[HTML][HTML] Induction of potent antitumor immunity by in situ targeting of intratumoral DCs

K Furumoto, L Soares, EG Engleman… - The Journal of clinical …, 2004 - Am Soc Clin Investig
K Furumoto, L Soares, EG Engleman, M Merad
The Journal of clinical investigation, 2004Am Soc Clin Investig
Recent reports of tumor regression following delivery of autologous tumor antigen–pulsed
DCs suggest that defective antigen presentation may play a key role in tumor escape. Here
we show in two different murine tumor models, CT26 (colon adenocarcinoma) and B16
(melanoma), that the number and activation state of intratumoral DCs are critical factors in
the host response to tumors. We used CCL20/macrophage inflammatory protein-3α (MIP-3α)
chemokine to increase the number of tumoral DCs and intratumoral injections of CG-rich …
Recent reports of tumor regression following delivery of autologous tumor antigen–pulsed DCs suggest that defective antigen presentation may play a key role in tumor escape. Here we show in two different murine tumor models, CT26 (colon adenocarcinoma) and B16 (melanoma), that the number and activation state of intratumoral DCs are critical factors in the host response to tumors. We used CCL20/macrophage inflammatory protein-3α (MIP-3α) chemokine to increase the number of tumoral DCs and intratumoral injections of CG-rich motifs (CpGs) to activate such cells. Expression of CCL20 in the tumor site attracted large numbers of circulating DCs into the tumor mass and, in the case of CT26 tumors, led to complete tumor regression. Intratumoral CpG injections, in addition to CCL20, were required to induce therapeutic immunity against B16 tumors. In this model CpG overcame tumor-mediated inhibition of DC activation and enabled tumoral DCs to cross-present tumor antigens to naive CD8 T cells. CpG activation of tumoral DCs alone was not sufficient to induce tumor regression in either tumor model, nor was systemic delivery of the DC growth factor, Flt3 ligand, which dramatically increased the number of circulating DCs but not the number of tumoral DCs. These results indicate that the number of tumoral DCs as well as the tumor milieu determines the ability of tumor-bearing hosts to mount an effective antitumor immune response. Our results also suggest that DCs can be manipulated in vivo without delivery of defined tumor antigens to induce a specific T cell–mediated antitumor response and provide the basis for the use of chemokines in DC-targeted clinical strategies.
The Journal of Clinical Investigation