Please use this identifier to cite or link to this item: http://hdl.handle.net/123456789/2292
DC FieldValueLanguage
dc.contributor.authorNordin, M. L.en_US
dc.contributor.authorNorpi A.S.M.en_US
dc.contributor.authorNg P.Y.en_US
dc.contributor.authorYusoff K.en_US
dc.contributor.authorAbu N.en_US
dc.contributor.authorLim K.P.en_US
dc.contributor.authorAzmi F.en_US
dc.date.accessioned2022-01-06T03:25:17Z-
dc.date.available2022-01-06T03:25:17Z-
dc.date.issued2021-10-01-
dc.identifier.issn20726694-
dc.identifier.urihttp://hdl.handle.net/123456789/2292-
dc.descriptionWeb of Science / Scopusen_US
dc.description.abstractBreast cancer is the most common invasive cancer diagnosed among women. A cancer vaccine has been recognized as a form of immunotherapy with a prominent position in the prevention and treatment of breast cancer. The majority of current breast cancer vaccination strategies aim to stimulate antitumor T-cell responses of the HER2/neu oncogene, which is abnormally expressed in breast cancer cells. However, the role of the B-cell humoral response is often underappreciated in the cancer vaccine design. We have advanced this idea by elucidating the role of B-cells in cancer vaccination by designing a chimeric antigenic peptide possessing both cytotoxic T lymphocytes (GP2) and B-cell (P4) peptide epitopes derived from HER2/neu. The chimeric peptide (GP2–P4) was further conjugated to a carrier protein (KLH), forming a KLH–GP2–P4 conjugate. The immunogenicity of KLH–GP2–P4 was compared with KLH–GP2 (lacking the B-cell epitope) in BALB/c mice. Mice immunized with KLH–GP2–P4 elicited more potent antigen-specific neutralizing antibodies against syngeneic TUBO cells (cancer cell line overexpressing HER2/neu) that was governed by a balanced Th1/Th2 polarization in comparison to KLH–GP2. Subsequently, these immune responses led to greater inhibition of tumor growth and longer survival in TUBO tumor-bearing mice in both prophylactic and therapeutic challenge experiments. Overall, our data demonstrated that the B-cell epitope has a profound effect in orchestrating an efficacious antitumor immunity. Thus, a multi-epitope peptide vaccine encompassing cytotoxic T-lymphocytes, T-helper and B-cell epitopes represents a promising strategy in developing cancer vaccines with a preventive and therapeutic modality for the effective management of breast cancer.en_US
dc.language.isoenen_US
dc.publisherMDPIen_US
dc.relation.ispartofCancersen_US
dc.subjectAntitumoren_US
dc.subjectB-cell epitopeen_US
dc.subjectBreast canceren_US
dc.subjectHER2/neuen_US
dc.subjectMulti-epitopeen_US
dc.subjectPeptide vaccinesen_US
dc.titleHER2/neu-based peptide vaccination-pulsed with B-cell epitope induced efficient prophylactic and therapeutic antitumor activities in TUBO breast cancer mice modelen_US
dc.typeNationalen_US
dc.identifier.doi10.3390/cancers13194958-
dc.volume13 (19)en_US
dc.description.articleno4958en_US
dc.description.typeArticleen_US
dc.description.impactfactor6.639en_US
dc.description.quartileQ1en_US
item.languageiso639-1en-
item.openairetypeNational-
item.fulltextWith Fulltext-
item.grantfulltextopen-
Appears in Collections:Faculty of Veterinary Medicine - Journal (Scopus/WOS)
Show simple item record

Google ScholarTM

Check

Altmetric

Altmetric


Items in DSpace are protected by copyright, with all rights reserved, unless otherwise indicated.