oncolytic virotherapy
Recently Published Documents


TOTAL DOCUMENTS

515
(FIVE YEARS 176)

H-INDEX

38
(FIVE YEARS 8)

2021 ◽  
Vol 45 (2) ◽  
pp. 26-32
Author(s):  
Qayssar A Obaid ◽  
Khalisa K Khudair ◽  
Ahmed M Al-Shammari

One of the "hallmarks of cancer" is altered energy metabolism, which is increased glycolysis in cancer cells, the primary source of energy that uses this metabolic pathway to generate ATP. Oncolytic virotherapy with aerobic glycolysis inhibitor smart therapeutic approach to induce apoptosis in cancer cells. The current study aimed to use the 2-Deoxyglucose (2DG), a specific glycolysis inhibitor, to enhance the Newcastle disease virus (NDV). In this study, a mouse model of breast cancer allograft with mammary adenocarcinoma tumor cells (AN3) was used and treated with 2DG, NDV, and a combination of both. Anti-tumor efficacy and glycolysis analysis (hexokinase -1 (HK-1), pyruvate, and ATP) were determined. The induction of oxidative stress was investigated by reactive oxygen species (ROS) and total glutathione assay examination. Apoptosis induction was investigated using immunohistochemistry (cleaved Caspase-3) and histopathology. The result showed that combination therapy enhances anti-tumor efficacy (decrease in relative tumor volume and increase in tumor growth inhibition) of NDV against breast cancer. This effect was accompanied by a reduction in HK-1 concentration, pyruvate, and ATP (glycolysis products). Moreover, NDV+2DG therapy induces oxidative stress (decreases total glutathione and increases ROS). Immunohistochemistry and histopathological examination showed the apoptotic area in tumor tissues in treated groups. In conclusion, the present study found that the combination therapy could be considered as an effective cancer therapy through induction of glycolysis inhibition, oxidative stress, and apoptosis selectively in cancer cells.


Science ◽  
2021 ◽  
Vol 374 (6573) ◽  
pp. 1325-1326
Author(s):  
Alan Melcher ◽  
Kevin Harrington ◽  
Richard Vile

2021 ◽  
Author(s):  
Nasser Hashemi Goradel ◽  
Arezoo Alizadeh ◽  
Shahnaz Hosseinzadeh ◽  
Mitra Taghipour ◽  
Zeinab Ghesmati ◽  
...  

Oncolytic virotherapy has currently emerged as a powerful therapeutic approach in cancer treatment. Although the history of using viruses goes back to the early 20th century, the approval of talimogene laherparepvec (T-VEC) in 2015 increased interest in oncolytic viruses (OVs). OVs are multifaceted biotherapeutic agents because they replicate in and kill tumor cells and augment immune responses by releasing immunostimulatory molecules from lysed cells. Despite promising results, some limitations hinder the efficacy of oncolytic virotherapy. The delivery challenges and the upregulation of checkpoints following oncolytic virotherapy also mediate resistance to OVs by diminishing immune responses. Furthermore, the localization of receptors of viruses in the tight junctions, interferon responses, and the aberrant expression of genes involved in the cell cycle of the virus, including their infection and replication, reduce the efficacy of OVs. In this review, we present different mechanisms of resistance to OVs and strategies to overcome them.


2021 ◽  
Vol 23 (Supplement_6) ◽  
pp. vi177-vi177
Author(s):  
Eleni Panagioti ◽  
Michal Nowicki ◽  
William Goins ◽  
Jorge Jimenez Macias ◽  
Julia Escobar ◽  
...  

