An Engineered M13 Filamentous Nanoparticle as an Antigen Carrier for a Malignant Melanoma Immunotherapeutic Strategy.

Autor: Brišar N; Faculty of Health Sciences, University of Primorska, 6310 Izola, Slovenia.; Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia., Šuster K; Valdoltra Orthopaedic Hospital, 6280 Ankaran, Slovenia., Brezar SK; Department of Experimental Oncology, Institute of Oncology Ljubljana, 1000 Ljubljana, Slovenia., Vidmar R; Department of Biochemistry, Molecular and Structural Biology, Jozef Stefan Institute, 1000 Ljubljana, Slovenia., Fonović M; Department of Biochemistry, Molecular and Structural Biology, Jozef Stefan Institute, 1000 Ljubljana, Slovenia., Cör A; Valdoltra Orthopaedic Hospital, 6280 Ankaran, Slovenia.; Faculty of Education, University of Primorska, 6310 Izola, Slovenia.
Jazyk: angličtina
Zdroj: Viruses [Viruses] 2024 Feb 01; Vol. 16 (2). Date of Electronic Publication: 2024 Feb 01.
DOI: 10.3390/v16020232
Abstrakt: Bacteriophages, prokaryotic viruses, hold great potential in genetic engineering to open up new avenues for vaccine development. Our study aimed to establish engineered M13 bacteriophages expressing MAGE-A1 tumor peptides as a vaccine for melanoma treatment. Through in vivo experiments, we sought to assess their ability to induce robust immune responses. Using phage display technology, we engineered two M13 bacteriophages expressing MAGE-A1 peptides as fusion proteins with either pVIII or pIIII coat proteins. Mice were intraperitoneally vaccinated three times, two weeks apart, using two different engineered bacteriophages; control groups received a wild-type bacteriophage. Serum samples taken seven days after each vaccination were analyzed by ELISA assay, while splenocytes harvested seven days following the second boost were evaluated by ex vivo cytotoxicity assay. Fusion proteins were confirmed by Western blot and nano-LC-MS/MS. The application of bacteriophages was safe, with no adverse effects on mice. Engineered bacteriophages effectively triggered immune responses, leading to increased levels of anti-MAGE-A1 antibodies in proportion to the administered bacteriophage dosage. Anti-MAGE-A1 antibodies also exhibited a binding capability to B16F10 tumor cells in vitro, as opposed to control samples. Splenocytes demonstrated enhanced CTL cytotoxicity against B16F10 cells. We have demonstrated the immunogenic capabilities of engineered M13 bacteriophages, emphasizing their potential for melanoma immunotherapy.
Databáze: MEDLINE
Nepřihlášeným uživatelům se plný text nezobrazuje