Our system is currently under heavy load due to increased usage. We're actively working on upgrades to improve performance. Thank you for your patience.
2021
DOI: 10.1016/j.ymthe.2020.12.024
|View full text |Cite
|
Sign up to set email alerts
|

Fusogenic oncolytic vaccinia virus enhances systemic antitumor immune response by modulating the tumor microenvironment

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
2
1
1
1

Citation Types

0
13
0

Year Published

2021
2021
2024
2024

Publication Types

Select...
9

Relationship

1
8

Authors

Journals

citations
Cited by 15 publications
(13 citation statements)
references
References 37 publications
0
13
0
Order By: Relevance
“…However, it fails to develop oncolytic viruselicited antitumor immune responses, which play a critical role in the efficacy of oncolytic virotherapy. Our previous studies demonstrated that VGF and O1-deleted oncolytic VVs elicited potent antitumor effects via systemic antitumor activity rather than local viral oncolytic activity in syngeneic tumor mouse models using melanoma, colon, and lung carcinomas, and its antitumor effects were enhanced by arming VV with the expression of IL-12 and IL-7 [51] or induction of cell-cell fusion [52]. Thus, it is worth evaluating the combinational effects of armed MDRVV and 5-FC in syngenic mouse tumor models in the future.…”
Section: Discussionmentioning
confidence: 99%
“…However, it fails to develop oncolytic viruselicited antitumor immune responses, which play a critical role in the efficacy of oncolytic virotherapy. Our previous studies demonstrated that VGF and O1-deleted oncolytic VVs elicited potent antitumor effects via systemic antitumor activity rather than local viral oncolytic activity in syngeneic tumor mouse models using melanoma, colon, and lung carcinomas, and its antitumor effects were enhanced by arming VV with the expression of IL-12 and IL-7 [51] or induction of cell-cell fusion [52]. Thus, it is worth evaluating the combinational effects of armed MDRVV and 5-FC in syngenic mouse tumor models in the future.…”
Section: Discussionmentioning
confidence: 99%
“…FUVAC showed antitumor effect in a CD8+ T-cell-dependent manner and also inhibited infiltration of tumor-associated immune suppressive cells. 36 These results suggested that cell fusion is involved in oncolytic and antitumor effect of VV. A34 R is an EV glycoprotein of VV and involved in cell-to-cell transmission.…”
Section: Preclinical Investigation Of Oncolytic Vv In Crcmentioning
confidence: 91%
“… Immune checkpoint inhibitor i.t. 36 VVs Western reserve Deletion in N1 L, K1 L, K3 L, A46 R, or A52 R ΔK1 L VV, ΔA46 R VV, and ΔA52 R VV demonstrated improved antitumor effect and survival compared to vvDD. i.p.…”
Section: Preclinical Investigation Of Oncolytic Vv In Crcmentioning
confidence: 99%
“…FUVAC exhibited improved direct oncolytic activity and indirect anti-tumor immunity. FUVAC was reported to significantly inhibit tumor growth and improve the tumor immune microenvironment by reducing the tumor-associated immune suppressive cells, such as Treg cells, TAMs, and M-MDSCs, and increasing cytotoxic CD8+ T cells systemically [71]. In another study, VV was demonstrated to infect tumor infiltrating Tregs, leading to the depletion of viral-mediated Tregs, which is required for tumor regression [72].…”
Section: Vaccinia Virus For the Modulation Of Tumor Microenvironmentmentioning
confidence: 99%