Abstract

Neoantigens are the main targets of tumor-specific T cells reactivated by immune checkpoint-blocking antibodies or when using tumor-infiltrating T cells for adoptive therapy. While cancers often accumulate hundreds of mutations and harbor several immunogenic neoantigens, the repertoire of mutation-specific T cells in patients might be restricted. To bypass suboptimal conditions, which impede the reactivation of existing T cells or the priming of neoantigen-specific T cells in a patient, we employ T cells of healthy donors with an overlapping HLA repertoire to target cancer neoantigens. In this study, we focus on streamlining the process of in vitro-induction of neoantigen-specific T cells and isolating their T cell receptors (TCRs) to establish a time-efficient protocol that will allow the patient to benefit from subsequent therapy. We first optimized the priming of T cells to omit multiple restimulations and extended culturing. Neoantigen-specific T cells were enriched using specific dextramers and next-generation sequencing was applied to determine the TCR repertoire. This allowed us to circumvent the laborious process of expanding T cell clones. Using this protocol, we successfully identified HLA-A-restricted TCRs specific for neoantigens found in an esophageal cancer cell line (TE-8) and a primary ovarian cancer. To verify TCR specificity, we generated TCR-engineered T cells and confirmed recognition of the tumor-derived neoantigens. Our results also emphasize the importance of neoepitope selection in order to avoid cross-reactivity to corresponding wild-type peptide sequences. In conclusion, we established a 2-week protocol for generating and identifying neoantigen-specific TCRs from third-party donors making this strategy applicable for clinical use.

Highlights

  • Cancer immunotherapies have revolutionized and improved cancer treatment through boosting the patients’ anti-tumor immune responses

  • DPY19L4L143F T cell receptor (TCR)-engineered T cells co-cultured with human leucocyte antigen (HLA)-A*24:02- or mock-transfected TE-8 cells

  • Several types of immune checkpoint inhibitor emerged as a novel cancer treatment after the first approval of a fully humanized antibody against cytotoxic T-lymphocyteassociated protein 4 (CTLA-4) for treatment of advanced melanoma and showed significant survival benefit in various types of cancer [2, 29]

Read more

Summary

Introduction

Cancer immunotherapies have revolutionized and improved cancer treatment through boosting the patients’ anti-tumor immune responses. The success of immune checkpoint inhibitors has highlighted the importance of an effective anti-cancer immune response in cancer patients. Several studies of cancer immunotherapy revealed that T cells recognizing cancer-specific antigens (neoantigens and shared antigens) may have significant impact on the clinical response. The higher numbers of somatic mutations may increase a chance to generate highly immunogenic neoantigens that induce/ activate T cells, which are thought to drive the clinical effects of immune checkpoint inhibitors [1, 3,4,5]. The success of adoptive therapy using autologous tumor-infiltrating lymphocytes (TILs) was correlated with the presence of T cells targeting cancer-specific antigens [9]

Methods
Results
Conclusion
Full Text
Published version (Free)

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