Oncolytic Orf virus licenses NK cells via cDC1 to activate innate and adaptive antitumor mechanisms and extends survival in a murine model of late-stage ovarian cancer

Background Novel therapies are needed to improve outcomes for women diagnosed with ovarian cancer. Oncolytic viruses are multifunctional immunotherapeutic biologics that preferentially infect cancer cells and stimulate inflammation with the potential to generate antitumor immunity. Herein we describ...

Full description

Bibliographic Details
Main Authors: Jacob P van Vloten, Kathy Matuszewska, Mark A A Minow, Jessica A Minott, Lisa A Santry, Madison Pereira, Ashley A Stegelmeier, Thomas M McAusland, Elaine M Klafuric, Khalil Karimi, Joseph Colasanti, D Grant McFadden, James J Petrik, Byram W Bridle, Sarah K Wootton
Format: Article
Language:English
Published: BMJ Publishing Group 2022-03-01
Series:Journal for ImmunoTherapy of Cancer
Online Access:https://jitc.bmj.com/content/10/3/e004335.full
_version_ 1797771674309885952
author Jacob P van Vloten
Kathy Matuszewska
Mark A A Minow
Jessica A Minott
Lisa A Santry
Madison Pereira
Ashley A Stegelmeier
Thomas M McAusland
Elaine M Klafuric
Khalil Karimi
Joseph Colasanti
D Grant McFadden
James J Petrik
Byram W Bridle
Sarah K Wootton
author_facet Jacob P van Vloten
Kathy Matuszewska
Mark A A Minow
Jessica A Minott
Lisa A Santry
Madison Pereira
Ashley A Stegelmeier
Thomas M McAusland
Elaine M Klafuric
Khalil Karimi
Joseph Colasanti
D Grant McFadden
James J Petrik
Byram W Bridle
Sarah K Wootton
author_sort Jacob P van Vloten
collection DOAJ
description Background Novel therapies are needed to improve outcomes for women diagnosed with ovarian cancer. Oncolytic viruses are multifunctional immunotherapeutic biologics that preferentially infect cancer cells and stimulate inflammation with the potential to generate antitumor immunity. Herein we describe Parapoxvirus ovis (Orf virus (OrfV)), an oncolytic poxvirus, as a viral immunotherapy for ovarian cancer.Methods The immunotherapeutic potential of OrfV was tested in the ID8 orthotopic mouse model of end-stage epithelial ovarian carcinoma. Immune cell profiling, impact on secondary lesion development and survival were evaluated in OrfV-treated mice as well as in Batf3 knockout, mice depleted of specific immune cell subsets and in mice where the primary tumor was removed. Finally, we interrogated gene expression datasets from primary human ovarian tumors from the International Cancer Genome Consortium database to determine whether the interplay we observed between natural killer (NK) cells, classical type 1 dendritic cells (cDC1s) and T cells exists and influences outcomes in human ovarian cancer.Results OrfV was an effective monotherapy in a murine model of advanced-stage epithelial ovarian cancer. OrfV intervention relied on NK cells, which when depleted abrogated antitumor CD8+ T-cell responses. OrfV therapy was shown to require cDC1s in experiments with BATF3 knockout mice, which do not have mature cDC1s. Furthermore, cDC1s governed antitumor NK and T-cell responses to mediate antitumor efficacy following OrfV. Primary tumor removal, a common treatment option in human patients, was effectively combined with OrfV for optimal therapeutic outcome. Analysis of human RNA sequencing datasets revealed that cDC1s correlate with NK cells in human ovarian cancer and that intratumoral NK cells correlate positively with survival.Conclusions The data herein support the translational potential of OrfV as an NK stimulating immunotherapeutic for the treatment of advanced-stage ovarian cancer.
