Your browser doesn't support javascript.
loading
Intratumoral Delivery of Interleukin 9 via Oncolytic Vaccinia Virus Elicits Potent Antitumor Effects in Tumor Models.
Ye, Junjie; Chen, Lingjuan; Waltermire, Julia; Zhao, Jinshun; Ren, Jinghua; Guo, Zongsheng; Bartlett, David L; Liu, Zuqiang.
Afiliação
  • Ye J; Allegheny Health Network Cancer Institute, Pittsburgh, PA 15212, USA.
  • Chen L; Department of Surgery, Drexel University College of Medicine, Philadelphia, PA 19104, USA.
  • Waltermire J; Department of Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China.
  • Zhao J; Allegheny Health Network Cancer Institute, Pittsburgh, PA 15212, USA.
  • Ren J; Department of Surgery, Drexel University College of Medicine, Philadelphia, PA 19104, USA.
  • Guo Z; Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
  • Bartlett DL; Allegheny Health Network Cancer Institute, Pittsburgh, PA 15212, USA.
  • Liu Z; Allegheny Health Network Cancer Institute, Pittsburgh, PA 15212, USA.
Cancers (Basel) ; 16(5)2024 Feb 29.
Article em En | MEDLINE | ID: mdl-38473379
ABSTRACT
The success of cancer immunotherapy is largely associated with immunologically hot tumors. Approaches that promote the infiltration of immune cells into tumor beds are urgently needed to transform cold tumors into hot tumors. Oncolytic viruses can transform the tumor microenvironment (TME), resulting in immunologically hot tumors. Cytokines are good candidates for arming oncolytic viruses to enhance their function in this transformation. Here, we used the oncolytic vaccinia virus (oVV) to deliver interleukin-9 (IL-9) into the tumor bed and explored its antitumor effects in colon and lung tumor models. Our data show that IL-9 prolongs viral persistence, which is probably mediated by the up-regulation of IL-10. The vvDD-IL-9 treatment elevated the expression of Th1 chemokines and antitumor factors such as IFN-γ, granzyme B, and perforin. IL-9 expression increased the percentages of CD4+ and CD8+ T cells in the TME and decreased the percentage of oVV-induced immune suppressive myeloid-derived suppressor cells (MDSC), leading to potent antitumor effects compared with parental virus treatment. The vvDD-IL-9 treatment also increased the percentage of regulatory T cells (Tregs) in the TME and elevated the expression of immune checkpoint molecules such as PD-1, PD-L1, and CTLA-4, but not GITR. The combination therapy of vvDD-IL-9 and the anti-CTLA-4 antibody, but not the anti-GITR antibody, induced systemic tumor-specific antitumor immunity and significantly extended the overall survival of mice, indicating a potential translation of the IL-9-expressing oncolytic virus into a clinical trial to enhance the antitumor effects elicited by an immune checkpoint blockade for cancer immunotherapy.
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Idioma: En Revista: Cancers (Basel) Ano de publicação: 2024 Tipo de documento: Article País de afiliação: Estados Unidos

Texto completo: 1 Base de dados: MEDLINE Idioma: En Revista: Cancers (Basel) Ano de publicação: 2024 Tipo de documento: Article País de afiliação: Estados Unidos