1
|
Zhu S, Fu K, Li S, Yang C, Pan C, Wang X, Wang F, Yu X, To KKW, Fu L. Cardiotoxicity of small-molecule kinase inhibitors in cancer therapy. Exp Hematol Oncol 2025; 14:68. [PMID: 40346640 PMCID: PMC12063284 DOI: 10.1186/s40164-025-00660-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 04/22/2025] [Indexed: 05/11/2025] Open
Abstract
Cancer is one of the leading causes of death worldwide. Recent advances in precision oncology have enabled many specific cancer patient populations to respond well and achieve longer survival with small-molecule kinase inhibitors, which have become a new therapeutic strategy for tumors. Since 2001, the Food and Drug Administration has approved 108 and 63 new anticancer drugs for treating solid tumors and hematological malignancies, respectively, 89 of which belong to the large group of small-molecule kinase inhibitors (SMKIs). Compared to conventional chemotherapeutic agents such as cyclophosphamide, doxorubicin, and 5-FU, SMKIs offer better efficacy with fewer toxic side effects. Nevertheless, with the development of more novel SMKIs and their wider clinical application to a larger population of cancer patients, variable degrees of cardiotoxic adverse events have emerged for some SMKIs during cancer therapy. This review comprehensively summarizes the most updated progress in the cardiotoxicity of SMKIs in cancer therapy and discusses the new findings and mechanisms, which will provide emerging strategies for the prevention of cardiotoxicity caused by small molecule targeted drugs and the design of the next generation of low cardiotoxicity targeted drugs.
Collapse
Affiliation(s)
- Shuangli Zhu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Kai Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Sijia Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Chuan Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Can Pan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Xueping Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Fang Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Xiyong Yu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangdong, Guangzhou Medical University, Guangzhou, 511436, China
| | - Kenneth Kin Wah To
- School of Pharmacy, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Liwu Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China.
| |
Collapse
|
2
|
Cai JQ, Wang YM, Lin X, Xie M, Zhang G, Wei XX, Sun H. Cardiovascular toxicity of anaplastic lymphoma kinase inhibitors for patients with non-small cell lung cancer: a network meta-analysis. Future Oncol 2025; 21:1125-1135. [PMID: 39400073 DOI: 10.1080/14796694.2024.2370239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 06/17/2024] [Indexed: 10/15/2024] Open
Abstract
Aim: We conducted network meta-analysis to assess cardiovascular toxicity of anaplastic lymphoma kinase-tyrosine kinase inhibitors (ALK-TKIs).Materials & methods: Eleven articles involving 2855 patients and six interventions including crizotinib, alectinib, ceritinib, lorlatinib, brigatinib and chemotherapy were analyzed.Results: No significant difference was observed in overall cardiovascular risk among ALK-TKIs. Subgroup analysis showed that for cardiac toxicity, crizotinib and alectinib were more likely to cause myocardial rhythm abnormalities. Crizotinib and ceritinib had a higher risk of Q-T prolongation than chemotherapy. For vascular toxicity, crizotinib and ceritinib had a higher risk of thrombotic events than brigatinib. Crizotinib and lorlatinib were more likely to cause blood pressure abnormalities.Conclusion: Clinicians should carefully monitoring cardiovascular events when ALK-TKIs used in NSCLCs patients with baseline cardiovascular diseases.
