1
|
Costa IBSDS, Furtado RHM, Drager LF, de Barros E Silva PGM, Melo MDTD, Araruna P, Bacchiega BC, Cauduro S, Walter E, Fialho GL, Silvestre O, Damiani LP, Barbosa LM, Luz MN, Silva ACA, de Mattos RR, Saretta R, Rehder MHHS, Hajjar LA, Lopes-Fernandez T, Dent S, Gibson CM, Lopes RD, Kalil Filho R. Effects of carvedilol on the prevention of cardiotoxicity induced by anthracyclines: Design and rationale of the CARDIOTOX trial. Am Heart J 2025; 285:1-11. [PMID: 39988204 DOI: 10.1016/j.ahj.2025.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 02/25/2025]
Abstract
BACKGROUND Patients with cancer undergoing chemotherapy with an anthracycline-based regimen are at increased risk of cardiotoxicity, predisposing to heart failure, arrhythmias and death. Whether carvedilol may confer benefit to prevent anthracycline-induced cardiotoxicity remains to be determined. DESIGN CARDIOTOX is a double-blind, placebo controlled randomized clinical trial that plan to enroll 1,018 patients across 25 study sites in Brazil. Patients with active cancer scheduled to undergo an anthracycline-based chemotherapy regimen are eligible. Patients with prior HF or cardiomyopathy are excluded. Patients are randomized in 1:1 ratio to carvedilol (starting dose 6.25mg BID up titrated to 25mg BID or maximum tolerated dose) or placebo, stratified by site and use of renin-angiotensin blockers at baseline. Study drug is administered through the duration of chemotherapy and up to 30 days after the last dose of anthracycline. Patients are scheduled to undergo echocardiographic evaluations at baseline and at 3, 6, and 12 months. The study primary endpoint is the composite of new left ventricle ejection fraction (LVEF) reduction by at least 10% leading to an LVEF <50%, cardiovascular death, myocardial infarction, urgent care visit or hospitalization for heart failure, or clinically significant arrhythmias at 12 months. Echocardiographic images will be analyzed by a central core lab, clinical outcomes will be adjudicated, and safety endpoints include serious adverse events and adverse events of special interest (symptomatic bradycardia, hypotension, syncope and bronchospasm). SUMMARY The CARDIOTOX trial is the largest trial to date analyzing the potential role of beta-blockers as prophylactic therapy to prevent cardiotoxicity induced by anthracyclines. TRIAL REGISTRATION Effects of Carvedilol on Cardiotoxicity in Cancer Patients Submitted to Anthracycline Therapy (CardioTox). CLINICALTRIALS gov ID NCT04939883. https://clinicaltrials.gov/study/NCT04939883.
Collapse
Affiliation(s)
- Isabela Bispo Santos da Silva Costa
- Hospital Sírio-Libanês Research and Education Institute, São Paulo, Brazil; Instituto do Cancer do Estado de São Paulo, Hospital das Clinicas da Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Remo H M Furtado
- Hospital Sírio-Libanês Research and Education Institute, São Paulo, Brazil; Brazilian Clinical Research Institute, São Paulo, Brazil; Instituto do Coração (InCor), Hospital das Clinicas da Faculdade de Medicina, Sao Paulo, Brazil
| | - Luciano F Drager
- Hospital Sírio-Libanês Research and Education Institute, São Paulo, Brazil; Instituto do Coração (InCor), Hospital das Clinicas da Faculdade de Medicina, Sao Paulo, Brazil
| | | | | | | | | | | | | | - Guilherme Loureiro Fialho
- Hospital Universitario Professor Polydoro Ernani de São Thiago, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | | | - Lucas P Damiani
- Brazilian Clinical Research Institute, São Paulo, Brazil; Instituto Dante Pazzanese de Cardiologia, Sao Paulo, Brazil
| | | | | | | | | | - Roberta Saretta
- Hospital Sírio-Libanês Research and Education Institute, São Paulo, Brazil
| | | | - Ludhmila Abrahao Hajjar
- Instituto do Cancer do Estado de São Paulo, Hospital das Clinicas da Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil; Brazilian Clinical Research Institute, São Paulo, Brazil; Instituto D´Or de Ensino e Pesquisa, Sao Paulo, Brazil
| | - Teresa Lopes-Fernandez
- Department of Cardiology, La Paz University Hospital, IdiPAZ Research Institute, Madrid, Spain; Department of Cardiology, Hospital Universitario Quirónsalud Madrid, Madrid, Spain.
