Abstract

Simple SummaryCancer immunotherapy has now attracted much attention because of the recent success of immune checkpoint inhibitors. However, they are only beneficial in a limited fraction of patients most probably due to lack of sufficient CD8+ cytotoxic T-lymphocytes against tumor antigens in the host. In this regard, dendritic cells are useful tools to induce host immune responses against exogenous antigens. In particular, recently characterized cross-presenting dendritic cells are capable of inducing CD8+ cytotoxic T-lymphocytes against exogenous antigens such as tumor antigens and uniquely express the chemokine receptor XCR1. Here we focus on the recent progress in DC-based cancer vaccines and especially the use of the XCR1 and its ligand XCL1 axis for the targeted delivery of cancer vaccines to cross-presenting dendritic cells. Cancer immunotherapy aims to treat cancer by enhancing cancer-specific host immune responses. Recently, cancer immunotherapy has been attracting much attention because of the successful clinical application of immune checkpoint inhibitors targeting the CTLA-4 and PD-1/PD-L1 pathways. However, although highly effective in some patients, immune checkpoint inhibitors are beneficial only in a limited fraction of patients, possibly because of the lack of enough cancer-specific immune cells, especially CD8+ cytotoxic T-lymphocytes (CTLs), in the host. On the other hand, studies on cancer vaccines, especially DC-based ones, have made significant progress in recent years. In particular, the identification and characterization of cross-presenting DCs have greatly advanced the strategy for the development of effective DC-based vaccines. In this review, we first summarize the surface markers and functional properties of the five major DC subsets. We then describe new approaches to induce antigen-specific CTLs by targeted delivery of antigens to cross-presenting DCs. In this context, the chemokine receptor XCR1 and its ligand XCL1, being selectively expressed by cross-presenting DCs and mainly produced by activated CD8+ T cells, respectively, provide highly promising molecular tools for this purpose. In the near future, CTL-inducing DC-based cancer vaccines may provide a new breakthrough in cancer immunotherapy alone or in combination with immune checkpoint inhibitors.

Highlights

  • Cancer is the major cause of death worldwide

  • Recombinant mouse XCL1 fused with the red fluorescent protein further confirmed that XC chemokine receptor 1 (XCR1) was selectively expressed by CD103+ cDC1s in the skin and migratory CD103+ cDC1s in the draining lymph nodes [157]

  • XCL1-V21C/A59C induced accumulation of XCR1+CD103+ cDC1s in the injection site and migration to draining lymph nodes and left migratory XCR1+CD103+ cDC1s in the draining lymph nodes for a prolonged period of time compared with toll-like receptors (TLRs) ligands

Read more

Summary

Introduction

Cancer is the major cause of death worldwide. surgery, radiation, and chemotherapy represent the three pillars of cancer therapy, the prognosis still remains poor in advanced stages of cancer often with metastases [1]. Because DCs were known to play a pivotal role in the induction of various immune responses, including CD8+ CTL responses, ex vivo-derived moDCs were tried as DC-based cancer vaccines, but clinical outcomes were poor, possibly due to a limited capacity of in vitro-derived DCs to induce CD8+ CTL responses. Because recent studies have identified cDC1s as the key cross-presenting DC subset capable of inducing CD8+ CTLs [32,33,34], efforts have been focused on how to target cDC1s in antigen delivery. DEC-205 and CLEC9A are promising surface molecules for targeted delivery of antigens to cDC1s, the vaccines based on these molecules require adjuvants for the efficient induction of CD8+ CTL responses.

Differential Expression of Chemokine Receptors by DC Subsets
Use of Chemokines to Target DCs
Use of Fusion Antigens Targeting XCR1 as CTL-inducing Vaccines
Immunization Method Injection Injection
Conclusions
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call