35
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Macrophages impede CD8 T cells from reaching tumor cells and limit the efficacy of anti–PD-1 treatment

      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 is a promising therapeutic intervention. However, complete and durable responses are seen in only a fraction of cancer patients. A key factor that limits therapeutic success is the lack of T cells in tumor cell regions, a profile termed “immune-excluded.” Here, we provide evidence that tumor-associated macrophages (TAMs) are an important determinant of the establishment of a T cell-excluded tumor phenotype. In human and murine tumors, we found that CD8 T cells poorly migrate and invade tumor nests due to long-lasting interactions with TAMs. The depletion of TAMs restores T cell migration and infiltration into tumor islets and improves the efficacy of anti–PD-1 immunotherapy. This study highlights the rationale of combining approaches targeting TAM and immune checkpoint proteins.

          Abstract

          In a large proportion of cancer patients, CD8 T cells are excluded from the vicinity of cancer cells. The inability of CD8 T cells to reach tumor cells is considered an important mechanism of resistance to cancer immunotherapy. We show that, in human lung squamous-cell carcinomas, exclusion of CD8 T cells from tumor islets is correlated with a poor clinical outcome and with a low lymphocyte motility, as assessed by dynamic imaging on fresh tumor slices. In the tumor stroma, macrophages mediate lymphocyte trapping by forming long-lasting interactions with CD8 T cells. Using a mouse tumor model with well-defined stromal and tumor cell areas, macrophages were depleted with PLX3397, an inhibitor of colony-stimulating factor-1 receptor (CSF-1R). Our results reveal that a CSF-1R blockade enhances CD8 T cell migration and infiltration into tumor islets. Although this treatment alone has minor effects on tumor growth, its combination with anti–PD-1 therapy further increases the accumulation of CD8 T cells in close contact with malignant cells and delays tumor progression. These data suggest that the reduction of macrophage-mediated T cell exclusion increases tumor surveillance by CD8 T cells and renders tumors more responsive to anti–PD-1 treatment.

          Related collections

          Most cited references29

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

          Leukocyte complexity predicts breast cancer survival and functionally regulates response to chemotherapy.

          Immune-regulated pathways influence multiple aspects of cancer development. In this article we demonstrate that both macrophage abundance and T-cell abundance in breast cancer represent prognostic indicators for recurrence-free and overall survival. We provide evidence that response to chemotherapy is in part regulated by these leukocytes; cytotoxic therapies induce mammary epithelial cells to produce monocyte/macrophage recruitment factors, including colony stimulating factor 1 (CSF1) and interleukin-34, which together enhance CSF1 receptor (CSF1R)-dependent macrophage infiltration. Blockade of macrophage recruitment with CSF1R-signaling antagonists, in combination with paclitaxel, improved survival of mammary tumor-bearing mice by slowing primary tumor development and reducing pulmonary metastasis. These improved aspects of mammary carcinogenesis were accompanied by decreased vessel density and appearance of antitumor immune programs fostering tumor suppression in a CD8+ T-cell-dependent manner. These data provide a rationale for targeting macrophage recruitment/response pathways, notably CSF1R, in combination with cytotoxic therapy, and identification of a breast cancer population likely to benefit from this novel therapeutic approach. These findings reveal that response to chemotherapy is in part regulated by the tumor immune microenvironment and that common cytotoxic drugs induce neoplastic cells to produce monocyte/macrophage recruitment factors, which in turn enhance macrophage infiltration into mammary adenocarcinomas. Blockade of pathways mediating macrophage recruitment, in combination with chemotherapy, significantly decreases primary tumor progression, reduces metastasis, and improves survival by CD8+ T-cell-dependent mechanisms, thus indicating that the immune microenvironment of tumors can be reprogrammed to instead foster antitumor immunity and improve response to cytotoxic therapy.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Obstacles Posed by the Tumor Microenvironment to T cell Activity: A Case for Synergistic Therapies.

