A novel potential effective strategy for enhancing the antitumor immune response in breast cancer patients using a viable cancer cell-dendritic cell-based vaccine.

Autor: Abdellateif MS; Medical Biochemistry and Molecular Biology Unit, Department of Cancer Biology, National Cancer Institute, Cairo University, Cairo 11976, Egypt., Shaarawy SM; Medical Biochemistry and Molecular Biology Unit, Department of Cancer Biology, National Cancer Institute, Cairo University, Cairo 11976, Egypt., Kandeel EZ; Department of Clinical Pathology, National Cancer Institute, Cairo University, Cairo 11976, Egypt., El-Habashy AH; Department of Pathology, National Cancer Institute, Cairo University, Cairo 11976, Egypt., Salem ML; Department of Zoology, Faculty of Science, Tanta University, Tanta, Gharbia 31511, Egypt., El-Houseini ME; Medical Biochemistry and Molecular Biology Unit, Department of Cancer Biology, National Cancer Institute, Cairo University, Cairo 11976, Egypt.
Jazyk: angličtina
Zdroj: Oncology letters [Oncol Lett] 2018 Jul; Vol. 16 (1), pp. 529-535. Date of Electronic Publication: 2018 May 04.
DOI: 10.3892/ol.2018.8631
Abstrakt: Dendritic cells (DCs) have been used in a number of clinical trials for cancer immunotherapy; however, they have achieved limited success in solid tumors. Consequently the aim of the present study was to identify a novel potential immunotherapeutic target for breast cancer patients through in vitro optimization of a viable DC-based vaccine. Immature DCs were primed by viable MCF-7 breast cancer cells and the activity and maturation of DCs were assessed through measuring CD83, CD86 and major histocompatibility complex (MHC)-II expression, in addition to different T cell subpopulations, namely CD4 + T cells, CD8 + T cells, and CD4 + CD25 + forkhead box protein 3 (Foxp3) + regulatory T cells (Tregs), by flow cytometric analysis. Foxp3 level was also measured by enzyme-linked immunosorbent assay (ELISA) in addition to reverse-transcription quantitative polymerase chain reaction. The levels of interleukin-12 (IL-12) and interferon-γ (IFN-γ) were determined by ELISA. Finally, the cytotoxicity of cytotoxic T lymphocytes (CTLs) was evaluated through measuring lactate dehydrogenase (LDH) release by ELISA. The results demonstrated that CD83 + , CD86 + and MHC-II + DCs were significantly elevated (P<0.001) following priming with breast cancer cells. In addition, there was increased activation of CD4 + and CD8 + T-cells, with a significant decrease of CD4 + CD25 + Foxp3 + Tregs (P<0.001). Furthermore, a significant downregulation of FOXP3 gene expression (P<0.001) was identified, and a significant decrease in the level of its protein following activation (P<0.001) was demonstrated by ELISA. Additionally, significant increases in the secretion of IL-12 and IFN-γ (P=0.001) were observed. LDH release was significantly increased (P<0.001), indicating a marked cytotoxicity of CTLs against cancer cells. Therefore viable breast cancer cell-DC-based vaccines could expose an innovative avenue for a novel breast cancer immunotherapy.
Databáze: MEDLINE
Nepřihlášeným uživatelům se plný text nezobrazuje