Collapse
Affiliation(s)
- Jia Qin Cai
- Department of Pharmacy, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Yi Ming Wang
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Department of Pharmacy, Fuzhou, Fujian, China
| | - Xinmiao Lin
- School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Mumu Xie
- Department of Pharmacy, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Guifeng Zhang
- Department of Oncology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Xiao Xia Wei
- Department of Pharmacy, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Hong Sun
- Department of Pharmacy, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| |
Collapse
|
3
|
Perelli RM, Dewars ER, Cope H, Behura AS, Ponek AQ, Sala AM, Zhang Z, Muralidharan P, Moya-Mendez ME, Berkman A, Monaco GG, Sullivan MC, Ezekian JE, Yang Q, Sun B, Kurzlechner LM, Asokan T, Breglio AM, Jay Campbell M, Spector ZZ, Rehder CW, Tang PC, James CA, Calkins H, Shashi V, Landstrom AP. TAX1BP3 Causes TRPV4-Mediated Autosomal Recessive Arrhythmogenic Cardiomyopathy. Circ Res 2025; 136:667-684. [PMID: 39963794 PMCID: PMC11949706 DOI: 10.1161/circresaha.124.325180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 02/01/2025] [Accepted: 02/05/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND Arrhythmogenic cardiomyopathy (ACM) is one of the leading causes of sudden cardiac death in children, young adults, and athletes and is characterized by the fibro-fatty replacement of the myocardium, predominantly of the right ventricle. Sixty percent of patients with ACM have a known genetic cause, but for the remainder, the pathogenesis is unknown. This lack of mechanistic understanding has slowed the development of disease-modifying therapies, and children with ACM have a high degree of morbidity and mortality. METHODS Induced pluripotent stem cells (iPSCs) from 3 family members were differentiated into cardiac myocytes (CMs). Calcium imaging was conducted by labeling calcium with CAL-520 and confocal imaging to capture calcium sparks after iPSC-CMs were electrically paced. A cardiac-specific, inducible knockout mouse (Tax1bp3-/-) was made and intracardiac electrophysiology studies conducted to observe arrhythmia inducibility following pacing. RESULTS We identified a kindred with multiple members affected by ACM cosegregating with biallelic variants in the gene TAX1BP3, which encodes the protein TAX1BP3 (Tax1-binding protein 3). iPSC-CMs derived from this kindred demonstrated increased intracellular lipid droplets, induction of TRPV4 (transient receptor potential vanilloid type 4) expression, and inducible TRPV4 current. This was associated with depletion of the intracellular sarcoplasmic reticulum Ca2+ store and increased RyR2 (ryanodine receptor 2)-mediated store Ca2+ leak and delayed afterdepolarizations, a known mechanism of Ca2+-mediated arrhythmogenesis. Similarly, Tax1bp3 cardiac-specific knockout mice had increased Ca2+ leak and were predisposed to ventricular arrhythmias compared with wild-type mice. Ca2+ leak in both the iPSC-CMs and mouse ventricular myocytes was rescued by small molecule TRPV4 inhibition. This strategy also effectively reduced Ca2+ leak in a PKP2 (plakophilin 2) p.His773AlafsX8 iPSC-CM model of ACM. CONCLUSIONS We conclude that TAX1BP3 is associated with rare autosomal recessive ACM through TRPV4-mediated Ca2+ leak from RyR2. Further, TRPV4 current inhibition has the potential to be a new therapeutic target for ACM.
Collapse
Affiliation(s)
- Robin M. Perelli
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina, United States
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Enya R. Dewars
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
- Department of Cell and Molecular Biology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Heidi Cope
- Department of Pediatrics, Division of Pediatric Cardiology, UT Southwestern Medical Center, Dallas, Texas, United States
| | - Alexander S. Behura
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Anna Q. Ponek
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Angelina M. Sala
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Zhushan Zhang
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Padmapriya Muralidharan
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Mary E. Moya-Mendez
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Amy Berkman
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Gabrielle G. Monaco
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Molly C. Sullivan
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Jordan E. Ezekian
- Department of Pediatrics, Division of Pediatric Cardiology, UT Southwestern Medical Center, Dallas, Texas, United States
| | - Qixin Yang
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Bo Sun
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Leonie M. Kurzlechner
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Tulsi Asokan
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Andrew M. Breglio
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
- Department of Anesthesiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - M. Jay Campbell
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Zebulon Z. Spector
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Catherine W. Rehder
- Department of Pathology and Duke University Health Systems Clinical Laboratories, Durham, North Carolina, United States
| | | | - Paul C. Tang
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, Michigan, United States
| | - Cynthia A. James
- Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States
| | - Hugh Calkins
- Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States
| | - Vandana Shashi
- Department of Pediatrics, Division of Pediatric Cardiology, UT Southwestern Medical Center, Dallas, Texas, United States
| | - Andrew P. Landstrom
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina, United States
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| |
Collapse
|
4
|
Wright JC, Sharma S, Potter AS. Arrhythmia Challenges in Cardio-Oncology: High-Risk Therapies, Management, and Anticoagulation. Cardiol Clin 2025; 43:43-56. [PMID: 39551561 DOI: 10.1016/j.ccl.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Cardiovascular disease and cancer are the leading cause of mortality in the United States. In 2021, there were 695,547 and 605,213 deaths due to heart disease and cancer, respectively. With novel oncologic and cardiac therapies, survival has improved leading to increased life-expectancy albeit with chronic illness burden. Arrhythmia management in patients with cancer, whether active or in remission, can be quite challenging. In this review, we will discuss high-risk oncological therapies, prevention, and management of Atrial fibrillation, Ventricular Arrhythmias, and Bradyarrhythmias.
