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

      Biomarker and pharmacodynamic activity of the transforming growth factor‐beta (TGFβ) inhibitor SAR439459 as monotherapy and in combination with cemiplimab in a phase I clinical study in patients with advanced solid tumors

      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

          SAR439459, a ‘second‐generation’ human anti‐transforming growth factor‐beta (TGFβ) monoclonal antibody, inhibits all TGFβ isoforms and improves the antitumor activity of anti‐programmed cell death protein‐1 therapeutics. This study reports the pharmacodynamics (PD) and biomarker results from phase I/Ib first‐in‐human study of SAR439459 ± cemiplimab in patients with advanced solid tumors (NCT03192345). In dose‐escalation phase (Part 1), SAR439459 was administered intravenously at increasing doses either every 2 weeks (Q2W) or every 3 weeks (Q3W) with cemiplimab IV at 3 mg/kg Q2W or 350 mg Q3W, respectively, in patients with advanced solid tumors. In dose‐expansion phase (Part 2), patients with melanoma received SAR439459 IV Q3W at preliminary recommended phase II dose (pRP2D) of 22.5/7.5 mg/kg or at 22.5 mg/kg with cemiplimab 350 mg IV Q3W. Tumor biopsy and peripheral blood samples were collected for exploratory biomarker analyses to assess target engagement and PD, and results were correlated with patients' clinical parameters. SAR439459 ± cemiplimab showed decreased plasma and tissue TGFβ, downregulation of TGFβ‐pathway activation signature, modulation of peripheral natural killer (NK) and T cell expansion, proliferation, and increased secretion of CXCL10. Conversion of tumor tissue samples from ‘immune‐excluded’ to ‘immune‐infiltrated’ phenotype in a representative patient with melanoma SAR439459 22.5 mg/kg with cemiplimab was observed. In paired tumor and plasma, active and total TGFβ1 was more consistently elevated followed by TGFβ2, whereas TGFβ3 was only measurable (lower limit of quantitation ≥2.68 pg/mg) in tumors. SAR439459 ± cemiplimab showed expected peripheral PD effects and TGFβ alteration. However, further studies are needed to identify biomarkers of response.

          Related collections

          Most cited references12

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

          TGF-β attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells

          Therapeutic antibodies that block the programmed death-ligand 1 (PD-L1)/programmed death-1 (PD-1) pathway can induce robust and durable responses in patients with various cancers, including metastatic urothelial cancer (mUC) 1–5 . However, these responses only occur in a subset of patients. Elucidating the determinants of response and resistance is key to improving outcomes and developing new treatment strategies. Here, we examined tumours from a large cohort of mUC patients treated with an anti–PD-L1 agent (atezolizumab) and identified major determinants of clinical outcome. Response was associated with CD8+ T-effector cell phenotype and, to an even greater extent, high neoantigen or tumour mutation burden (TMB). Lack of response was associated with a signature of transforming growth factor β (TGF-β) signalling in fibroblasts, particularly in patients with CD8+ T cells that were excluded from the tumour parenchyma and instead found in the fibroblast- and collagen-rich peritumoural stroma—a common phenotype among patients with mUC. Using a mouse model that recapitulates this immune excluded phenotype, we found that therapeutic administration of a TGF-β blocking antibody together with anti–PD-L1 reduced TGF-β signalling in stromal cells, facilitated T cell penetration into the centre of the tumour, and provoked vigorous anti-tumour immunity and tumour regression. Integration of these three independent biological features provides the best basis for understanding outcome in this setting and suggests that TGF-β shapes the tumour microenvironment to restrain anti-tumour immunity by restricting T cell infiltration.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            TGFβ drives immune evasion in genetically reconstituted colon cancer metastasis

            Most patients with colorectal cancer die as a result of the disease spreading to other organs. However, no prevalent mutations have been associated with metastatic colorectal cancers. Instead, particular features of the tumour microenvironment, such as lack of T-cell infiltration, low type 1 T-helper cell (TH1) activity and reduced immune cytotoxicity or increased TGFβ levels predict adverse outcomes in patients with colorectal cancer. Here we analyse the interplay between genetic alterations and the tumour microenvironment by crossing mice bearing conditional alleles of four main colorectal cancer mutations in intestinal stem cells. Quadruple-mutant mice developed metastatic intestinal tumours that display key hallmarks of human microsatellite-stable colorectal cancers, including low mutational burden, T-cell exclusion and TGFβ-activated stroma. Inhibition of the PD-1-PD-L1 immune checkpoint provoked a limited response in this model system. By contrast, inhibition of TGFβ unleashed a potent and enduring cytotoxic T-cell response against tumour cells that prevented metastasis. In mice with progressive liver metastatic disease, blockade of TGFβ signalling rendered tumours susceptible to anti-PD-1-PD-L1 therapy. Our data show that increased TGFβ in the tumour microenvironment represents a primary mechanism of immune evasion that promotes T-cell exclusion and blocks acquisition of the TH1-effector phenotype. Immunotherapies directed against TGFβ signalling may therefore have broad applications in treating patients with advanced colorectal cancer.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Enhanced preclinical antitumor activity of M7824, a bifunctional fusion protein simultaneously targeting PD-L1 and TGF-β

