-forming melanoma cells, soluble growth factors, or regimes of hypoxia. Highly invasive and metastatic human melanoma cells, but not poorly invasive melanoma cells, contracted and remodeled floating hydrated gels, providing a biomechanical explanation for the generation of microvessels in vitro. cDNA microarray analysis of highly invasive versus poorly invasive melanoma tumor cells confirmed a genetic reversion to a pluripotent embryonic-like genotype in the highly aggressive melanoma cells. These observations strongly suggest that aggressive melanoma cells may generate vascular channels that facilitate tumor perfusion independent of tumor angiogenesis.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Accelerated metastasis after short-term treatment with a potent inhibitor of tumor angiogenesis.

            Herein we report that the VEGFR/PDGFR kinase inhibitor sunitinib/SU11248 can accelerate metastatic tumor growth and decrease overall survival in mice receiving short-term therapy in various metastasis assays, including after intravenous injection of tumor cells or after removal of primary orthotopically grown tumors. Acceleration of metastasis was also observed in mice receiving sunitinib prior to intravenous implantation of tumor cells, suggesting possible "metastatic conditioning" in multiple organs. Similar findings with additional VEGF receptor tyrosine kinase inhibitors implicate a class-specific effect for such agents. Importantly, these observations of metastatic acceleration were in contrast to the demonstrable antitumor benefits obtained when the same human breast cancer cells, as well as mouse or human melanoma cells, were grown orthotopically as primary tumors and subjected to identical sunitinib treatments.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Glioblastoma stem cells generate vascular pericytes to support vessel function and tumor growth.

              Glioblastomas (GBMs) are highly vascular and lethal brain tumors that display cellular hierarchies containing self-renewing tumorigenic glioma stem cells (GSCs). Because GSCs often reside in perivascular niches and may undergo mesenchymal differentiation, we interrogated GSC potential to generate vascular pericytes. Here, we show that GSCs give rise to pericytes to support vessel function and tumor growth. In vivo cell lineage tracing with constitutive and lineage-specific fluorescent reporters demonstrated that GSCs generate the majority of vascular pericytes. Selective elimination of GSC-derived pericytes disrupts the neovasculature and potently inhibits tumor growth. Analysis of human GBM specimens showed that most pericytes are derived from neoplastic cells. GSCs are recruited toward endothelial cells via the SDF-1/CXCR4 axis and are induced to become pericytes predominantly by transforming growth factor β. Thus, GSCs contribute to vascular pericytes that may actively remodel perivascular niches. Therapeutic targeting of GSC-derived pericytes may effectively block tumor progression and improve antiangiogenic therapy. Copyright © 2013 Elsevier Inc. All rights reserved.
                Bookmark

                Author and article information

                Contributors
                aw.griffioen@vumc.nl
                Journal
                J Pathol
                J. Pathol
                10.1002/(ISSN)1096-9896
                PATH
                The Journal of Pathology
                John Wiley & Sons, Ltd (Chichester, UK )
                0022-3417
                1096-9896
                30 October 2018
                December 2018
                : 246
                : 4 ( doiID: 10.1002/path.2018.246.issue-4 )
                : 447-458
                Affiliations
                [ 1 ] Angiogenesis Laboratory, Department of Medical Oncology VU University Medical Center Amsterdam The Netherlands
                [ 2 ] Department of Radiation Oncology VU University Medical Center Amsterdam The Netherlands
                [ 3 ] Division of Medical Oncology, GROW – School for Oncology and Developmental Biology Maastricht University Medical Center Maastricht The Netherlands
                [ 4 ] School of Pharmaceutical Sciences University of Geneva Geneva Switzerland
                [ 5 ] Department of Microbiology, Tumor and Cell Biology Karolinska Institute Stockholm Sweden
                [ 6 ] Department of Microscopic Morphology, Histology, Angiogenesis Research Center Victor Babes University of Medicine and Pharmacy Timisoara Romania
                [ 7 ] Department of Pathology Antwerp University Hospital Edegem Belgium
                [ 8 ] Laboratory of Translational Cell and Tissue Research University of Leuven Leuven Belgium
                [ 9 ] Department of Biology, Shepherd University Shepherdstown University WV USA
                [ 10 ] Department of Medical Biochemistry and Microbiology, Science for Life Laboratory Uppsala University Uppsala Sweden
                Author notes
                [*] [* ] Correspondence to: AW Griffioen, Department of Medical Oncology, Angiogenesis Laboratory, VU University Medical Center, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands. E‐mail: aw.griffioen@ 123456vumc.nl

                [†]

                These authors contributed equally to this work.

                Author information
                https://orcid.org/0000-0002-8508-4754
                Article
                PATH5152
                10.1002/path.5152
                6587443
                30101525
                14c5f364-5d31-41e6-b028-0fa58e7bfda4
                © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.

                This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

                History
                : 22 May 2018
                : 12 July 2018
                : 07 August 2018
                Page count
                Figures: 4, Tables: 0, Pages: 12, Words: 7800
                Funding
                Funded by: Dutch Cancer Society
                Award ID: VUMC2014‐7234
                Funded by: Profileringsfonds AZM
                Categories
                Original Paper
                Original Papers
                Custom metadata
                2.0
                path5152
                December 2018
                Converter:WILEY_ML3GV2_TO_NLMPMC version:5.6.4 mode:remove_FC converted:21.06.2019

                Pathology
                tumor angiogenesis,imatinib,endothelial cells,melanoma,ewing sarcoma,perivascular cells,vasculogenic mimicry,vessel stabilization,cancer

                Comments

                Comment on this article