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

      VEGFR2 activity on myeloid cells mediates immune suppression in the tumor microenvironment

      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

          Angiogenesis, a hallmark of cancer, is induced by vascular endothelial growth factor–A (hereafter VEGF). As a result, anti-VEGF therapy is commonly used for cancer treatment. Recent studies have found that VEGF expression is also associated with immune suppression in patients with cancer. This connection has been investigated in preclinical and clinical studies by evaluating the therapeutic effect of combining antiangiogenic reagents with immune therapy. However, the mechanisms of how anti-VEGF strategies enhance immune therapy are not fully understood. We and others have shown selective elevation of VEGFR2 expression on tumor-associated myeloid cells in tumor-bearing animals. Here, we investigated the function of VEGFR2 + myeloid cells in regulating tumor immunity and found VEGF induced an immunosuppressive phenotype in VEGFR2 + myeloid cells, including directly upregulating the expression of programmed cell death 1 ligand 1. Moreover, we found that VEGF blockade inhibited the immunosuppressive phenotype of VEGFR2 + myeloid cells, increased T cell activation, and enhanced the efficacy of immune checkpoint blockade. This study highlights the function of VEGFR2 on myeloid cells and provides mechanistic insight on how VEGF inhibition potentiates immune checkpoint blockade.

          Related collections

          Most cited references50

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

          The blockade of immune checkpoints in cancer immunotherapy.

          Among the most promising approaches to activating therapeutic antitumour immunity is the blockade of immune checkpoints. Immune checkpoints refer to a plethora of inhibitory pathways hardwired into the immune system that are crucial for maintaining self-tolerance and modulating the duration and amplitude of physiological immune responses in peripheral tissues in order to minimize collateral tissue damage. It is now clear that tumours co-opt certain immune-checkpoint pathways as a major mechanism of immune resistance, particularly against T cells that are specific for tumour antigens. Because many of the immune checkpoints are initiated by ligand-receptor interactions, they can be readily blocked by antibodies or modulated by recombinant forms of ligands or receptors. Cytotoxic T-lymphocyte-associated antigen 4 (CTLA4) antibodies were the first of this class of immunotherapeutics to achieve US Food and Drug Administration (FDA) approval. Preliminary clinical findings with blockers of additional immune-checkpoint proteins, such as programmed cell death protein 1 (PD1), indicate broad and diverse opportunities to enhance antitumour immunity with the potential to produce durable clinical responses.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Atezolizumab plus Bevacizumab in Unresectable Hepatocellular Carcinoma

            The combination of atezolizumab and bevacizumab showed encouraging antitumor activity and safety in a phase 1b trial involving patients with unresectable hepatocellular carcinoma.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Molecular mechanisms and clinical applications of angiogenesis.

              Blood vessels deliver oxygen and nutrients to every part of the body, but also nourish diseases such as cancer. Over the past decade, our understanding of the molecular mechanisms of angiogenesis (blood vessel growth) has increased at an explosive rate and has led to the approval of anti-angiogenic drugs for cancer and eye diseases. So far, hundreds of thousands of patients have benefited from blockers of the angiogenic protein vascular endothelial growth factor, but limited efficacy and resistance remain outstanding problems. Recent preclinical and clinical studies have shown new molecular targets and principles, which may provide avenues for improving the therapeutic benefit from anti-angiogenic strategies.
                Bookmark

                Author and article information

                Contributors
                Journal
                JCI Insight
                JCI Insight
                JCI Insight
                JCI Insight
                American Society for Clinical Investigation
                2379-3708
                8 December 2021
                8 December 2021
                8 December 2021
                : 6
                : 23
                : e150735
                Affiliations
                [1 ]Hamon Center for Therapeutic Oncology Research,
                [2 ]Department of Surgery,
                [3 ]Cancer Biology Graduate Program, and
                [4 ]Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Texas (UT) Southwestern, Dallas, Texas, USA.
                [5 ]Program of Mathematics in Medicine, Houston Methodist Research Institute, Houston, Texas, USA.
                [6 ]Department of Pathology and
                [7 ]Division of Hematology and Oncology, Department of Medicine, UT Southwestern, Dallas, Texas, USA.
                Author notes
                Address correspondence to: Rolf A. Brekken, Hamon Center for Therapeutic Oncology Research, UT Southwestern, 6000 Harry Hines Blvd., Dallas, Texas 75390-8593, USA. Phone: 214.648.5151; Email: rolf.brekken@ 123456utsouthwestern.edu .
                Author information
                http://orcid.org/0000-0003-0740-5310
                http://orcid.org/0000-0002-6345-4566
                http://orcid.org/0000-0002-3367-367X
                http://orcid.org/0000-0003-2704-2377
                Article
                150735
                10.1172/jci.insight.150735
                8675197
                34673569
                4d23807c-2662-41a6-9961-1920eef561f6
                © 2021 Zhang et al.

                This work is licensed under the Creative Commons Attribution 4.0 International License. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

                History
                : 21 April 2021
                : 20 October 2021
                Funding
                Funded by: OncXerna (formerly Oncologie); NIH; Effie Marie Cain Fellowship
                Award ID: U54 CA210181 (Project 2),R01 CA243577
                Categories
                Research Article

                immunology,oncology,cancer immunotherapy,macrophages
                immunology, oncology, cancer immunotherapy, macrophages

                Comments

                Comment on this article