Recombinant Newcastle disease viruses expressing immunological checkpoint inhibitors induce a pro-inflammatory state and enhance tumor-specific immune responses in two murine models of cancer

IntroductionTumor microenvironments are immunosuppressive due to progressive accumulation of mutations in cancer cells that can drive expression of a range of inhibitory ligands and cytokines, and recruitment of immunomodulatory cells, including myeloid-derived suppressor cells (MDSC), tumor-associa...

Full description

Bibliographic Details
Main Authors: Lisa A. Santry, Jacob P. van Vloten, Amanda W. K. AuYeung, Robert C. Mould, Jacob G. E. Yates, Thomas M. McAusland, James J. Petrik, Pierre P. Major, Byram W. Bridle, Sarah K. Wootton
Format: Article
Language:English
Published: Frontiers Media S.A. 2024-01-01
Series:Frontiers in Microbiology
Subjects:
Online Access:https://www.frontiersin.org/articles/10.3389/fmicb.2024.1325558/full
_version_ 1797347864544804864
author Lisa A. Santry
Jacob P. van Vloten
Amanda W. K. AuYeung
Robert C. Mould
Jacob G. E. Yates
Thomas M. McAusland
James J. Petrik
Pierre P. Major
Byram W. Bridle
Sarah K. Wootton
author_facet Lisa A. Santry
Jacob P. van Vloten
Amanda W. K. AuYeung
Robert C. Mould
Jacob G. E. Yates
Thomas M. McAusland
James J. Petrik
Pierre P. Major
Byram W. Bridle
Sarah K. Wootton
author_sort Lisa A. Santry
collection DOAJ
description IntroductionTumor microenvironments are immunosuppressive due to progressive accumulation of mutations in cancer cells that can drive expression of a range of inhibitory ligands and cytokines, and recruitment of immunomodulatory cells, including myeloid-derived suppressor cells (MDSC), tumor-associated macrophages, and regulatory T cells (Tregs).MethodsTo reverse this immunosuppression, we engineered mesogenic Newcastle disease virus (NDV) to express immunological checkpoint inhibitors anti-cytotoxic T lymphocyte antigen-4 and soluble programmed death protein-1.ResultsIntratumoral administration of recombinant NDV (rNDV) to mice bearing intradermal B16-F10 melanomas or subcutaneous CT26LacZ colon carcinomas led to significant changes in the tumor-infiltrating lymphocyte profiles. Vectorizing immunological checkpoint inhibitors in NDV increased activation of intratumoral natural killer cells and cytotoxic T cells and decreased Tregs and MDSCs, suggesting induction of a pro-inflammatory state with greater infiltration of activated CD8+ T cells. These notable changes translated to higher ratios of activated effector/suppressor tumor-infiltrating lymphocytes in both cancer models, which is a promising prognostic marker. Whereas all rNDV-treated groups showed evidence of tumor regression and increased survival in the CT26LacZ and B16-F10, only treatment with NDV expressing immunological checkpoint blockades led to complete responses compared to tumors treated with NDV only.DiscussionThese data demonstrated that NDV expressing immunological checkpoint inhibitors could reverse the immunosuppressive state of tumor microenvironments and enhance tumor-specific T cell responses.
first_indexed 2024-03-08T11:54:01Z
format Article
id doaj.art-0dc2b238fb124dce87ad725117251258
institution Directory Open Access Journal
issn 1664-302X
language English
last_indexed 2024-03-08T11:54:01Z
publishDate 2024-01-01
publisher Frontiers Media S.A.
record_format Article
series Frontiers in Microbiology
spelling doaj.art-0dc2b238fb124dce87ad7251172512582024-01-24T04:49:10ZengFrontiers Media S.A.Frontiers in Microbiology1664-302X2024-01-011510.3389/fmicb.2024.13255581325558Recombinant Newcastle disease viruses expressing immunological checkpoint inhibitors induce a pro-inflammatory state and enhance tumor-specific immune responses in two murine models of cancerLisa A. Santry0Jacob P. van Vloten1Amanda W. K. AuYeung2Robert C. Mould3Jacob G. E. Yates4Thomas M. McAusland5James J. Petrik6Pierre P. Major7Byram W. Bridle8Sarah K. Wootton9Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, CanadaDepartment of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, CanadaDepartment of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, CanadaDepartment of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, CanadaDepartment of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, CanadaDepartment of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, CanadaDepartment of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, CanadaJuravinski Cancer Center, Hamilton, ON, CanadaDepartment of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, CanadaDepartment of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, CanadaIntroductionTumor microenvironments are immunosuppressive due to progressive accumulation of mutations in cancer cells that can drive expression of a range of inhibitory ligands and cytokines, and recruitment of immunomodulatory cells, including myeloid-derived suppressor cells (MDSC), tumor-associated macrophages, and regulatory T cells (Tregs).MethodsTo reverse this immunosuppression, we engineered mesogenic Newcastle disease virus (NDV) to express immunological checkpoint inhibitors anti-cytotoxic T