8
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Optimized antiangiogenic reprogramming of the tumor microenvironment potentiates CD40 immunotherapy

      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.

          Significance

          Cancer immunotherapy demonstrates clinical efficacy in selected cancer types. Yet, as the majority of patients do not respond to the most effective immunotherapeutics, novel immunotherapy combinations are extensively investigated. There is increasing clinical interest in combining cancer immunotherapies with antiangiogenic agents, particularly VEGFA pathway inhibitors. Here we show that anti-CD40 immunotherapy increases CD8 + T cell infiltration in the tumor, yet tumor regression is achieved more robustly when anti-CD40 is combined with dual Ang2 and VEGFA blockade, but not VEGFA inhibition alone. Tumor regression was associated with proinflammatory skewing of the tumor microenvironment and intratumoral redistribution of CD8 + T cells. These data emphasize the rationale for blocking Ang2 as a vascular-modulatory strategy in combination with T cell-targeting immunotherapies.

          Abstract

          Cancer immunotherapies are increasingly combined with targeted therapies to improve therapeutic outcomes. We show that combination of agonistic anti-CD40 with antiangiogenic antibodies targeting 2 proangiogenic factors, vascular endothelial growth factor A (VEGFA) and angiopoietin 2 (Ang2/ANGPT2), induces pleiotropic immune mechanisms that facilitate tumor rejection in several tumor models. On the one hand, VEGFA/Ang2 blockade induced regression of the tumor microvasculature while decreasing the proportion of nonperfused vessels and reducing leakiness of the remaining vessels. On the other hand, both anti-VEGFA/Ang2 and anti-CD40 independently promoted proinflammatory macrophage skewing and increased dendritic cell activation in the tumor microenvironment, which were further amplified upon combination of the 2 treatments. Finally, combined therapy provoked brisk infiltration and intratumoral redistribution of cytotoxic CD8 + T cells in the tumors, which was mainly driven by Ang2 blockade. Overall, these nonredundant synergistic mechanisms endowed T cells with improved effector functions that were conducive to more efficient tumor control, underscoring the therapeutic potential of antiangiogenic immunotherapy in cancer.

          Related collections

          Most cited references36

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

          Combined antiangiogenic and anti–PD-L1 therapy stimulates tumor immunity through HEV formation

          Inhibitors of VEGF (vascular endothelial growth factor)/VEGFR2 (vascular endothelial growth factor receptor 2) are commonly used in the clinic, but their beneficial effects are only observed in a subset of patients and limited by induction of diverse relapse mechanisms. We describe the up-regulation of an adaptive immunosuppressive pathway during antiangiogenic therapy, by which PD-L1 (programmed cell death ligand 1), the ligand of the negative immune checkpoint regulator PD-1 (programmed cell death protein 1), is enhanced by interferon-γ-expressing T cells in distinct intratumoral cell types in refractory pancreatic, breast, and brain tumor mouse models. Successful treatment with a combination of anti-VEGFR2 and anti-PD-L1 antibodies induced high endothelial venules (HEVs) in PyMT (polyoma middle T oncoprotein) breast cancer and RT2-PNET (Rip1-Tag2 pancreatic neuroendocrine tumors), but not in glioblastoma (GBM). These HEVs promoted lymphocyte infiltration and activity through activation of lymphotoxin β receptor (LTβR) signaling. Further activation of LTβR signaling in tumor vessels using an agonistic antibody enhanced HEV formation, immunity, and subsequent apoptosis and necrosis in pancreatic and mammary tumors. Finally, LTβR agonists induced HEVs in recalcitrant GBM, enhanced cytotoxic T cell (CTL) activity, and thereby sensitized tumors to antiangiogenic/anti-PD-L1 therapy. Together, our preclinical studies provide evidence that anti-PD-L1 therapy can sensitize tumors to antiangiogenic therapy and prolong its efficacy, and conversely, antiangiogenic therapy can improve anti-PD-L1 treatment specifically when it generates intratumoral HEVs that facilitate enhanced CTL infiltration, activity, and tumor cell destruction.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Targeting the tumour vasculature: insights from physiological angiogenesis.

