Myocarditis, pathologic inflammation of the heart, is a serious cause of sudden cardiac death affecting patients of all age groups. Immune checkpoint inhibitors (ICIs) are monoclonal antibodies to cytotoxic T-cell antigen-4 (CTLA-4) or programmed death-1 (PD-1)/programmed death-1 ligand (PD-1L) used as novel cancer therapeutics to release intrinsic brakes on T-cell cytotoxicity against tumor cells. Although ICIs are now relied upon to treat many advanced cancers, fulminant myocarditis has been reported as a life-threatening side effect of these drugs, leading to severe arrhythmias, heart failure and death. Under histopathology, an acute lymphocytic infiltrate is found in the heart, and multiple lines of evidence point to a T-cell and antigen-mediated phenomenon. Although T-cell clonal analysis of patient heart tissues suggest the existence of a cardiac-specific antigen in ICI-induced myocarditis, the identity of such antigen(s) remains elusive. Understanding the culprit antigens in this disease may lead to novel insights on the mechanism of T-cell mediated myocardial damage. I hypothesize that ICI-induced myocarditis is an autoimmune disorder caused by cardiac-specific auto- antigens that trigger the activation and clonal expansion of T-cells, leading to myocardial inflammation. The goal of this study is to use an advanced immunophenotyping technique called CyTOF to characterize T-cell subsets, and then use single-cell T-cell receptor sequencing and a novel computational algorithm called GLIPH (Grouping Lymphocyte Interactions by Paratope Hotspots) to identify specific epitopes responsible for T-cell activation in ICI- induced myocardial damage in mice and humans. By completing this project, I will uncover key insights into the biological mechanism of ICI-induced myocarditis which will bridge a major knowledge gap regarding the role of T-cells in mediating myocardial damage. My results will answer the key question of which immune cell types play a dominant role in ICI-induced myocarditis, as well as identify disease-causing antigens causing cardiotoxicity. There is increasing evidence to suggest that immune-mediated cardiac damage may be at the heart of many major cardiovascular diseases, ranging from autoimmune myocarditis to heart failure to atherosclerosis. My long-term goal as a physician-scientist is to elucidate mechanisms of pathological inflammation in the heart to help cure cardiovascular disease and become a leader in the field of cardiac inflammation.

Public Health Relevance

Myocarditis, a serious inflammatory reaction in the heart causing cardiac death, can occur as a result of immune checkpoint inhibitor therapy. Immune checkpoint inhibitors (ICIs) are a novel class of cancer therapeutics which release the brakes on T-cell cytotoxicity against tumor cells but may also lead to T-cell mediated myocarditis. This project aims to define the precise T-cell subsets in ICI-induced myocarditis and uncover the culprit antigens in this disease, thus paving the way to prevention and cure of this serious side effect of ICIs and helping ensure the cardiovascular safety of cancer patients.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Postdoctoral Individual National Research Service Award (F32)
Project #
1F32HL149188-01
Application #
9833406
Study Section
Special Emphasis Panel (ZRG1)
Program Officer
Meadows, Tawanna
Project Start
2019-08-14
Project End
2022-08-13
Budget Start
2019-08-14
Budget End
2020-08-13
Support Year
1
Fiscal Year
2019
Total Cost
Indirect Cost
Name
Stanford University
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
009214214
City
Stanford
State
CA
Country
United States
Zip Code
94305