cowpea mosaic virus
Recently Published Documents


TOTAL DOCUMENTS

336
(FIVE YEARS 26)

H-INDEX

48
(FIVE YEARS 5)

Author(s):  
Edward C. Koellhoffer ◽  
Chenkai Mao ◽  
Veronique Beiss ◽  
Lu Wang ◽  
Steven N. Fiering ◽  
...  

Biomaterials ◽  
2021 ◽  
pp. 120914
Author(s):  
Chenkai Mao ◽  
Veronique Beiss ◽  
Jennifer Fields ◽  
Nicole F. Steinmetz ◽  
Steven Fiering

Cancers ◽  
2021 ◽  
Vol 13 (4) ◽  
pp. 627
Author(s):  
Courtney T. Stump ◽  
Gregory Ho ◽  
Chenkai Mao ◽  
Frank A. Veliz ◽  
Veronique Beiss ◽  
...  

Ovarian cancer is the deadliest gynecological malignancy. Though most patients enter remission following initial interventions, relapse is common and often fatal. Accordingly, there is a substantial need for ovarian cancer therapies that prevent relapse. Following remission generated by surgical debulking and chemotherapy, but prior to relapse, resected and inactivated tumor tissue could be used as a personalized vaccine antigen source. The patient’s own tumor contains relevant antigens and, when combined with the appropriate adjuvant, could generate systemic antitumor immunity to prevent relapse. Here, we model this process in mice to investigate the optimal tumor preparation and vaccine adjuvant. Cowpea mosaic virus (CPMV) has shown remarkable efficacy as an immunostimulatory cancer therapy in ovarian cancer mouse models, so we use CPMV as an adjuvant in a prophylactic vaccine against a murine ovarian cancer model. Compared to its codelivery with tumor antigens prepared in three other ways, we show that CPMV co-delivered with irradiated ovarian cancer cells constitutes an effective prophylactic vaccine against a syngeneic model of ovarian cancer in C57BL/6J mice. Following two vaccinations, 72% of vaccinated mice reject tumor challenges, and all those mice survived subsequent rechallenges, demonstrating immunologic memory formation. This study supports remission-stage vaccines using irradiated patient tumor tissue as a promising option for treating ovarian cancer, and validates CPMV as an antitumor vaccine adjuvant for that purpose.


2021 ◽  
Author(s):  
Paul L. Chariou ◽  
Veronique Beiss ◽  
Yifeng Ma ◽  
Nicole F. Steinmetz

Cowpea mosaic virus (CPMV) is currently in the development pipeline for multiple biomedical applications, including cancer immunotherapy.


2020 ◽  
Vol 10 ◽  
Author(s):  
Kayla E. A. Duval ◽  
Robert J. Wagner ◽  
Veronique Beiss ◽  
Steven N. Fiering ◽  
Nicole F. Steinmetz ◽  
...  

IntroductionVirus and virus-like nanoparticles (VNPs) have been used for a variety of preclinical treatments, including in situ anti-cancer vaccination. The Cowpea mosaic virus (CPMV) is a VNP that has shown the ability to stimulate an anti-cancer immune response. The hypothesis of this study is two-fold: that intratumoral CPMV enhances the immunogenetic and cytotoxic response of hypofractionated radiation (15 Gy or 3 x 8 Gy), and that the effect differs between fraction regimens in the murine B16 flank melanoma model.MethodsCPMV nanoparticles were delivered intratumorally, 100 μg/tumor to B16 murine melanoma flank tumors alone, and in combination with either 15 Gy or 3 x 8 Gy (3 consecutive days). Tumors were assessed for immune and cytotoxic gene and protein expression, and cytotoxic T cell infiltration 4 days post treatment. Treatment based tumor control was assessed by a 3-fold tumor growth assay.ResultsBoth CPMV and radiation alone demonstrated the activation of a number of important immune and cytotoxic genes including natural killer cell and T cell mediated cytotoxicity pathways. However, the combination treatment activated greater expression than either treatment alone. CPMV combined with a single dose of 15 Gy demonstrated greater immune and cytotoxic gene expression, protein expression, CD8+ T cell infiltration activity, and greater tumor growth delay compared to 3 x 8 Gy with CPMV.ConclusionCPMV presents a unique and promising hypofractionated radiation adjuvant that leads to increased anti-tumor cytotoxic and immune signaling, especially with respect to the immune mediated cytotoxicity, immune signaling, and toll-like receptor signaling pathways. This improvement was greater with a single dose than with a fractionated dose.


2020 ◽  
Vol 3 (7) ◽  
pp. 4179-4187
Author(s):  
Bindi K. Patel ◽  
Chao Wang ◽  
Braulio Lorens ◽  
Alan D. Levine ◽  
Nicole F. Steinmetz ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document