first_indexed 2024-03-12T21:41:00Z
format Article
id doaj.art-042f07917dda4af9ab08f790da95d074
institution Directory Open Access Journal
issn 2051-1426
language English
last_indexed 2024-03-12T21:41:00Z
publishDate 2022-03-01
publisher BMJ Publishing Group
record_format Article
series Journal for ImmunoTherapy of Cancer
spelling doaj.art-042f07917dda4af9ab08f790da95d0742023-07-26T22:55:07ZengBMJ Publishing GroupJournal for ImmunoTherapy of Cancer2051-14262022-03-0110310.1136/jitc-2021-004335Oncolytic Orf virus licenses NK cells via cDC1 to activate innate and adaptive antitumor mechanisms and extends survival in a murine model of late-stage ovarian cancerJacob P van Vloten0Kathy Matuszewska1Mark A A Minow2Jessica A Minott3Lisa A Santry4Madison Pereira5Ashley A Stegelmeier6Thomas M McAusland7Elaine M Klafuric8Khalil Karimi9Joseph Colasanti10D Grant McFadden11James J Petrik12Byram W Bridle13Sarah K Wootton14Department of Pathobiology, University of Guelph, Guelph, Ontario, CanadaDepartment of Biomedical Sciences, University of Guelph, Guelph, Ontario, CanadaMolecular and Cellular Biology, University of Guelph, Guelph, Ontario, CanadaDepartment of Pathobiology, University of Guelph, Guelph, Ontario, CanadaDepartment of Pathobiology, University of Guelph, Guelph, Ontario, CanadaDepartment of Biomedical Sciences, University of Guelph, Guelph, Ontario, CanadaDepartment of Pathobiology, University of Guelph, Guelph, Ontario, CanadaDepartment of Pathobiology, University of Guelph, Guelph, Ontario, CanadaDepartment of Pathobiology, University of Guelph, Guelph, Ontario, CanadaDepartment of Pathobiology, University of Guelph, Guelph, Ontario, CanadaDepartment of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, CanadaBiodesign Institute, Arizona State University, Tempe, Arizona, USADepartment of Biomedical Sciences, University of Guelph, Guelph, Ontario, CanadaDepartment of Pathobiology, University of Guelph, Guelph, Ontario, CanadaDepartment of Pathobiology, University of Guelph, Guelph, Ontario, CanadaBackground Novel therapies are needed to improve outcomes for women diagnosed with ovarian cancer. Oncolytic viruses are multifunctional immunotherapeutic biologics that preferentially infect cancer cells and stimulate inflammation with the potential to generate antitumor immunity. Herein we describe Parapoxvirus ovis (Orf virus (OrfV)), an oncolytic poxvirus, as a viral immunotherapy for ovarian cancer.Methods The immunotherapeutic potential of OrfV was tested in the ID8 orthotopic mouse model of end-stage epithelial ovarian carcinoma. Immune cell profiling, impact on secondary lesion development and survival were evaluated in OrfV-treated mice as well as in Batf3 knockout, mice depleted of specific immune cell subsets and in mice where the primary tumor was removed. Finally, we interrogated gene expression datasets from primary human ovarian tumors from the International Cancer Genome Consortium database to determine whether the interplay we observed between natural killer (NK) cells, classical type 1 dendritic cells (cDC1s) and T cells exists and influences outcomes in human ovarian cancer.Results OrfV was an effective monotherapy in a murine model of advanced-stage epithelial ovarian cancer. OrfV intervention relied on NK cells, which when depleted abrogated antitumor CD8+ T-cell responses. OrfV therapy was shown to require cDC1s in experiments with BATF3 knockout mice, which do not have mature cDC1s. Furthermore, cDC1s governed antitumor NK and T-cell responses to mediate antitumor efficacy following OrfV. Primary tumor removal, a common treatment option in human patients, was effectively combined with OrfV for optimal therapeutic outcome. Analysis of human RNA sequencing datasets revealed that cDC1s correlate with NK cells in human ovarian cancer and that intratumoral NK cells correlate positively with survival.Conclusions The data herein support the translational potential of OrfV as an NK stimulating immunotherapeutic for the treatment of advanced-stage ovarian cancer.https://jitc.bmj.com/content/10/3/e004335.full