Abstract Glioblastoma (GBM) is the most common primary malignant brain tumor in adults with median overall survival of 14-16 months. Glioblastoma progression involves multiple immunosuppressive pathways hindering the success of cancer immunotherapy. Oncolytic virotherapy is a promising approach to reprogram the glioblastoma microenvironment and restore anti-tumor immunity. Interleukin (IL)-27 receptor signaling pathway regulates development of effector T cells, IL-10 producing T regulatory type 1 (Tr1) cells and induction of co-inhibitory receptors associated with T cell exhaustion in cancer. In this study, we sought to tackle local and systemic GBM-induced immunosuppression using the novel rQNestin34.5v2 oncolytic herpes simplex virus type-1 (HSV-1), engineered to selectively replicate in glioma cells together with genetic disruptions of the IL-27 receptor signaling pathway. The antitumor activity of HSV-1 rQNestin34.5v2 was evaluated in IL-27ra-/- and wild-type C57BL/6 mice harboring orthotopic CT-2A and GL261 glioblastoma and in athymic nude mice harboring orthotopic patient derived glioblastoma xenografts (PDXs) selected from patients that had been treated in a clinical trial of intratumoral administration of HSV-1 rQNestin34.5v2 (ClinicalTrials.gov: NCT03152318). HSV-1 rQNestin34.5v2 exhibited superior capacity to infect glioma cells in vitro triggering release of proinflammatory cytokines and inducing immunogenic cell death in infected cells. Immune phenotyping of GL261 and CT-2A gliomas revealed astrocytes and microglia express the highest levels of IL-27R. Lack of IL-27R signaling decreased expression of the co-inhibitory receptors PD-1 and Tim-3 on circulating CD4+ T cells suggesting altered CD4+ T cell responses. In the tumor microenvironment, lack of IL-27R signaling decreased immune suppression as evidenced by reduced interleukin (IL)-10 secretion by glioma cells. Despite positive functional modifications on CD4+ T cells, global lack of IL-27R signaling promoted tumor growth accompanied by increased circulating myeloid derived suppressor cells (MDSCs) in the GL261 model. Experiments to address the immunoregulatory role of IL-27R signaling during HSV-1 rQNestin34.5v2 virotherapy-induced glioma tissue destruction are ongoing.


2021 ◽  
Vol 23 (Supplement_6) ◽  
pp. vi175-vi175
Author(s):  
Kazuhide Shimizu ◽  
Andranik Kahramanian ◽  
Hiroaki Wakimoto

Abstract BACKGROUND Photodynamic therapy (PDT) is a targeted cancer therapy utilizing tumor-specific accumulation of photosensitizers and generation of reactive oxygen species (ROS) upon receiving specific light. The deadly CNS malignancies, high-grade gliomas and malignant meningioma, represent excellent candidates for this therapeutic method due to accessibility to light irradiation at the time of surgery. On the other hand, oncolytic virotherapy using a genetically engineered oncolytic herpes simplex virus (oHSV), has been intensively investigated as a multi-mechanistic therapy against these tumors. One of the advantages of oHSV is its ability to incorporate therapeutic transgenes. Our study aims to address our hypothesis that incorporating KillerRed, the first fully genetically encoded photosensitizing fluorescent protein, into oHSV will establish photodynamic oncolytic virotherapy that enhances tumoricidal efficacy as a novel treatment approach to CNS neoplasms. METHOD The optical properties of the intracellular KillerRed protein expressed in cells were determined by scanning by a multi-mode microplate reader to determine the optimal irradiation wavelength. In vitro efficacy of KillerRed-mediated PDT was tested using human glioblastoma and malignant meningioma cell lines. oHSV G47delta expressing KillerRed was constructed by a bacterial artificial chromosome-based method. KillerRed-transduced cells were confirmed to express red fluorescence, followed by irradiation by an amber color LED. Cell death and viability were assessed by DAPI staining and MTS assay, respectively. ROS generation post light treatment was assessed by DCF-DA cellular ROS assay. RESULTS KillerRed had an excitation peak at 580-585nm in transduced cells. Light irradiation by an amber LED after infection with G47delta-KillerRed induced increased cell growth inhibition and death compared with virus infection without light or light alone. Increased ROS production was observed following KillerRed PDT. CONCLUSION G47delta-KillerRed enables a combination of oncolytic virus therapy and PDT to augment tumor killing. This approach is being tested in in vivo mouse models using potent focused laser irradiation.


Sign in / Sign up

Export Citation Format

Share Document