            The cardiovascular system ensures the delivery of nutrients, oxygen, and blood and immune cells to all organs and tissues: it is also responsible for the removal of waste metabolites. The vascular system develops and matures through two tightly regulated processes: vasculogenesis and angiogenesis. Angiogenesis is active only under specific physiological conditions in healthy adults but the vasculature can be aberrantly activated to generate new blood vessels during pathological conditions such as cancer and chronic inflammation. In this Opinion article we discuss the parallels and differences in the angiogenic process under either a physiological or a pathological state, especially tumorigenesis.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Dual angiopoietin-2 and VEGFA inhibition elicits antitumor immunity that is enhanced by PD-1 checkpoint blockade

              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.
                Bookmark

                Author and article information

                Journal
                Proc Natl Acad Sci U S A
                Proc. Natl. Acad. Sci. U.S.A
                pnas
                pnas
                PNAS
                Proceedings of the National Academy of Sciences of the United States of America
                National Academy of Sciences
                0027-8424
                1091-6490
                7 January 2020
                30 December 2019
                30 December 2019
                : 117
                : 1
                : 541-551
                Affiliations
                [1] aDepartment of Biomedicine, University Hospital Basel and University of Basel , 4031 Basel, Switzerland;
                [2] bSwiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland;
                [3] cPharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel , 4070 Basel, Switzerland;
                [4] dInstitute of Medical Genetics and Pathology, University Hospital Basel , 4031 Basel, Switzerland;
                [5] ePharma Research and Early Development, Roche Innovation Center Munich , 82377 Penzberg, Germany;
                [6] fPharma Research and Early Development, Roche Innovation Center Zurich , 8952 Schlieren, Switzerland;
                [7] gMedical Oncology, University Hospital Basel , 4031 Basel, Switzerland
                Author notes

                Edited by Napoleone Ferrara, University of California San Diego, La Jolla, CA, and approved November 15, 2019 (received for review February 6, 2019)

                Author contributions: A.S.K., M.S., P.M., F.H., Y.K., C.K., C.H.R., E.C., M.D.P., and A.Z. designed research; A.S.K., M.S., N. Rigamonti, D.P.-F., P.M., M.B., M.K., P.H., A.G., N. Rieder, R.B., F.H., and S.D. performed research; R.B., S.H., and C.H.R. contributed new reagents/analytic tools; A.S.K., M.S., N. Rigamonti, D.P.-F., P.M., C.-H.O., A.G., R.B., F.H., Y.K., S.D., C.K., S.H., and E.C. analyzed data; and A.S.K., M.S., A.G., E.C., M.D.P., and A.Z. wrote the paper.

                1A.S.K., M.S., M.D.P., and A.Z. contributed equally to this work.

                3Present address: Pharma Research and Early Development, Roche Innovation Center Munich, 82377 Penzberg, Germany.

                4Present address: Molecular Partners, 8952 Schlieren, Switzerland.

                5Present address: Boehringer Ingelheim Pharma GmbH & Co, 88397 Biberach an der Riss, Germany.

                6Present address: Centre for Systems Biology, Massachusetts General Hospital, Boston, MA 02114.

                Author information
                http://orcid.org/0000-0003-2856-373X
                http://orcid.org/0000-0001-6759-6433
                http://orcid.org/0000-0002-4953-7234
                Article
                201902145
                10.1073/pnas.1902145116
                6955310
                31889004
                992cd858-38ed-4f5e-86b6-12d234098cf8
                Copyright © 2020 the Author(s). Published by PNAS.

                This open access article is distributed under Creative Commons Attribution-NonCommercial-NoDerivatives License 4.0 (CC BY-NC-ND).

                History
                Page count
                Pages: 11
                Funding
                Funded by: Krebsliga Schweiz (Swiss Cancer League) 501100004361
                Award ID: KFS-3394-02-2014
                Award Recipient : Michele De Palma Award Recipient : Alfred Zippelius
                Funded by: Krebsliga Schweiz (Swiss Cancer League) 501100004361
                Award ID: KFS-3759-08-2015
                Award Recipient : Michele De Palma Award Recipient : Alfred Zippelius
                Categories
                PNAS Plus
                Biological Sciences
                Immunology and Inflammation
                PNAS Plus

                angiogenesis,immunotherapy,cd40,vegfa,angiopoetin
                angiogenesis, immunotherapy, cd40, vegfa, angiopoetin

                Comments

                Comment on this article