Abstract

Successful outcome of immune checkpoint blockade in patients with solid cancers is in part associated with a high tumor mutational burden (TMB) and the recognition of private neoantigens by T-cells. The quality and quantity of target recognition is determined by the repertoire of ‘neoepitope’-specific T-cell receptors (TCRs) in tumor-infiltrating lymphocytes (TIL), or peripheral T-cells. Interferon gamma (IFN-γ), produced by T-cells and other immune cells, is essential for controlling proliferation of transformed cells, induction of apoptosis and enhancing human leukocyte antigen (HLA) expression, thereby increasing immunogenicity of cancer cells. TCR αβ-dependent therapies should account for tumor heterogeneity and availability of the TCR repertoire capable of reacting to neoepitopes and functional HLA pathways. Immunogenic epitopes in the tumor-stroma may also be targeted to achieve tumor-containment by changing the immune-contexture in the tumor microenvironment (TME). Non protein-coding regions of the tumor-cell genome may also contain many aberrantly expressed, non-mutated tumor-associated antigens (TAAs) capable of eliciting productive anti-tumor immune responses. Whole-exome sequencing (WES) and/or RNA sequencing (RNA-Seq) of cancer tissue, combined with several layers of bioinformatic analysis is commonly used to predict possible neoepitopes present in clinical samples. At the ImmunoSurgery Unit of the Champalimaud Centre for the Unknown (CCU), a pipeline combining several tools is used for predicting private mutations from WES and RNA-Seq data followed by the construction of synthetic peptides tailored for immunological response assessment reflecting the patient’s tumor mutations, guided by MHC typing. Subsequent immunoassays allow the detection of differential IFN-γ production patterns associated with (intra-tumoral) spatiotemporal differences in TIL or peripheral T-cells versus TIL. These bioinformatics tools, in addition to histopathological assessment, immunological readouts from functional bioassays and deep T-cell ‘adaptome’ analyses, are expected to advance discovery and development of next-generation personalized precision medicine strategies to improve clinical outcomes in cancer in the context of i) anti-tumor vaccination strategies, ii) gauging mutation-reactive T-cell responses in biological therapies and iii) expansion of tumor-reactive T-cells for the cellular treatment of patients with cancer.

Highlights

  • ‘Personalized immunotherapy is all the rage, but neoantigen discovery and validation remains a daunting problem’ echoed an Editorial in Nature Biotechnology 2017 [1]

  • Advances in next-generation sequencing (NGS) techniques such as whole exome sequencing (WES) and RNA-seq form the bedrock of personalized precision medicine in neoantigen-directed immuno-oncology [22, 23]

  • Treatments affecting the activity of cancer-associated fibroblasts (CAF) or tumorassociated macrophages (TAM) redirect T-cells to these nominal target cells which appear to be associated with improved anti-tumor responses in a clinical setting [31,32,33,34,35,36]

Read more

Summary

Introduction

‘Personalized immunotherapy is all the rage, but neoantigen discovery and validation remains a daunting problem’ echoed an Editorial in Nature Biotechnology 2017 [1]. Building on the therapeutic value of targeting cancer-associated mutations, mutation-directed cancer immunotherapy based on PCVs represent a highly specialized approach to induce clinically relevant and tailored anti-tumor immune responses in patients with advanced malignancies [54, 58, 59].

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