IDO vaccine ablates immune-suppressive myeloid populations and enhances antitumor effects independent of tumor cell IDO status

R Nandre, V Verma, P Gaur, V Patil, X Yang… - Cancer immunology …, 2022 - AACR
R Nandre, V Verma, P Gaur, V Patil, X Yang, Z Ramlaoui, N Shobaki, MH Andersen…
Cancer immunology research, 2022AACR
The immunosuppressive tumor microenvironment (TME) does not allow generation and
expansion of antitumor effector cells. One of the potent immunosuppressive factors present
in the TME is the indoleamine-pyrrole 2, 3-dioxygenase (IDO) enzyme, produced mainly by
cancer cells and suppressive immune cells of myeloid origin. In fact, IDO+ myeloid-derived
suppressor cells (MDSC) and dendritic cells (DC) tend to be more suppressive than their
IDO− counterparts. Hence, therapeutic approaches that would target the IDO+ cells in the …
Abstract
The immunosuppressive tumor microenvironment (TME) does not allow generation and expansion of antitumor effector cells. One of the potent immunosuppressive factors present in the TME is the indoleamine-pyrrole 2,3-dioxygenase (IDO) enzyme, produced mainly by cancer cells and suppressive immune cells of myeloid origin. In fact, IDO+ myeloid-derived suppressor cells (MDSC) and dendritic cells (DC) tend to be more suppressive than their IDO counterparts. Hence, therapeutic approaches that would target the IDO+ cells in the TME, while sparing the antigen-presenting functions of IDO myeloid populations, are needed. Using an IDO-specific peptide vaccine (IDO vaccine), we explored the possibility of generating effector cells against IDO and non-IDO tumor-derived antigens. For this, IDO-secreting (B16F10 melanoma) and non–IDO-secreting (TC-1) mouse tumor models were employed. We showed that the IDO vaccine significantly reduced tumor growth and enhanced survival of mice in both the tumor models, which associated with a robust induction of IDO-specific effector cells in the TME. The IDO vaccine significantly enhanced the antitumor efficacy of non-IDO tumor antigen–specific vaccines, leading to an increase in the number of total and antigen-specific activated CD8+ T cells (IFNγ+ and granzyme B+). Treatment with the IDO vaccine significantly reduced the numbers of IDO+ MDSCs and DCs, and immunosuppressive regulatory T cells in both tumor models, resulting in enhanced therapeutic ratios. Together, we showed that vaccination against IDO is a promising therapeutic option for both IDO-producing and non–IDO-producing tumors. The IDO vaccine selectively ablates the IDO+ compartment in the TME, leading to a significant enhancement of the immune responses against other tumor antigen–specific vaccines.
AACR