| | - Susan Dent
- Wilmot Cancer Institute, University of Rochester, Rochester NY, USA
| | - C Michael Gibson
- Baim Research Institute and Harvard Medical School, Boston, MA, USA
| | - Renato D Lopes
- Hospital Sírio-Libanês Research and Education Institute, São Paulo, Brazil; Brazilian Clinical Research Institute, São Paulo, Brazil; Duke Clinical Research Institute, Duke University Medical Center, Durham, NC, USA
| | - Roberto Kalil Filho
- Hospital Sírio-Libanês Research and Education Institute, São Paulo, Brazil; Instituto do Coração (InCor), Hospital das Clinicas da Faculdade de Medicina, Sao Paulo, Brazil
| |
Collapse
|
2
|
Lescroart M, Kemp H, Imauven O, Raphalen JH, Bagate F, Schmidt J, Issa N, Decavele M, Moreau AS, Tamion F, Mourvillier B, Calvet L, Canet E, Lebert C, Pons S, Lacave G, Wallet F, Winiszewski H, Merdji H, De Chambrun MP, Argaud L, Kimmoun A, Dumas G, Zafrani L. Cardiogenic shock in patients with active onco-hematological malignancies: A multicenter retrospective study. J Crit Care 2025; 87:155028. [PMID: 39848115 DOI: 10.1016/j.jcrc.2025.155028] [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: 10/28/2024] [Revised: 12/28/2024] [Accepted: 01/16/2025] [Indexed: 01/25/2025]
Abstract
PURPOSE Onco-hematological (OH) patients face significant cardiovascular risks due to malignancy and drug toxicity. Data are limited on the characteristics and outcomes of OH patients with cardiogenic shock (CS) in intensive care units (ICUs). METHODS This multicenter retrospective study included 214 OH patients with CS across 22 ICUs (2010-2021). The objectives were to (i) identify risk factors for 30-day mortality, (ii) describe early and long-term outcomes, and (iii) assess the prognostic impact of malignancy by comparing OH patients to a control group of CS patients. RESULTS The 30-day survival rate was 44.8 %. Multivariate analysis identified previous cardiomyopathy (OR = 1.61), acute kidney injury (OR = 1.62), lactate levels (OR = 1.08 per 1 mmol/L), pulmonary embolism (OR = 3.04), invasive mechanical ventilation (OR = 3.48), and epinephrine use (OR = 2.09) as factors associated with 30-day mortality. Among ICU survivors, 54 % were alive at 1 year with a median left ventricular ejection fraction of 52 %. OH malignancy was significantly associated with 30-day mortality (HR 2.54). CONCLUSION The prognosis for OH patients with CS in the ICU is poor, with epinephrine use associated with worse outcomes. Further research is needed to refine risk stratification and improve treatments for this population.
