Abstract

Immune checkpoint inhibitors (ICI) revolutionized the field of immuno-oncology and opened new avenues towards the development of novel assets to achieve durable immune control of cancer. Yet, the presence of tumor immune evasion mechanisms represents a challenge for the development of efficient treatment options. Therefore, combination therapies are taking the center of the stage in immuno-oncology. Such combination therapies should boost anti-tumor immune responses and/or target tumor immune escape mechanisms, especially those created by major players in the tumor microenvironment (TME) such as tumor-associated macrophages (TAM). Natural killer (NK) cells were recently positioned at the forefront of many immunotherapy strategies, and several new approaches are being designed to fully exploit NK cell antitumor potential. One of the most relevant NK cell-activating receptors is NKG2D, a receptor that recognizes 8 different NKG2D ligands (NKG2DL), including MICA and MICB. MICA and MICB are poorly expressed on normal cells but become upregulated on the surface of damaged, transformed or infected cells as a result of post-transcriptional or post-translational mechanisms and intracellular pathways. Their engagement of NKG2D triggers NK cell effector functions. Also, MICA/B are polymorphic and such polymorphism affects functional responses through regulation of their cell-surface expression, intracellular trafficking, shedding of soluble immunosuppressive isoforms, or the affinity of NKG2D interaction. Although immunotherapeutic approaches that target the NKG2D-NKG2DL axis are under investigation, several tumor immune escape mechanisms account for reduced cell surface expression of NKG2DL and contribute to tumor immune escape. Also, NKG2DL polymorphism determines functional NKG2D-dependent responses, thus representing an additional challenge for leveraging NKG2DL in immuno-oncology. In this review, we discuss strategies to boost MICA/B expression and/or inhibit their shedding and propose that combination strategies that target MICA/B with antibodies and strategies aimed at promoting their upregulation on tumor cells or at reprograming TAM into pro-inflammatory macrophages and remodeling of the TME, emerge as frontrunners in immuno-oncology because they may unleash the antitumor effector functions of NK cells and cytotoxic CD8 T cells (CTL). Pursuing several of these pipelines might lead to innovative modalities of immunotherapy for the treatment of a wide range of cancer patients.

Highlights

  • The therapeutic alternatives to treat tumors received a formidable boost when immune checkpoint inhibitors (ICI) were developed [1]

  • Overall, integrating data about the effect of histone deacetylase inhibitors (HDACi) on MICA/B expression, on NKG2D expression and on natural killer (NK) cell effector functions, and taking into consideration the few HDACi approved for the treatment of human patients, vorinostsat, belinostat and entinostat emerge as frontrunners to be used to induced heightened expression of MICA/B and sensitize tumor cells to anti-MICA/B Ab

  • ICD inducers might be combined with anti-MICA/B Ab to enhance antibodydependent cell-mediated cytotoxicity (ADCC) against MICA/B expressed on tumor cells mediated by NK cells and to prevent their exhaustion through an effect that involves the remodeling of the tumor microenvironment (TME) with an associated repolarization of tumor-associated macrophages (TAM)

Read more

Summary

Introduction

The therapeutic alternatives to treat tumors received a formidable boost when immune checkpoint inhibitors (ICI) were developed [1].

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