            T cell dysfunction in solid tumors results from multiple mechanisms. Altered signaling pathways in tumor cells help produce a suppressive tumor microenvironment enriched for inhibitory cells, posing a major obstacle for cancer immunity. Metabolic constraints to cell function and survival shape tumor progression and immune cell function. In the face of persistent antigen, chronic T cell receptor signaling drives T lymphocytes to a functionally exhausted state. Here we discuss how the tumor and its microenvironment influences T cell trafficking and function with a focus on melanoma, and pancreatic and ovarian cancer, and discuss how scientific advances may help overcome these hurdles.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              In vivo imaging reveals a tumor-associated macrophage–mediated resistance pathway in anti–PD-1 therapy

              Monoclonal antibodies (mAbs) targeting the immune checkpoint anti–programmed cell death protein 1 (aPD-1) have demonstrated impressive benefits for the treatment of some cancers; however, these drugs are not always effective, and we still have a limited understanding of the mechanisms that contribute to their efficacy or lack thereof. We used in vivo imaging to uncover the fate and activity of aPD-1 mAbs in real time and at subcellular resolution in mice. We show that aPD-1 mAbs effectively bind PD-1+ tumor-infiltrating CD8+ T cells at early time points after administration. However, this engagement is transient, and aPD-1 mAbs are captured within minutes from the T cell surface by PD-1– tumor-associated macrophages. We further show that macrophage accrual of aPD-1 mAbs depends both on the drug’s Fc domain glycan and on Fc receptors (FcRs) expressed by host myeloid cells and extend these findings to the human setting. Finally, we demonstrate that in vivo blockade of FcRs before aPD-1 mAb administration substantially prolongs aPD-1 mAb binding to tumor-infiltrating CD8+ T cells and enhances immunotherapy-induced tumor regression in mice. These investigations yield insight into aPD-1 target engagement in vivo and identify specific Fc/FcR interactions that can be modulated to improve checkpoint blockade 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
                24 April 2018
                9 April 2018
                9 April 2018
                : 115
                : 17
                : E4041-E4050
                Affiliations
                [1] aINSERM, U1016, Institut Cochin , 75014 Paris, France;
                [2] bCNRS , UMR8104, 75014 Paris, France;
                [3] cUniversité Paris Descartes , Sorbonne Paris Cité, 75014 Paris, France;
                [4] dINSERM U1138, Cordeliers Research Center, Team Cancer, Immune Control and Escape, University Pierre and Marie Curie , 75006 Paris, France;
                [5] eDepartment of Pathology, Paris Centre University Hospitals, University Paris Descartes , INSERM U1138, 75006 Paris, France;
                [6] fDepartment of Thoracic Surgery, Paris Centre University Hospitals, Université Paris Descartes , 75014 Paris, France
                Author notes
                2To whom correspondence may be addressed. Email: elisa.peranzoni@ 123456servier.com or emmanuel.donnadieu@ 123456inserm.fr .

                Edited by Douglas T. Fearon, Cornell University, Cambridge, United Kingdom, and approved March 19, 2018 (received for review December 2, 2017)

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

                1Present address: Translational and Clinical Research, Center for Therapeutic Innovation in Oncology, Servier Research Institute, 78290 Croissy-sur-Seine, France.

                Article
                201720948
                10.1073/pnas.1720948115
                5924916
                29632196
                b315bf7e-c47b-4f6c-93cb-2b89162a2eaf
                Copyright © 2018 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: 10
                Funding
                Funded by: Ligue Contre le Cancer 501100004099
                Award ID: Equipe Labélisée
                Funded by: Plan cancer France
                Award ID: Tumor heterogeneity and ecosystem program
                Funded by: SIRIC France
                Award ID: CARPEM
                Funded by: Fondation de France 501100004431
                Award ID: Post-doc fellowship
                Funded by: Associazione Italiana per la Ricerca sul Cancro (AIRC) 501100005010
                Award ID: Post-doc fellowship
                Categories
                PNAS Plus
                Biological Sciences
                Immunology and Inflammation
                PNAS Plus

                cancer,immunotherapy,t cells,macrophages,migration
                cancer, immunotherapy, t cells, macrophages, migration

                Comments

                Comment on this article