Abstract

Immune checkpoint inhibitors have gained an established role in the treatment of different tumors. Indeed, their use has dramatically changed the landscape of cancer care, especially for tumor types traditionally known to have poor outcomes. However, stimulating anticancer immune responses may also elicit an unusual pattern of immune-related adverse events (irAEs), different from those of conventional chemotherapy, likely due to a self-tolerance impairment featuring the production of autoreactive lymphocytes and autoantibodies, or a non-specific autoinflammatory reaction. Ionizing radiation has proven to promote both positive pro-inflammatory and immunostimolatory activities, and negative anti-inflammatory and immunosuppressive mechanisms, as a result of cross-linked interactions among radiation dose, the tumor microenvironment and the host genetic predisposition. Several publications argue in favor of combining immunotherapy and a broad range of radiation schedules, based on the recent evidence of superior treatment responses and patient survival. The synergistic modulation of the immune response by radiation therapy and immunotherapeutics, particularly those manipulating T-cell activation, may also affect the type and severity of irAEs, suggesting a relationship between the positive antitumor and adverse autoimmune effects of these agents. As yet, information on factors that may help to predict immune toxicity is still lacking. The aim of our work is to provide an overview of the biological mechanisms underlying irAEs and possible crosslinks with radiation-induced anticancer immune responses. We believe such an overview may support the optimization of immunotherapy and radiotherapy as essential components of multimodal anticancer therapeutic approaches. Challenges in translating these to clinical practice are discussed.

Highlights

  • The advent of immune checkpoint inhibitors (ICIs) has dramatically changed the landscape of cancer care, since immunotherapeutic strategies are emerging as potentially curative systemic therapy for several tumors, especially for tumor types traditionally known to have poor outcomes

  • Increased circulating levels of IL-6 and C-reactive protein (CRP) have been recorded in patients with different tumor types developing a broad range of immune-related adverse events (irAEs), and a synergistic antitumor activity has been demonstrated in mouse models combining ICIs with inhibition of the IL-6 axis (Esfahani et al, 2020a)

  • Distinct human leukocyte antigen (HLA) haplotypes, and polymorphisms in immunoregulatory genes such as Cytotoxic T-lymphocyte–associated protein 4 (CTLA-4) and Programmed cell death 1 (PD-1) have been associated with a variety of classical autoimmune diseases, and likely play an important role in the development of irAEs (e.g., HLA-DQ8 and HLA-DR53 for lymphocytic hypophysitis), and the predominance of HLA-DR4 among patients treated with PD1- or programmed death-ligand 1 (PD-L1)-directed ICIs developing autoimmune insulindependent diabetes (Khan et al, 2018))

Read more

Summary

INTRODUCTION

The advent of immune checkpoint inhibitors (ICIs) has dramatically changed the landscape of cancer care, since immunotherapeutic strategies are emerging as potentially curative systemic therapy for several tumors, especially for tumor types traditionally known to have poor outcomes. Combination strategies (PD-1 or PD-L1 agents with other immunotherapy agents such as anti-CTLA-4 antibodies, molecular targeted therapy, vaccines, chemotherapies, radiotherapy, or chemoradiotherapies) may improve outcomes, as supported by recent evidence of superior treatment responses and patient survival. Several factors may influence the development of irAEs during treatment with ICIs, including the cancer type, molecular target PD-L1), single agent administration or combined checkpoint blockade and/or sequential use of different types of ICIs. Treatment duration and prior chemotherapy likely influence immunotherapy tolerance, together with underlying host factors such as the patient’s age, genetic predisposition to autoimmunity and/or pre-existing autoimmune disorders, and the host microbiome (Weinmann and Pisetsky, 2019). The aim of our work was to provide an overview of the current evidence on the biological bases underlying irAEs, and to speculate about possible crosslinks with radiation-induced anticancer immune responses

THE BALANCE BETWEEN SELF TOLERANCE AND ANTICANCER IMMUNE RESPONSE
PROPOSED IMMUNOPATHOGENIC MECHANISMS FOR irAEs
Adaptive and Innate Immunity
Autoinflammatory Mechanisms
Genetic Susceptibilities
The Role of the Microbiome
RADIOTHERAPY AND IMMUNOTHERAPY
POTENTIAL MARKERS PREDICTIVE FOR irAEs
Nonspecific biomarkers
DISCUSSION
Findings
AUTHOR CONTRIBUTIONS
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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.