Collapse
Affiliation(s)
- Jonathan C Wright
- Cardio-oncology Program, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Sneha Sharma
- Cardio-oncology Program, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Division of Cardiovascular Medicine, The Ohio State University, Columbus, OH, USA
| | - Adam S Potter
- Cardio-oncology Program, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Internal Medicine, The Ohio State University, Columbus, OH, USA; Division of Cardiovascular Medicine, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
5
|
Torresan S, Bortolot M, De Carlo E, Bertoli E, Stanzione B, Del Conte A, Spina M, Bearz A. Matters of the Heart: Cardiotoxicity Related to Target Therapy in Oncogene-Addicted Non-Small Cell Lung Cancer. Int J Mol Sci 2025; 26:554. [PMID: 39859270 PMCID: PMC11765312 DOI: 10.3390/ijms26020554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/30/2024] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
The treatment of Non Small Cell Lung Cancer (NSCLC) has been revolutionised by the introduction of targeted therapies. With the improvement of response and frequently of overall survival, however, a whole new set of adverse events emerged. In fact, due to the peculiar mechanism of action of each one of the tyrosine kinase inhibitors and other targeted therapies, every drug has its own specific safety profile. In addition, this safety profile could not fully emerge from clinical trials data, as patients in clinical practice usually have more comorbidities and frailties. Cardiotoxicity is a well-known and established adverse event of anti-cancer therapies. However, only recently it has become a central topic for targeted therapies in NSCLC, due to the unknown real range and frequency. Management of this toxicity begins with prevention, and must balance the need of continuing an effective anticancer treatment versus low risk of even fatal events and the preservation of long-term quality of life. The aim of this review is to summarise the current knowledge focusing on currently used targeted therapies in NSCLC.
Collapse
Affiliation(s)
- Sara Torresan
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), Istituto di Ricovero e Cura a Carattere Scientifico, 33081 Aviano, Italy
- Department of Medicine (DME), University of Udine, 33100 Udine, Italy
| | - Martina Bortolot
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), Istituto di Ricovero e Cura a Carattere Scientifico, 33081 Aviano, Italy
- Department of Medicine (DME), University of Udine, 33100 Udine, Italy
| | - Elisa De Carlo
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), Istituto di Ricovero e Cura a Carattere Scientifico, 33081 Aviano, Italy
| | - Elisa Bertoli
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), Istituto di Ricovero e Cura a Carattere Scientifico, 33081 Aviano, Italy
| | - Brigida Stanzione
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), Istituto di Ricovero e Cura a Carattere Scientifico, 33081 Aviano, Italy
| | - Alessandro Del Conte
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), Istituto di Ricovero e Cura a Carattere Scientifico, 33081 Aviano, Italy
| | - Michele Spina
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), Istituto di Ricovero e Cura a Carattere Scientifico, 33081 Aviano, Italy
| | - Alessandra Bearz
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), Istituto di Ricovero e Cura a Carattere Scientifico, 33081 Aviano, Italy
| |
Collapse
|
6
|
Zhao J, Ma Z, Li H, Sun D, Hu Y, Zhang C, Zhang Y. Risks of cardiovascular toxicities associated with ALK tyrosine kinase inhibitors in patients with non-small-cell lung cancer: a meta-analysis of randomized control trials. Expert Opin Drug Saf 2023; 22:581-588. [PMID: 36803384 DOI: 10.1080/14740338.2023.2182284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 01/30/2023] [Indexed: 02/20/2023]
Abstract
BACKGROUND Anaplastic lymphoma kinases (ALK) tyrosine kinase inhibitors (TKIs) are effective and safe targeted therapies used in advanced ALK-positive non-small cell lung cancers (NSCLC). However, ALK-TKIs associated cardiovascular toxicities in patients with ALK-positive NSCLCremain incompletely characterized. We conducted the first meta-analysis to investigate this. RESEARCH DESIGN AND METHODS To determine the cardiovascular toxicities associated with these agents, we carried out a meta-analysis comparing ALK-TKIs with chemotherapy and a meta-analysis comparing crizotinib with other ALK-TKIs. Statistical analysis was conducted to calculate the RRs and 95% confidence intervals (CIs) by using either random effects or fixed-effect models according to the heterogeneity of the included studies. RESULTS A total of 11 studies (2855 patients) were included. ALK-TKIs ranked to have more severe cardiovascular toxicities than chemotherapy (RR 5.03, 95% CI 1.97-12.84, P = 0.0007) . Compared with other ALK-TKIs, increased risks of cardiac disorders and VTEs associated with crizotinib were found (cardiac disorders RR 1.75, 95% CI 1.07-2.86, P = 0.03; risk of VTEs RR 3.97, 95% CI 1.69-9.31, P = 0.002; respectively). CONCLUSION ALK-TKIs were associated with higher risks of cardiovascular toxicities. Special attention should be given to the risks of cardiac disorders and VTEs related to crizotinib therapy.