Collapse
Affiliation(s)
- Mickael Lescroart
- Hospital Saint-Louis et Université Paris Cité, Assistance Publique-Hôpitaux de Paris, France; CHRU de Nancy, Médecine Intensive et Réanimation Brabois, Université de Lorraine, Vandœuvre-Lès-Nancy, France
| | - Hélène Kemp
- Hospital Saint-Louis et Université Paris Cité, Assistance Publique-Hôpitaux de Paris, France
| | - Olivier Imauven
- Groupe hospitalier Diaconesses - Croix Saint-Simon, institut d'anesthésie de l'Est Parisien, Paris, France
| | - Jean Herlé Raphalen
- Intensive Care Unit, Necker University Hospital, Assistance Publique-Hôpitaux de Paris, 149 rue de Sèvres, 75015 Paris, France
| | - François Bagate
- Service de Médecine Intensive Réanimation, AP-HP, CHU Henri Mondor, DHU A-TVB, 51, avenue du Mal de Lattre de Tassigny, 94010 Créteil Cedex, France
| | - Julien Schmidt
- Unité de médecine intensive et réanimation, Assistance Publique-Hôpitaux de Paris, Avicenne Hospital, Groupe Hospitalier Paris Seine Saint-Denis, Bobigny, France
| | - Nahema Issa
- Réanimation médicale, groupe hospitalier Saint-André, 1, rue Jean-Burguet, 33075 Bordeaux cedex, France; Médecine interne et maladies infectieuses, groupe hospitalier Saint-André, 1, rue Jean-Burguet, 33075 Bordeaux cedex, France
| | - Maxens Decavele
- APHP, Groupe Hospitalier Universitaire APHP-Sorbonne Université, site Pitié-Salpêtrière, Service Médecine Intensive et Réanimation (Département R3S), 75013, Paris, France
| | - Anne-Sophie Moreau
- CHU de Lille, Hôpital Salengro, Service de Médecine Intensive Réanimation, rue Emile-Laine, 59037 Lille, France
| | - Fabienne Tamion
- Service de Réanimation Médicale, Normandie Univ, UNIROUEN, U1096, CHU de Rouen, F 76000 Rouen, France
| | - Bruno Mourvillier
- Centre Hospitalo-Universitaire de Reims (CHU), Hôpital Robert-Debré, Service de Réanimation médicale, Reims, France
| | - Laure Calvet
- Service de Reanimation Medicale, Hopital Gabriel Monpied, CHU, Clermont-Ferrand, France
| | - Emmanuel Canet
- Medecine Intensive Reanimation, University Hospital Center, Nantes, France
| | - Christine Lebert
- Service Medico-Chirurgical, Unité de soins Intensifs, Centre Hospitalier de La Roche-sur-Yon, France
| | - Stephanie Pons
- Sorbonne University, GRC 29, AP-HP, DMU DREAM, Department of Anaesthesiology and Critical Care, Pitié-Salpêtrière Hospital, Paris, France; Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Guillaume Lacave
- Service de Réanimation Médico-Chirurgicale, Centre Hospitalier de Versailles, Le Chesnay, France
| | - Florent Wallet
- Médecine Intensive et Réanimation, Hôpital Lyon Sud, Pierre-Bénite 69495, France
| | - Hadrien Winiszewski
- Médecine Intensive et Réanimation, Centre Hospitalier Universitaire de Besançon, Université de Franche-Comté, France
| | - Hamid Merdji
- Service de Médecine Intensive et Réanimation, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, INSERM UMR 1260, Regenerative NanoMedicine, FMTS, Strasbourg, France
| | - Marc Pineton De Chambrun
- Sorbonne Université, UPMC Univ Paris 06, INSERM UMRS_1166-iCAN, Institute of Cardiometabolism and Nutrition, 75651 Paris Cedex 13, France; Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Medical Intensive Care Unit, 75651 Paris Cedex 13, France
| | - Laurent Argaud
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Service de Médecine Intensive-Réanimation, 5, Place d'Arsonval, 69437 Lyon Cedex 03, France
| | - Antoine Kimmoun
- CHRU de Nancy, Médecine Intensive et Réanimation Brabois, Université de Lorraine, Vandœuvre-Lès-Nancy, France; INSERM U942, MASCOT, Paris, France
| | - Guillaume Dumas
- Medical Intensive Care Unit, Service de Médecine Intensive-Réanimation, CHU Grenoble-Alpes, Université Grenoble-Alpes, INSERM, U1042-HP2, Grenoble, France
| | - Lara Zafrani
- Hospital Saint-Louis et Université Paris Cité, Assistance Publique-Hôpitaux de Paris, France.