lymphocyte antigen-4 and soluble programmed death protein-1.ResultsIntratumoral administration of recombinant NDV (rNDV) to mice bearing intradermal B16-F10 melanomas or subcutaneous CT26LacZ colon carcinomas led to significant changes in the tumor-infiltrating lymphocyte profiles. Vectorizing immunological checkpoint inhibitors in NDV increased activation of intratumoral natural killer cells and cytotoxic T cells and decreased Tregs and MDSCs, suggesting induction of a pro-inflammatory state with greater infiltration of activated CD8+ T cells. These notable changes translated to higher ratios of activated effector/suppressor tumor-infiltrating lymphocytes in both cancer models, which is a promising prognostic marker. Whereas all rNDV-treated groups showed evidence of tumor regression and increased survival in the CT26LacZ and B16-F10, only treatment with NDV expressing immunological checkpoint blockades led to complete responses compared to tumors treated with NDV only.DiscussionThese data demonstrated that NDV expressing immunological checkpoint inhibitors could reverse the immunosuppressive state of tumor microenvironments and enhance tumor-specific T cell responses.https://www.frontiersin.org/articles/10.3389/fmicb.2024.1325558/fullNewcastle disease virus (NDV)oncolytic virusimmunological checkpoint inhibitorsanti-CTLA-4PD-1PD-L1
spellingShingle Lisa A. Santry
Jacob P. van Vloten
Amanda W. K. AuYeung
Robert C. Mould
Jacob G. E. Yates
Thomas M. McAusland
James J. Petrik
Pierre P. Major
Byram W. Bridle
Sarah K. Wootton
Recombinant Newcastle disease viruses expressing immunological checkpoint inhibitors induce a pro-inflammatory state and enhance tumor-specific immune responses in two murine models of cancer
Frontiers in Microbiology
Newcastle disease virus (NDV)
oncolytic virus
immunological checkpoint inhibitors
anti-CTLA-4
PD-1
PD-L1
title Recombinant Newcastle disease viruses expressing immunological checkpoint inhibitors induce a pro-inflammatory state and enhance tumor-specific immune responses in two murine models of cancer
title_full Recombinant Newcastle disease viruses expressing immunological checkpoint inhibitors induce a pro-inflammatory state and enhance tumor-specific immune responses in two murine models of cancer
title_fullStr Recombinant Newcastle disease viruses expressing immunological checkpoint inhibitors induce a pro-inflammatory state and enhance tumor-specific immune responses in two murine models of cancer
title_full_unstemmed Recombinant Newcastle disease viruses expressing immunological checkpoint inhibitors induce a pro-inflammatory state and enhance tumor-specific immune responses in two murine models of cancer
title_short Recombinant Newcastle disease viruses expressing immunological checkpoint inhibitors induce a pro-inflammatory state and enhance tumor-specific immune responses in two murine models of cancer
title_sort recombinant newcastle disease viruses expressing immunological checkpoint inhibitors induce a pro inflammatory state and enhance tumor specific immune responses in two murine models of cancer
topic Newcastle disease virus (NDV)
oncolytic virus
immunological checkpoint inhibitors
anti-CTLA-4
PD-1
PD-L1
url https://www.frontiersin.org/articles/10.3389/fmicb.2024.1325558/full
work_keys_str_mv AT lisaasantry recombinantnewcastlediseasevirusesexpressingimmunologicalcheckpointinhibitorsinduceaproinflammatorystateandenhancetumorspecificimmuneresponsesintwomurinemodelsofcancer
AT jacobpvanvloten recombinantnewcastlediseasevirusesexpressingimmunologicalcheckpointinhibitorsinduceaproinflammatorystateandenhancetumorspecificimmuneresponsesintwomurinemodelsofcancer
AT amandawkauyeung recombinantnewcastlediseasevirusesexpressingimmunologicalcheckpointinhibitorsinduceaproinflammatorystateandenhancetumorspecificimmuneresponsesintwomurinemodelsofcancer
AT robertcmould recombinantnewcastlediseasevirusesexpressingimmunologicalcheckpointinhibitorsinduceaproinflammatorystateandenhancetumorspecificimmuneresponsesintwomurinemodelsofcancer
AT jacobgeyates recombinantnewcastlediseasevirusesexpressingimmunologicalcheckpointinhibitorsinduceaproinflammatorystateandenhancetumorspecificimmuneresponsesintwomurinemodelsofcancer
AT thomasmmcausland recombinantnewcastlediseasevirusesexpressingimmunologicalcheckpointinhibitorsinduceaproinflammatorystateandenhancetumorspecificimmuneresponsesintwomurinemodelsofcancer
AT jamesjpetrik recombinantnewcastlediseasevirusesexpressingimmunologicalcheckpointinhibitorsinduceaproinflammatorystateandenhancetumorspecificimmuneresponsesintwomurinemodelsofcancer
AT pierrepmajor recombinantnewcastlediseasevirusesexpressingimmunologicalcheckpointinhibitorsinduceaproinflammatorystateandenhancetumorspecificimmuneresponsesintwomurinemodelsofcancer
AT byramwbridle recombinantnewcastlediseasevirusesexpressingimmunologicalcheckpointinhibitorsinduceaproinflammatorystateandenhancetumorspecificimmuneresponsesintwomurinemodelsofcancer
AT sarahkwootton recombinantnewcastlediseasevirusesexpressingimmunologicalcheckpointinhibitorsinduceaproinflammatorystateandenhancetumorspecificimmuneresponsesintwomurinemodelsofcancer