Collapse
Affiliation(s)
- Jin Zhao
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing, Hebei, China
| | - Zhuo Ma
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, Hebei, China
| | - Hao Li
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing, Hebei, China
| | - Dan Sun
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing, Hebei, China
| | - Yi Hu
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing, Hebei, China
| | - Chen Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing, Hebei, China
| | - Yuhui Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing, Hebei, China
| |
Collapse
|
7
|
Cheng L, Duan J, Tse G, Liu T, Li G. Sacubitril/Valsartan Ameliorates Crizotinib-Induced Cardiotoxicity in Mice. Rev Cardiovasc Med 2023; 24:192. [PMID: 39077026 PMCID: PMC11266459 DOI: 10.31083/j.rcm2407192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/14/2023] [Accepted: 02/03/2023] [Indexed: 10/11/2023] Open
Abstract
BACKGROUND Lung cancer is one of the major cause of death globally. Crizotinib is a first-line drug used in treating non-small-cell lung cancer (NSCLC). However, the pathophysiological mechanisms underlying its cardiotoxicity are unknown. This study investigated the mechanisms of crizotinib-induced cardiotoxicity and explored whether this toxicity can be prevented by the angiotensin receptor/neprilysin inhibitor sacubitril/valsartan. METHODS Male C57BL/6 mice were randomly divided into three groups: control, crizotinib (40 mg ⋅ kg - 1 ⋅ d - 1 for four weeks), and crizotinib + sacubitril/valsartan (40 mg ⋅ kg - 1 ⋅ d - 1 /60 mg ⋅ kg - 1 ⋅ d - 1 for four weeks). Expression of genes in myocardial tissue were detected by transcriptomic sequencing, with verification of the differentially expressed genes (DEGs) using Real time-polymerase chain reaction (RT-PCR). Blood pressure (BP) and cardiac function of animals were measured using non-invasive monitoring and echocardiography approaches. Ventricular refractory period (RP), as well as the induction rate and score of ventricular arrhythmias (VAs) were detected by in vivo electrophysiology. Epicardial conductance was measured by mapping. Expression of Myh7 in myocardium was detected by western blot and RT-PCR. RESULTS DEGs detected using transcriptomic sequencing included 10 up-regulated and 20 down-regulated genes. The first 5 DEGs identified were Myh7, Ngp, Lcn2, Ciart and Ptgds. Kyoto Encyclopedia of Genes and Genomes (KEGG) result indicated that Myh7 is involved in myocarditis, cardiomyopathy, and cardiac muscle contraction. Crizotinib treatment increased blood pressure, prolonged QTc interval, shortened ventricular RP, increased the incidence and score of right VAs, and increased Myh7 expression. Most of these responses were limited by sacubitril/valsartan. CONCLUSIONS Crizotinib induced a range of cardiotoxic side effects in a mouse model and increased Myh7 expression represents a biomarker for this response. These cardiovascular toxic responses can be largely prevented by sacubitril/valsartan.