spellingShingle Jacob P van Vloten
Kathy Matuszewska
Mark A A Minow
Jessica A Minott
Lisa A Santry
Madison Pereira
Ashley A Stegelmeier
Thomas M McAusland
Elaine M Klafuric
Khalil Karimi
Joseph Colasanti
D Grant McFadden
James J Petrik
Byram W Bridle
Sarah K Wootton
Oncolytic Orf virus licenses NK cells via cDC1 to activate innate and adaptive antitumor mechanisms and extends survival in a murine model of late-stage ovarian cancer
Journal for ImmunoTherapy of Cancer
title Oncolytic Orf virus licenses NK cells via cDC1 to activate innate and adaptive antitumor mechanisms and extends survival in a murine model of late-stage ovarian cancer
title_full Oncolytic Orf virus licenses NK cells via cDC1 to activate innate and adaptive antitumor mechanisms and extends survival in a murine model of late-stage ovarian cancer
title_fullStr Oncolytic Orf virus licenses NK cells via cDC1 to activate innate and adaptive antitumor mechanisms and extends survival in a murine model of late-stage ovarian cancer
title_full_unstemmed Oncolytic Orf virus licenses NK cells via cDC1 to activate innate and adaptive antitumor mechanisms and extends survival in a murine model of late-stage ovarian cancer
title_short Oncolytic Orf virus licenses NK cells via cDC1 to activate innate and adaptive antitumor mechanisms and extends survival in a murine model of late-stage ovarian cancer
title_sort oncolytic orf virus licenses nk cells via cdc1 to activate innate and adaptive antitumor mechanisms and extends survival in a murine model of late stage ovarian cancer
url https://jitc.bmj.com/content/10/3/e004335.full
work_keys_str_mv AT jacobpvanvloten oncolyticorfviruslicensesnkcellsviacdc1toactivateinnateandadaptiveantitumormechanismsandextendssurvivalinamurinemodeloflatestageovariancancer
AT kathymatuszewska oncolyticorfviruslicensesnkcellsviacdc1toactivateinnateandadaptiveantitumormechanismsandextendssurvivalinamurinemodeloflatestageovariancancer
AT markaaminow oncolyticorfviruslicensesnkcellsviacdc1toactivateinnateandadaptiveantitumormechanismsandextendssurvivalinamurinemodeloflatestageovariancancer
AT jessicaaminott oncolyticorfviruslicensesnkcellsviacdc1toactivateinnateandadaptiveantitumormechanismsandextendssurvivalinamurinemodeloflatestageovariancancer
AT lisaasantry oncolyticorfviruslicensesnkcellsviacdc1toactivateinnateandadaptiveantitumormechanismsandextendssurvivalinamurinemodeloflatestageovariancancer
AT madisonpereira oncolyticorfviruslicensesnkcellsviacdc1toactivateinnateandadaptiveantitumormechanismsandextendssurvivalinamurinemodeloflatestageovariancancer
AT ashleyastegelmeier oncolyticorfviruslicensesnkcellsviacdc1toactivateinnateandadaptiveantitumormechanismsandextendssurvivalinamurinemodeloflatestageovariancancer
AT thomasmmcausland oncolyticorfviruslicensesnkcellsviacdc1toactivateinnateandadaptiveantitumormechanismsandextendssurvivalinamurinemodeloflatestageovariancancer
AT elainemklafuric oncolyticorfviruslicensesnkcellsviacdc1toactivateinnateandadaptiveantitumormechanismsandextendssurvivalinamurinemodeloflatestageovariancancer
AT khalilkarimi oncolyticorfviruslicensesnkcellsviacdc1toactivateinnateandadaptiveantitumormechanismsandextendssurvivalinamurinemodeloflatestageovariancancer
AT josephcolasanti oncolyticorfviruslicensesnkcellsviacdc1toactivateinnateandadaptiveantitumormechanismsandextendssurvivalinamurinemodeloflatestageovariancancer
AT dgrantmcfadden oncolyticorfviruslicensesnkcellsviacdc1toactivateinnateandadaptiveantitumormechanismsandextendssurvivalinamurinemodeloflatestageovariancancer
AT jamesjpetrik oncolyticorfviruslicensesnkcellsviacdc1toactivateinnateandadaptiveantitumormechanismsandextendssurvivalinamurinemodeloflatestageovariancancer
AT byramwbridle oncolyticorfviruslicensesnkcellsviacdc1toactivateinnateandadaptiveantitumormechanismsandextendssurvivalinamurinemodeloflatestageovariancancer
AT sarahkwootton oncolyticorfviruslicensesnkcellsviacdc1toactivateinnateandadaptiveantitumormechanismsandextendssurvivalinamurinemodeloflatestageovariancancer