scispace - formally typeset
Open AccessJournal ArticleDOI

Cardiotoxicity of immune checkpoint inhibitors

TLDR
The mechanisms of the most prominent checkpoint inhibitors are described, specifically ipilimumab (anti-CTLA-4, the godfather of checkpoint inhibitors) patient and monoclonal antibodies targeting PD-1 and PD-L1 (eg, atezolizumab).
Abstract
Cardiac toxicity after conventional antineoplastic drugs (eg, anthracyclines) has historically been a relevant issue. In addition, targeted therapies and biological molecules can also induce cardiotoxicity. Immune checkpoint inhibitors are a novel class of anticancer drugs, distinct from targeted or tumour type-specific therapies. Cancer immunotherapy with immune checkpoint blockers (ie, monoclonal antibodies targeting cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), programmed cell death 1 (PD-1) and its ligand (PD-L1)) has revolutionised the management of a wide variety of malignancies endowed with poor prognosis. These inhibitors unleash antitumour immunity, mediate cancer regression and improve the survival in a percentage of patients with different types of malignancies, but can also produce a wide spectrum of immune-related adverse events. Interestingly, PD-1 and PD-L1 are expressed in rodent and human cardiomyocytes, and early animal studies have demonstrated that CTLA-4 and PD-1 deletion can cause autoimmune myocarditis. Cardiac toxicity has largely been underestimated in recent reviews of toxicity of checkpoint inhibitors, but during the last years several cases of myocarditis and fatal heart failure have been reported in patients treated with checkpoint inhibitors alone and in combination. Here we describe the mechanisms of the most prominent checkpoint inhibitors, specifically ipilimumab (anti-CTLA-4, the godfather of checkpoint inhibitors) patient and monoclonal antibodies targeting PD-1 (eg, nivolumab, pembrolizumab) and PD-L1 (eg, atezolizumab). We also discuss what is known and what needs to be done about cardiotoxicity of checkpoint inhibitors in patients with cancer. Severe cardiovascular effects associated with checkpoint blockade introduce important issues for oncologists, cardiologists and immunologists.

read more

Citations
More filters
Journal ArticleDOI

CD8+ cytotoxic T lymphocytes in cancer immunotherapy: A review.

TL;DR: CD8 + T cell priming is directed essentially as a corroboration work between cells of innate immunity including dendritic cells (DCs) and natural killer (NK) cells with CD4 + T cells in adoptive immunity for making durable and efficient antitumor immune responses.
Journal ArticleDOI

Immune checkpoint inhibitors and cardiovascular toxicity.

TL;DR: The epidemiology of immune checkpoint inhibitor-mediated cardiotoxic effects, as well as their clinical presentation, subtypes, risk factors, pathophysiology, and clinical management, are discussed, including the introduction of a new surveillance strategy.
References
More filters
Journal ArticleDOI

Cancer Immunotherapy Trials Not Immune from Imprecise Selection of Patients

TL;DR: A trial that compared the PD-1 inhibitor nivolumab with chemotherapy as first-line treatment for advanced NSCLC in a population of patients selected on the basis of PD-L1 expression level showed improvement in survival and progression-free survival.
Journal ArticleDOI

Chemotherapy-induced cardiotoxicity: importance of early detection.

TL;DR: The most clinically impacting manifestation, feared both by cardiologists and by oncologists, is the development of left ventricular dysfunction (LVD), which seriously limits patients’ therapeutic opportunities when adjunctive therapy for cancer relapse or persistence is required.
Journal ArticleDOI

Cancer immunotherapy innovator James Allison receives the 2015 Lasker~DeBakey Clinical Medical Research Award

TL;DR: James P. Allison has caused a paradigm shift in the treatment of cancer in which treatment is focused on helping the patient’s immune system defeat the tumor instead of targeting the tumor itself.
Related Papers (5)