Collapse
Affiliation(s)
- Lijun Cheng
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, 300211 Tianjin, China
| | - Junying Duan
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, 300211 Tianjin, China
| | - Gary Tse
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, 300211 Tianjin, China
- Department of Health Sciences, School of Nursing and Health Studies, Hong Kong Metropolitan University, 518057 Hong Kong, China
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, 300211 Tianjin, China
| | - Guangping Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, 300211 Tianjin, China
| |
Collapse
|
8
|
Wang F, Xu G, Wu X. Cardiac arrhythmias associated with anaplastic lymphoma kinase (ALK) inhibitors: an analysis of the FDA Adverse Event Reporting System (FAERS). Expert Opin Drug Saf 2023; 22:1127-1132. [PMID: 37428255 DOI: 10.1080/14740338.2023.2234279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 03/18/2023] [Indexed: 07/11/2023]
Abstract
BACKGROUND Anaplastic lymphoma kinase tyrosine kinase inhibitors (ALK-TKIs) may provoke cardiac arrhythmias. We conducted this pharmacovigilance analysis to research cardiac arrhythmias associated with ALK-TKIs using the Food and Drug Administration Adverse Event Reporting System (FAERS). RESEARCH DESIGN AND METHODS The first ALK-TKI, named crizotinib, was approved by the Food and Drug Administration (FDA) on 26 August 2011 for the treatment of ALK-rearranged non-small cell lung cancer (NSCLC). We evaluated ALK-TKIs-induced cardiac arrhythmias, by using the reporting odds ratio (ROR) and information component (IC) for mining the adverse event report signals in the FAERS database between January 2016 and June 2022. RESULTS We identified a total of 362 ALK-TKIs-related cardiac arrhythmia reports which appeared to influence more men (64.44%) than women (30.76%), with a median age of 68 (interquartile range [IQR] 7-74) years. Compared with the full database, ALK-TKIs were detected with pharmacovigilance of cardiac arrhythmias (ROR025 = 1.26, IC025 = 0.26). Crizotinib and alectinib were found to be related to higher reporting of arrhythmias. The median time to onset (TTO) among five ALK-TKI therapies was significantly different (p = 0.044). CONCLUSION ALK-TKIs present different frequencies of cardiac arrhythmias reporting, with only crizotinib and alectinib producing positive signals in high-level group term (HLGT) level arrhythmia. The time interval between the initial of drug treatment to the onset of arrhythmia varies greatly and cannot be predicted.
Collapse
Affiliation(s)
- Feifei Wang
- Department of Pharmacy, Hefei BOE Hospital, Hefei, P.R.China
| | - Guishui Xu
- Department of Orthopaedic, The First People's Hospital of Anqing, Anqing, P.R.China
| | - Xinan Wu
- Department of Pharmacy, Hefei BOE Hospital, Hefei, P.R.China
| |
Collapse
|
9
|
Leiva O, Bohart I, Ahuja T, Park D. Off-Target Effects of Cancer Therapy on Development of Therapy-Induced Arrhythmia: A Review. Cardiology 2023; 148:324-334. [PMID: 36702116 PMCID: PMC10614257 DOI: 10.1159/000529260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 01/17/2023] [Indexed: 01/27/2023]
Abstract
BACKGROUND Advances in cancer therapeutics have improved overall survival and prognosis in this patient population; however, this has come at the expense of cardiotoxicity including arrhythmia. SUMMARY Cancer and its therapies are associated with cardiotoxicity via several mechanisms including inflammation, cardiomyopathy, and off-target effects. Among cancer therapies, anthracyclines and tyrosine kinase inhibitors (TKIs) are particularly known for their pro-arrhythmia effects. In addition to cardiomyopathy, anthracyclines may be pro-arrhythmogenic via reactive oxygen species (ROS) generation and altered calcium handling. TKIs may mediate their cardiotoxicity via inhibition of off-target tyrosine kinases. Ibrutinib-mediated inhibition of CSK may be responsible for the increased prevalence of atrial fibrillation. Further investigation is warranted to further elucidate the mechanisms behind arrhythmias in cancer therapies. KEY MESSAGES Arrhythmias are a common cardiotoxicity of cancer therapies. Cancer therapies may induce arrhythmias via off-target effects. Understanding the mechanisms underlying arrhythmogenesis associated with cancer therapies may help design cancer therapies that can avoid these toxicities.
