scholarly journals A combinational therapy of EGFR-CAR NK cells and oncolytic herpes simplex virus 1 for breast cancer brain metastases

Oncotarget ◽  
2016 ◽  
Vol 7 (19) ◽  
pp. 27764-27777 ◽  
Author(s):  
Xilin Chen ◽  
Jianfeng Han ◽  
Jianhong Chu ◽  
Lingling Zhang ◽  
Jianying Zhang ◽  
...  
2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Bo Xu ◽  
Lei Tian ◽  
Jing Chen ◽  
Jing Wang ◽  
Rui Ma ◽  
...  

AbstractOncolytic herpes simplex virus-1 is capable of lysing tumor cells while alerting the immune system. CD47, in collaboration with SIRPα, represents an important immune checkpoint to inhibit phagocytosis by innate immune cells. Here we show locoregional control of glioblastoma by an oncolytic herpes virus expressing a full-length anti(α)-human CD47 IgG1 or IgG4 antibody. The antibodies secreted by the virus-infected glioblastoma cells block the CD47 ‘don’t eat me’ signal irrespective of the subclass; however, αCD47-IgG1 has a stronger tumor killing effect than αCD47-IgG4 due to additional antibody-dependent cellular phagocytosis by macrophages and antibody-dependent cellular cytotoxicity by NK cells. Intracranially injected αCD47-IgG1-producing virus continuously releases the respective antibody in the tumor microenvironment but not into systemic circulation; additionally, αCD47-IgG1-producing virus also improves the survival of tumor-bearing mice better than control oncolytic herpes virus combined with topical αCD47-IgG1. Results from immunocompetent mouse tumor models further confirm that macrophages, and to a lesser extent NK cells, mediate the anti-tumor cytotoxicity of antibody-producing oncolytic herpesviruses. Collectively, oncolytic herpes simplex virus-1 encoding full-length antibodies could improve immune-virotherapy for glioblastoma.


2012 ◽  
Vol 2012 ◽  
pp. 1-12 ◽  
Author(s):  
James J. Cody ◽  
Pietro Scaturro ◽  
Alan B. Cantor ◽  
G. Yancey Gillespie ◽  
Jacqueline N. Parker ◽  
...  

The metastasis of breast cancer to the brain and central nervous system (CNS) is a problem of increasing importance. As improving treatments continue to extend patient survival, the incidence of CNS metastases from breast cancer is on the rise. New treatments are needed, as current treatments are limited by deleterious side effects and are generally palliative. We have previously described an oncolytic herpes simplex virus (HSV), designated M002, which lacks both copies of theγ134.5 neurovirulence gene and carries a murine interleukin 12 (IL-12) expression cassette, and have validated its antitumor efficacy in a variety of preclinical models of primary brain tumors. However, M002 has not been yet evaluated for use against metastatic brain tumors. Here, we demonstrate the following: both human breast cancer and murine mammary carcinoma cells support viral replication and IL-12 expression from M002; M002 replicates in and destroys breast cancer cells from a variety of histological subtypes, including “triple-negative” and HER2 overexpressing; M002 improves survival in an immunocompetent model more effectively than does a non-cytokine control virus. Thus, we conclude from this proof-of-principle study that aγ134.5-deleted IL-12 expressing oncolytic HSV may be a potential new therapy for breast cancer brain metastases.


2019 ◽  
Vol 18 (6) ◽  
pp. 1127-1136 ◽  
Author(s):  
Tae Jin Lee ◽  
Mitra Nair ◽  
Yeshavanth Banasavadi-Siddegowda ◽  
Joseph Liu ◽  
Tejaswini Nallanagulagari ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document