The microtubule-depolymerizing agent ansamitocin P3 programs dendritic cells toward enhanced anti-tumor immunity

K Martin, P Müller, J Schreiner, SS Prince… - Cancer Immunology …, 2014 - Springer
K Martin, P Müller, J Schreiner, SS Prince, D Lardinois, VA Heinzelmann-Schwarz
Cancer Immunology, Immunotherapy, 2014Springer
In addition to direct tumor cell cytotoxicity, chemotherapy can mediate tumor reduction
through immune modulation of the tumor microenvironment to promote anti-tumor immunity.
Mature dendritic cells (DCs) play key roles in priming robust immune responses in tumor-
bearing hosts. Here, we screened a panel of 21 anticancer agents with defined molecular
targets for their ability to induce direct maturation of DCs. We identified ansamitocin P3, a
microtubule-depolymerizing agent, as a potent inducer of phenotypic and functional …
Abstract
In addition to direct tumor cell cytotoxicity, chemotherapy can mediate tumor reduction through immune modulation of the tumor microenvironment to promote anti-tumor immunity. Mature dendritic cells (DCs) play key roles in priming robust immune responses in tumor-bearing hosts. Here, we screened a panel of 21 anticancer agents with defined molecular targets for their ability to induce direct maturation of DCs. We identified ansamitocin P3, a microtubule-depolymerizing agent, as a potent inducer of phenotypic and functional maturation of DCs. Exposure of both murine spleen-derived and human monocyte-derived DCs to ansamitocin P3 triggered up-regulation of maturation markers and production of pro-inflammatory cytokines, resulting in an enhanced T cell stimulatory capacity. Local administration of ansamitocin P3 induced maturation of skin Langerhans cells in vivo and promoted antigen uptake and extensive homing of tumor-resident DCs to tumor-draining lymph nodes. When used as an adjuvant in a specific vaccination approach, ansamitocin P3 dramatically increased activation of antigen-specific T cells. Finally, we demonstrate that ansamitocin P3, due to its immunomodulatory properties, acts in synergy with antibody-mediated blockade of the T cell inhibitory receptors PD-1 and CTLA-4. The combination treatment was most effective and induced durable growth inhibition of established tumors. Mechanistically, we observed a reduced regulatory T cell frequency and improved T cell effector function at the tumor site. Taken together, our study unravels an immune-based anti-tumor mechanism exploited by microtubule-depolymerizing agents, including ansamitocin P3, and paves the way for future clinical trials combining this class of agents with immunotherapy.
Springer