Collapse
Affiliation(s)
- Orly Leiva
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York City, New York, USA
| | - Isaac Bohart
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York City, New York, USA
| | - Tania Ahuja
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York City, New York, USA
| | - David Park
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York City, New York, USA
| |
Collapse
|
10
|
Grela-Wojewoda A, Pacholczak-Madej R, Adamczyk A, Korman M, Püsküllüoğlu M. Cardiotoxicity Induced by Protein Kinase Inhibitors in Patients with Cancer. Int J Mol Sci 2022; 23:ijms23052815. [PMID: 35269958 PMCID: PMC8910876 DOI: 10.3390/ijms23052815] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 12/24/2022] Open
Abstract
Kinase inhibitors (KIs) represent a growing class of drugs directed at various protein kinases and used in the treatment of both solid tumors and hematologic malignancies. It is a heterogeneous group of compounds that are widely applied not only in different types of tumors but also in tumors that are positive for a specific predictive factor. This review summarizes common cardiotoxic effects of KIs, including hypertension, arrhythmias with bradycardia and QTc prolongation, and cardiomyopathy that can lead to heart failure, as well as less common effects such as fluid retention, ischemic heart disease, and elevated risk of thromboembolic events. The guidelines for cardiac monitoring and management of the most common cardiotoxic effects of protein KIs are discussed. Potential signaling pathways affected by KIs and likely contributing to cardiac damage are also described. Finally, the need for further research into the molecular mechanisms underlying the cardiovascular toxicity of these drugs is indicated.
Collapse
Affiliation(s)
- Aleksandra Grela-Wojewoda
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Kraków Branch, Garncarska 11, 31-115 Kraków, Poland; (R.P.-M.); (M.P.)
- Correspondence: ; Tel.: +48-1263-48350
| | - Renata Pacholczak-Madej
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Kraków Branch, Garncarska 11, 31-115 Kraków, Poland; (R.P.-M.); (M.P.)
- Department of Anatomy, Jagiellonian University Medical College, 31-008 Kraków, Poland
| | - Agnieszka Adamczyk
- Department of Tumour Pathology, Maria Sklodowska-Curie National Research Institute of Oncology, Kraków Branch, Garncarska 11, 31-115 Kraków, Poland;
| | - Michał Korman
- Faculty of Medicine, Jagiellonian University Medical College, 31-008 Kraków, Poland;
| | - Mirosława Püsküllüoğlu
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Kraków Branch, Garncarska 11, 31-115 Kraków, Poland; (R.P.-M.); (M.P.)
| |
Collapse
|
11
|
Tamargo J, Caballero R, Delpón E. Cancer Chemotherapy-Induced Sinus Bradycardia: A Narrative Review of a Forgotten Adverse Effect of Cardiotoxicity. Drug Saf 2022; 45:101-126. [PMID: 35025085 DOI: 10.1007/s40264-021-01132-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2021] [Indexed: 12/20/2022]
Abstract
Cardiotoxicity is a common adverse effect of anticancer drugs (ACDs), including the so-called targeted drugs, and increases morbidity and mortality in patients with cancer. Attention has focused mainly on ACD-induced heart failure, myocardial ischemia, hypertension, thromboembolism, QT prolongation, and tachyarrhythmias. Yet, although an increasing number of ACDs can produce sinus bradycardia (SB), this proarrhythmic effect remains an underappreciated complication, probably because of its low incidence and severity since most patients are asymptomatic. However, SB merits our interest because its incidence increases with the aging of the population and cancer is an age-related disease and because SB represents a risk factor for QT prolongation. Indeed, several ACDs that produce SB also prolong the QT interval. We reviewed published reports on ACD-induced SB from January 1971 to November 2020 using the PubMed and EMBASE databases. Published reports from clinical trials, case reports, and recent reviews were considered. This review describes the associations between ACDs and SB, their clinical relevance, risk factors, and possible mechanisms of onset and treatment.
Collapse
Affiliation(s)
- Juan Tamargo
- Department of Pharmacology, School of Medicine, Universidad Complutense, Institute of Health Gregorio Marañón, CIBERCV, 28040, Madrid, Spain.
| | - Ricardo Caballero
- Department of Pharmacology, School of Medicine, Universidad Complutense, Institute of Health Gregorio Marañón, CIBERCV, 28040, Madrid, Spain
| | - Eva Delpón
- Department of Pharmacology, School of Medicine, Universidad Complutense, Institute of Health Gregorio Marañón, CIBERCV, 28040, Madrid, Spain
| |
Collapse
|
12
|
ALK inhibitor-induced bradycardia: A systematic-review and meta-analysis. Lung Cancer 2021; 161:9-17. [PMID: 34492553 DOI: 10.1016/j.lungcan.2021.08.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/25/2021] [Accepted: 08/25/2021] [Indexed: 11/23/2022]
Abstract
INTRODUCTION Anaplastic Lymphoma Kinase (ALK) inhibitors have revolutionized the treatment of advanced ALK-positive non-small cell lung cancer (NSCLC), improving progression-free survival. Bradycardia is a potential adverse effect of these agents. We aimed to determine the risk of bradycardia associated with ALK inhibitors in patients with advanced NSCLC. MATERIALS AND METHODS We conducted a systematic search of MEDLINE, EMBASE, Cochrane Central Register of Controlled Trials, National clinical trial registry, and Web of Science Core Collection. We included all randomized controlled trials in which an ALK-inhibitor was compared with another ALK-inhibitor or standard chemotherapy. Meta-analyses were conducted to evaluate the pooled incidence rates of bradycardia and dizziness using fixed effect models. RESULTS The pooled incidence of bradycardia among 1737 individuals prescribed ALK inhibitors was 8% during a mean follow-up of 1.26 years. Crizotinib led to more bradycardia than standard chemotherapy (relative risk, RR 24.68, 95% CI 7.11-85.), while no difference was seen between crizotinib and alectinib (RR 1.12, 95% CI 0.79-1.59). The next-generation ALK inhibitors alectinib, brigatinib and lorlatinib combined resulted in a similar rate of bradycardia when compared to crizotinib (RR 0.77, 95% CI 0.57-1.04). All ALK inhibitors (as an aggregate) caused more dizziness (as a potential symptom of bradycardia) than standard chemotherapy (RR 1.88, 95% CI 1.44-2.44). CONCLUSION Crizotinib for the treatment of NSCLC is associated with a higher risk for bradycardia compared to standard chemotherapy. There is no evidence of a difference in bradycardia risk between crizotinib and newer ALK inhibitors.
Collapse
|
13
|
Rassaf T, Totzeck M, Backs J, Bokemeyer C, Hallek M, Hilfiker-Kleiner D, Hochhaus A, Lüftner D, Müller OJ, Neudorf U, Pfister R, von Haehling S, Lehmann LH, Bauersachs J. Onco-Cardiology: Consensus Paper of the German Cardiac Society, the German Society for Pediatric Cardiology and Congenital Heart Defects and the German Society for Hematology and Medical Oncology. Clin Res Cardiol 2020; 109:1197-1222. [PMID: 32405737 PMCID: PMC7515958 DOI: 10.1007/s00392-020-01636-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 03/13/2020] [Indexed: 12/11/2022]
Abstract
The acute and long-lasting side effects of modern multimodal tumour therapy significantly impair quality of life and survival of patients afflicted with malignancies. The key components of this therapy include radiotherapy, conventional chemotherapy, immunotherapy and targeted therapies. In addition to established tumour therapy strategies, up to 30 new therapies are approved each year with only incompletely characterised side effects. This consensus paper discusses the risk factors that contribute to the development of a potentially adverse reaction to tumour therapy and, in addition, defines specific side effect profiles for different treatment groups. The focus is on novel therapeutics and recommendations for the surveillance and treatment of specific patient groups.
Collapse
Affiliation(s)
- Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Centre Essen, University Hospital Essen, Hufelandstrasse 55, 45147, Essen, Germany.
