Abstract

Simple SummaryThe primary issue of adoptive cell therapy is the poor in vivo persistence. In this context, it is necessary to clarify the fundamental mechanisms of T cell dysfunction. Here we review common dysfunctional states, including exhaustion and senescence, and discuss the challenges associated with phenotypical characterization of these T cell subsets. We overview the heterogeneity among exhausted T cells as well as mechanisms by which T cells get reinvigorated by checkpoint inhibitors. We emphasize that some cancers not responding to such treatment may activate distinct T cell dysfunction programs. Finally, we describe the dysfunction-promoting mechanisms specific for CAR-T cells and the ways to mitigate them. Immunotherapy using chimeric antigen receptor (CAR) T cells is a promising option for cancer treatment. However, T cells and CAR-T cells frequently become dysfunctional in cancer, where numerous evasion mechanisms impair antitumor immunity. Cancer frequently exploits intrinsic T cell dysfunction mechanisms that evolved for the purpose of defending against autoimmunity. T cell exhaustion is the most studied type of T cell dysfunction. It is characterized by impaired proliferation and cytokine secretion and is often misdefined solely by the expression of the inhibitory receptors. Another type of dysfunction is T cell senescence, which occurs when T cells permanently arrest their cell cycle and proliferation while retaining cytotoxic capability. The first section of this review provides a broad overview of T cell dysfunctional states, including exhaustion and senescence; the second section is focused on the impact of T cell dysfunction on the CAR-T therapeutic potential. Finally, we discuss the recent efforts to mitigate CAR-T cell exhaustion, with an emphasis on epigenetic and transcriptional modulation.

Highlights

  • Cellular immunity dysfunction is a hallmark of cancer—a result of the disease’s natural evolution toward counteracting the immune pressure

  • argininosuccinate synthase (ASS)+ornithine transcarbamylase (OTC) enhanced proliferation of chimeric antigen receptor (CAR)-T; derived from multiple human donors, regardless of scFv in vitro, SkoV3-Her2+ reduced oxidative state, both basal and upon activation, enhanced proliferation and preserved target cell lysis in presence of H2O2 in vivo, NSG mice + D270, Hu08-BBz CAR-T cells showed enhanced tumor reduction when used in combination with anti-CTLA-4, whereas 2173BBz CAR-T cells did not benefit from CTLA-4 checkpoint blockade in vitro, HPAFII and CFPAC cells showed a reduction in survival, MiaPaCa2 cells showed no difference in survival; Celecoxib did not change the efficacy of CAR-T cells in vitro, Raji prolonged proliferation and inhibited cell senescence; in vivo, NPG/Vst mice + Raji, improved persistence, proliferation, and long-term antitumor effects in vivo, SCID-Beige mice + Raji/Raji-IDO

  • Immunotherapy and adoptive cell transfer have become the standard of care for patients with certain B-cell hematological malignancies and are included in the international treatment guidelines

Read more

Summary

Introduction

Cellular immunity dysfunction is a hallmark of cancer—a result of the disease’s natural evolution toward counteracting the immune pressure. Since the microenvironment varies significantly among different types of cancer, tumor-infiltrating lymphocytes will adopt tumor-specific dysfunctional states that, while similar to T cell exhaustion, possess distinct characteristics. A specific population of terminally differentiated CD8+ Tex superior in viral or tumor control was reported [43,44,45] It resembles short-lived effector cells (SLECs) from acute infection, forms in the presence of IL-21, is distinct from dysfunctional PD-1hi terminally CD8+ Tex, has higher cytolytic activity and cytokine secretion, and is characterized by increased expression of CX3CR1, KLRG1, T-bet, and Zeb-2. A recent study revealed that the dysfunction program might depend on TCR affinity with high-affinity clones adopting a classical exhaustion state, and low-affinity cells developing distinct dysfunction mechanism of “functional inertness” [63] This observation further supports the broader landscape of cancer-associated dysfunction. Though largely ignored by the scientific community, CD4+ T cell exhaustion is essential and should be considered when developing novel immunotherapies

How to Combat Exhaustion?
T cell Senescence
How to Identify Exhausted and Senescent T Cells
Dysfunctional States of CAR-T Cells
Adjusting Regulatory Networks to Counteract T Cell Dysfunction
Senescence in CAR-T Cells
Findings
Concluding Remarks
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