Your browser doesn't support javascript.
loading
A New Strategy Toward B Cell-Based Cancer Vaccines by Active Immunization With Mimotopes of Immune Checkpoint Inhibitors.
Tobias, Joshua; Battin, Claire; De Sousa Linhares, Annika; Lebens, Michael; Baier, Karin; Ambroz, Katharina; Drinic, Mirjana; Högler, Sandra; Inic-Kanada, Aleksandra; Garner-Spitzer, Erika; Preusser, Matthias; Kenner, Lukas; Kundi, Michael; Zielinski, Christoph C; Steinberger, Peter; Wiedermann, Ursula.
Affiliation
  • Tobias J; Center for Pathophysiology, Infectiology and Immunology, Institute of Specific Prophylaxis and Tropical Medicine, Medical University of Vienna, Vienna, Austria.
  • Battin C; Division of Immune Receptors and T Cell Activation, Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria.
  • De Sousa Linhares A; Division of Immune Receptors and T Cell Activation, Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria.
  • Lebens M; Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg Vaccine Research Institute (GUVAX), University of Gothenburg, Göteborg, Sweden.
  • Baier K; Center for Pathophysiology, Infectiology and Immunology, Institute of Specific Prophylaxis and Tropical Medicine, Medical University of Vienna, Vienna, Austria.
  • Ambroz K; Center for Pathophysiology, Infectiology and Immunology, Institute of Specific Prophylaxis and Tropical Medicine, Medical University of Vienna, Vienna, Austria.
  • Drinic M; Center for Pathophysiology, Infectiology and Immunology, Institute of Specific Prophylaxis and Tropical Medicine, Medical University of Vienna, Vienna, Austria.
  • Högler S; Unit of Laboratory Animal Pathology, Institute of Pathology, University of Veterinary Medicine Vienna, Vienna, Austria.
  • Inic-Kanada A; Center for Pathophysiology, Infectiology and Immunology, Institute of Specific Prophylaxis and Tropical Medicine, Medical University of Vienna, Vienna, Austria.
  • Garner-Spitzer E; Center for Pathophysiology, Infectiology and Immunology, Institute of Specific Prophylaxis and Tropical Medicine, Medical University of Vienna, Vienna, Austria.
  • Preusser M; Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria.
  • Kenner L; Unit of Laboratory Animal Pathology, Institute of Pathology, University of Veterinary Medicine Vienna, Vienna, Austria.
  • Kundi M; Department of Experimental Pathology, Medical University of Vienna, Vienna, Austria.
  • Zielinski CC; Department of Environmental Health, Center for Public Health, Medical University of Vienna, Vienna, Austria.
  • Steinberger P; Vienna Cancer Center (VCC), Medical University Vienna, and Vienna Hospital Association, Vienna, Austria.
  • Wiedermann U; Division of Immune Receptors and T Cell Activation, Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria.
Front Immunol ; 11: 895, 2020.
Article in En | MEDLINE | ID: mdl-32528470
ABSTRACT
Therapeutic monoclonal antibodies (mAbs), targeting tumor antigens, or immune checkpoints, have demonstrated a remarkable anti-tumor effect against various malignancies. However, high costs for mono- or combination therapies, associated with adverse effects or possible development of resistance in some patients, warrant further development and modification to gain more flexibility for this immunotherapy approach. An attractive alternative to passive immunization with therapeutic antibodies might be active immunization with mimotopes (B-cell peptides) representing the mAbs' binding epitopes, to activate the patient's own anti-tumor immune response following immunization. Here, we identified and examined the feasibility of inducing anti-tumor effects in vivo following active immunization with a mimotope of the immune checkpoint programmed cell death 1 (PD1), alone or in combination with a Her-2/neu B-cell peptide vaccine. Overlapping peptides spanning the extracellular domains of human PD1 (hPD1) were used to identify hPD1-derived mimotopes, using the therapeutic mAb Nivolumab as a proof of concept. Additionally, for in vivo evaluation in a tumor mouse model, a mouse PD1 (mPD1)-derived mimotope was identified using an anti-mPD1 mAb with mPD1/mPDL-1 blocking capacity. The identified mimotopes were characterized by in vitro assays, including a reporter cell-based assay, and their anti-tumor effects were evaluated in a syngeneic tumor mouse model stably expressing human Her-2/neu. The identified PD1-derived mimotopes were shown to significantly block the mAbs' capacity in inhibiting the respective PD1/PD-L1 interactions. A significant reduction in tumor growth in vivo was observed following active immunization with the mPD1-derived mimotope, associated with a significant reduction in proliferation and increased apoptotic rates in the tumors. Particularly, combined vaccination with the mPD1-derived mimotope and a multiple B-cell epitope Her-2/neu vaccine potentiated the vaccine's anti-tumor effect. Our results suggest active immunization with mimotopes of immune checkpoint inhibitors either as monotherapy or as combination therapy with tumor-specific vaccines, as a new strategy for cancer treatment.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Breast Neoplasms / B-Lymphocytes / Receptor, ErbB-2 / Cancer Vaccines / Programmed Cell Death 1 Receptor / Antineoplastic Agents, Immunological / Nivolumab / Immune Checkpoint Inhibitors / Epitopes Limits: Animals / Female / Humans Language: En Journal: Front Immunol Year: 2020 Document type: Article Affiliation country:

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Breast Neoplasms / B-Lymphocytes / Receptor, ErbB-2 / Cancer Vaccines / Programmed Cell Death 1 Receptor / Antineoplastic Agents, Immunological / Nivolumab / Immune Checkpoint Inhibitors / Epitopes Limits: Animals / Female / Humans Language: En Journal: Front Immunol Year: 2020 Document type: Article Affiliation country: