Val-boroPro accelerates T cell priming via modulation of dendritic cell trafficking resulting in complete regression of established murine tumors

Autor: Terry J. Fry, Meghaan P. Walsh, Martin Guimond, Jessica P. E. Davis, Shannon M. Larabee, Yongzhi Cui, William W. Bachovchin, Brynn B. Duncan, Su Young Kim, Aviva C. Krauss
Jazyk: angličtina
Rok vydání: 2013
Předmět:
CD4-Positive T-Lymphocytes
Male
Adoptive cell transfer
Mouse
medicine.medical_treatment
T-Lymphocytes
Priming (immunology)
lcsh:Medicine
CD8-Positive T-Lymphocytes
Immunotherapy
Adoptive

Mice
Neoplasms
Interferon gamma
lcsh:Science
Vaccines
Multidisciplinary
biology
Vaccination
Remission Induction
Animal Models
Dipeptides
Boronic Acids
medicine.anatomical_structure
Oncology
Medicine
Cytokines
Female
Immunotherapy
Antibody
Chemokines
Cancer Prevention
medicine.drug
Research Article
T cell
Immune Cells
Immunology
Antigen-Presenting Cells
Cancer Vaccines
Immune system
Model Organisms
Adjuvants
Immunologic

Cell Line
Tumor

Vaccine Development
medicine
Animals
Biology
lcsh:R
Immunity
Dendritic cell
Dendritic Cells
Disease Models
Animal

Cancer research
biology.protein
Clinical Immunology
lcsh:Q
Lymph Nodes
Zdroj: PLoS ONE, Vol 8, Iss 3, p e58860 (2013)
PLoS ONE
ISSN: 1932-6203
Popis: Although tumors naturally prime adaptive immune responses, tolerance may limit the capacity to control progression and can compromise effectiveness of immune-based therapies for cancer. Post-proline cleaving enzymes (PPCE) modulate protein function through N-terminal dipeptide cleavage and inhibition of these enzymes has been shown to have anti-tumor activity. We investigated the mechanism by which Val-boroPro, a boronic dipeptide that inhibits post-proline cleaving enzymes, mediates tumor regression and tested whether this agent could serve as a novel immune adjuvant to dendritic cell vaccines in two different murine syngeneic murine tumors. In mice challenged with MB49, which expresses the HY antigen complex, T cell responses primed by the tumor with and without Val-boroPro were measured using interferon gamma ELISPOT. Antibody depletion and gene-deficient mice were used to establish the immune cell subsets required for tumor regression. We demonstrate that Val-boroPro mediates tumor eradication by accelerating the expansion of tumor-specific T cells. Interestingly, T cells primed by tumor during Val-boroPro treatment demonstrate increased capacity to reject tumors following adoptive transfer without further treatment of the recipient. Val-boroPro -mediated tumor regression requires dendritic cells and is associated with enhanced trafficking of dendritic cells to tumor draining lymph nodes. Finally, dendritic cell vaccination combined with Val-boroPro treatment results in complete regression of established tumors. Our findings demonstrate that Val-boroPro has antitumor activity and a novel mechanism of action that involves more robust DC trafficking with earlier priming of T cells. Finally, we show that Val-boroPro has potent adjuvant properties resulting in an effective therapeutic vaccine.
Databáze: OpenAIRE