              Antibodies targeting immune checkpoints are emerging as potent and viable cancer therapies, but not all patients respond to these as single agents. Concurrently targeting additional immunosuppressive pathways is a promising approach to enhance immune checkpoint blockade, and bifunctional molecules designed to target two pathways simultaneously may provide a strategic advantage over the combination of two single agents. M7824 (MSB0011359C) is a bifunctional fusion protein composed of a monoclonal antibody against programmed death ligand 1 (PD-L1) fused to the extracellular domain of human transforming growth factor–β (TGF-β) receptor II, which functions as a "trap" for all three TGF-β isoforms. We demonstrate that M7824 efficiently, specifically, and simultaneously binds PD-L1 and TGF-β. In syngeneic mouse models, M7824 suppressed tumor growth and metastasis more effectively than treatment with either an anti–PD-L1 antibody or TGF-β trap alone; furthermore, M7824 extended survival and conferred long-term protective antitumor immunity. Mechanistically, the dual anti-immunosuppressive function of M7824 resulted in activation of both the innate and adaptive immune systems, which contributed to M7824’s antitumor activity. Finally, M7824 was an effective combination partner for radiotherapy or chemotherapy in mouse models. Collectively, our preclinical data demonstrate that simultaneous blockade of the PD-L1 and TGF-β pathways by M7824 elicits potent and superior antitumor activity relative to monotherapies.
                Bookmark

                Author and article information

                Contributors
                rui.wang@sanofi.com
                Journal
                Clin Transl Sci
                Clin Transl Sci
                10.1111/(ISSN)1752-8062
                CTS
                Clinical and Translational Science
                John Wiley and Sons Inc. (Hoboken )
                1752-8054
                1752-8062
                16 February 2024
                February 2024
                : 17
                : 2 ( doiID: 10.1111/cts.v17.2 )
                : e13736
                Affiliations
                [ 1 ] Medical Oncology Erasmus MC Cancer Institute Rotterdam The Netherlands
                [ 2 ] Dermatology and Oncology Service Aix Marseille University and Timone Hospital Marseille France
                [ 3 ] Department of General Medical Oncology Leuven Cancer Institute, University Hospitals Leuven, KU Leuven Leuven Belgium
                [ 4 ] Department of Biomedical Science Humanitas University Milan Italy
                [ 5 ] Department of Medical Oncology and Hematology IRCCS Humanitas Research Hospital Milan Italy
                [ 6 ] START Madrid‐FJD, Early Phase Clinical Trials Unit Hospital Universitario Fundación Jiménez Díaz Madrid Spain
                [ 7 ] Department of Hepatogastroenterology Cliniques Universitaires Saint‐Luc, Université Catholique de Louvain Brussels Belgium
                [ 8 ] Department of Medical Oncology and Hematology, Department of Immunology Princess Margaret Cancer Centre, University of Toronto Toronto Ontario Canada
                [ 9 ] Division of Medical Oncology, Department of Oncology University of Calgary Calgary Alberta Canada
                [ 10 ] Département d'Innovation Thérapeutique et d'Essais Précoces, Gustave Roussy Université Paris‐Saclay Villejuif France
                [ 11 ] Service d'oncologie médicale, CHU Nantes Université de Nantes Nantes France
                [ 12 ] Unit of Immunotherapy and Anticancer Innovative Therapeutics Fondazione IRCCS Istituto Nazionale Tumori Milan Italy
                [ 13 ] Division of Early Drug Development European Institute of Oncology IRCCS Milan Italy
                [ 14 ] Department of Oncology and Hemato‐Oncology University of Milan Milan Italy
                [ 15 ] Department of Oncology, Asan Medical Center University of Ulsan College of Medicine Seoul South Korea
                [ 16 ] Medical Oncology Department, Hospital del Mar, CIBERONC IMIM Research Institute Barcelona Spain
                [ 17 ] Department of Dermatology University Hospital Essen Essen Germany
                [ 18 ] German Cancer Consortium, partner site Essen Essen Germany
                [ 19 ] NCT‐West, Campus Essen Essen Germany
                [ 20 ] University Alliance Ruhr, Research Center One Health, University Duisburg‐Essen Essen Germany
                [ 21 ] Medical Oncology Department Vall d'Hebron University Hospital and Institute of Oncology Barcelona Spain
                [ 22 ] Sanofi Bridgewater New Jersey USA
                [ 23 ] Sanofi Vitry‐Sur‐Seine France
                [ 24 ] Sanofi Cambridge Massachusetts USA
                Author notes
                [*] [* ] Correspondence

                Rui Wang, Sanofi, Cambridge, Massachusetts, USA.

                Email: rui.wang@ 123456sanofi.com

                [†]

                Affiliation at the time of the study.

                Author information
                https://orcid.org/0000-0003-0667-3284
                Article
                CTS13736 CTS-2023-0339
                10.1111/cts.13736
                10870242
                38362837
                e1f0f705-96d3-4352-ab21-bdecec3072eb
                © 2024 The Authors. Clinical and Translational Science published by Wiley Periodicals LLC on behalf of American Society for Clinical Pharmacology and Therapeutics.

                This is an open access article under the terms of the http://creativecommons.org/licenses/by-nc-nd/4.0/ License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non‐commercial and no modifications or adaptations are made.

                History
                : 19 December 2023
                : 11 October 2023
                : 24 December 2023
                Page count
                Figures: 5, Tables: 0, Pages: 11, Words: 5869
                Categories
                Article
                Articles
                Custom metadata
                2.0
                February 2024
                Converter:WILEY_ML3GV2_TO_JATSPMC version:6.3.8 mode:remove_FC converted:16.02.2024

                Medicine
                Medicine

                Comments

                Comment on this article