| | - Matthias Totzeck
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Centre Essen, University Hospital Essen, Hufelandstrasse 55, 45147, Essen, Germany
| | - Johannes Backs
- Institute for Experimental Cardiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Carsten Bokemeyer
- Department of Oncology, Hematology and Bone Marrow Transplantation with the Section Pneumology, Centre for Oncology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Hallek
- Department I of Internal Medicine, Center for Integrated Oncology ABCD, University Hospital of Cologne, Cologne, Germany
| | | | - Andreas Hochhaus
- Department of Hematology and Medical Oncology, University Hospital Jena, Jena, Germany
| | - Diana Lüftner
- Department of Haematology, Oncology and Tumour Immunology, Charité, Humboldt University Berlin, Berlin, Germany
| | - Oliver J Müller
- Department of Internal Medicine III (Cardiology, Angiology and Internal Intensive Care Medicine), University Hospital Schleswig-Holstein, University of Kiel, Kiel, Germany
| | - Ulrich Neudorf
- Department of Pediatrics III, West German Heart and Vascular Centre Essen, University Hospital Essen, Essen, Germany
| | - Roman Pfister
- Clinic III for Internal Medicine, General and Interventional Cardiology, Electrophysiology, Angiology, Pneumology and Internal Intensive Care Medicine, University Hospital Cologne, Cologne, Germany
| | - Stephan von Haehling
- Department of Cardiology and Pneumology, Heart Center Göttingen, University of Göttingen Medical Center and German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
| | - Lorenz H Lehmann
- Department of Cardiology, Angiology, Pneumology, University Hospital Heidelberg, Heidelberg, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
14
|
Lee DH, Chandrashekhar S, Fradley MG. Electrophysiologic Complications in Cancer Patients. Methodist Debakey Cardiovasc J 2020; 15:282-288. [PMID: 31988689 DOI: 10.14797/mdcj-15-4-282] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In recent years, dramatic advances in both cancer diagnosis and treatment have led to significantly increased survival rates. As such, cardiovascular toxicities due to oncologic treatments are more frequently identified. Although heart failure and cardiomyopathy have historically been the cardiotoxicities most associated with cancer therapeutics, it is now recognized that all components of the cardiovascular system can be affected. In this review, we discuss electrophysiologic complications of cancer treatments, including atrial and ventricular tachyarrhythmias as well as bradyarrhythmias, and recommend a multidisciplinary approach with both cardiologists and oncologists to provide safe and effective care to these patients.
Collapse
Affiliation(s)
| | | | - Michael G Fradley
- UNIVERSITY OF SOUTH FLORIDA, TAMPA, FLORIDA.,H. LEE MOFFITT CANCER CENTER AND RESEARCH INSTITUTE, TAMPA, FLORIDA
| |
Collapse
|
15
|
Zaborowska-Szmit M, Krzakowski M, Kowalski DM, Szmit S. Cardiovascular Complications of Systemic Therapy in Non-Small-Cell Lung Cancer. J Clin Med 2020; 9:E1268. [PMID: 32349387 PMCID: PMC7287714 DOI: 10.3390/jcm9051268] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular diseases may determine therapy outcomes of non-small-cell lung cancer (NSCLC). The evidence for how iatrogenic cardiovascular complications contribute to ceasing anticancer treatment, decreasing the quality of life or even premature death, is unclear. Older patients and smokers are at risk of atherosclerosis and arterial thromboembolic events (TE), such as myocardial infarction or stroke. Venous TE can be observed in up to 15% of NSCLC patients, but the risk increases three to five times in ALK (anaplastic lymphoma kinase)-rearranged NSCLC. ALK inhibitors are associated with electrophysiological disorders. Cytotoxic agents and anti-VEGF inhibitors mainly cause vascular complications, including venous or arterial TE. Cardiac dysfunction and arrhythmias seem to be less frequent. Chemotherapy is often administered in two-drug regimens. Clinical events can be triggered by different mechanisms. Among epidermal growth factor inhibitors, erlotinib and gefitinib can lead to coronary artery events; however, afatinib and osimertinib can be associated with the development of heart failure. During anti-PD1/anti-PDL1 therapy, myocarditis is possible, which must be differentiated from acute coronary syndrome and heart failure. Awareness of all possible cardiovascular complications in NSCLC encourages vigilance in early diagnostics and treatment.
Collapse
Affiliation(s)
- Magdalena Zaborowska-Szmit
- Department of Lung Cancer and Thoracic Tumors, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (M.Z.-S.); (M.K.); (D.M.K.)
| | - Maciej Krzakowski
- Department of Lung Cancer and Thoracic Tumors, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (M.Z.-S.); (M.K.); (D.M.K.)
| | - Dariusz M. Kowalski
- Department of Lung Cancer and Thoracic Tumors, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (M.Z.-S.); (M.K.); (D.M.K.)
| | - Sebastian Szmit
- Department of Pulmonary Circulation, Thromboembolic Diseases and Cardiology, Centre of Postgraduate Medical Education, European Health Centre, 05-400 Otwock, Poland
| |
Collapse
|