| |
Collapse
|
3
|
Sutton AL, Zhao J, He J, Tossas KY, Bottinor W, Sheppard VB. Examining factors associated with experiencing cardiac arrhythmias in Black and White breast cancer survivors who received anthracyclines or trastuzumab. Breast Cancer Res Treat 2025; 211:537-544. [PMID: 40146434 PMCID: PMC12006251 DOI: 10.1007/s10549-025-07671-0] [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: 09/04/2024] [Accepted: 02/25/2025] [Indexed: 03/28/2025]
Abstract
PURPOSE Racial disparities exist regarding cardiovascular (CV) toxicities following breast cancer treatment; however, studies on racial differences in cardiac arrhythmias are lacking. This study examined associations between demographic and clinical factors and arrhythmia diagnosis in Black and White breast cancer survivors. METHODS This study included a retrospective cohort of Black and White women who were diagnosed with breast cancer and who received potentially cardiotoxic treatment. Cardiac arrhythmia data were captured via International Classification of Diseases, Tenth and Ninth Versions (ICD-10 and ICD-9). Experiences with cardiac arrhythmias were compared across racial groups. The associations of demographic and clinical factors with cardiac arrhythmias were evaluated using logistic regression for all women and in race-stratified models. RESULTS Thirty-three percent of the total 860 women in our study sample (mean (standard deviation) age 50.3 (10.7) years) old) experienced cardiac arrhythmias. In bivariate analyses, we observed a statistically discernible association between race and arrhythmia status following a breast cancer diagnosis (p = 0.004); however, this association was no longer significant in the multivariable model. In race-stratified multivariable analysis, the odds of experiencing arrhythmias in Black women over 50 years old are 51% lower than in Black women aged 50 years old or younger (adjusted odds ratio (OR): 0.49; 95% confidence interval (CI): 0.28, 0.86). All else being equal, Black women with hypertension had 2.68 times (95% CI: 1.51, 4.81) higher odds of experiencing arrhythmias than those without hypertension. White women with obesity had higher odds of experiencing arrhythmias than those with normal weight or underweight status. (adjusted OR 1.93: [1.17, 3.20]). CONCLUSION Survivors with chronic conditions like hypertension and obesity may require enhanced cardiac surveillance. Further investigation into hypertension management in Black survivors may shed light on its impact on CV toxicities in this group.
Collapse
Affiliation(s)
- Arnethea L Sutton
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, P.O. Box 843021, Richmond, VA, 23220, USA.
- VCU Massey Comprehensive Cancer Center, Richmond, VA, USA.
| | - Jinlei Zhao
- VCU Massey Comprehensive Cancer Center, Richmond, VA, USA
| | - Jian He
- VCU Massey Comprehensive Cancer Center, Richmond, VA, USA
| | - Katherine Y Tossas
- VCU Massey Comprehensive Cancer Center, Richmond, VA, USA
- Department of Social and Behavioral Sciences, Virginia Commonwealth University School of Public Health, Richmond, VA, USA
| | - Wendy Bottinor
- Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Vanessa B Sheppard
- Department of Social and Behavioral Sciences, Virginia Commonwealth University School of Public Health, Richmond, VA, USA
| |
Collapse
|
4
|
Li C, Jin Y, Ma J, Sun R, Huang J, Qi Y, Wang Q, Yu J. Diagnostic value of ROCK2 protein in immune checkpoint inhibitors-associated myocarditis. Int J Cardiol 2025; 427:133104. [PMID: 40037484 DOI: 10.1016/j.ijcard.2025.133104] [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: 12/16/2024] [Revised: 02/17/2025] [Accepted: 02/26/2025] [Indexed: 03/06/2025]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have advanced cancer treatment but are associated with serious cardiovascular side effects, including myocarditis, which is challenging to diagnose due to limited specificity of current biomarkers. Rho-associated kinase 2 (ROCK2) may play a role in myocarditis pathogenesis by mediating inflammation and myocardial remodeling. This study investigates the potential of ROCK2 protein detection as a diagnostic marker for ICIs-associated myocarditis. OBJECTIVES To evaluate ROCK2 protein levels in patients with ICIs-associated myocarditis and assess its diagnostic value, providing insights for improved identification and management of this condition. METHODS We conducted a study with 10 ICIs-treated gastric cancer patients diagnosed with myocarditis and a control group of 10 similar ICIs-treated patients without myocarditis. ROCK2 levels and inflammatory markers were measured via ELISA, and clinical assessments included cardiac imaging, electrocardiography, and echocardiography. Statistical analysis of group differences used independent t-tests and chi-square tests. RESULTS ROCK2 expression was significantly higher in myocarditis patients compared to controls (p < 0.001), alongside elevated inflammatory markers, specifically hypersensitive C-reactive protein (hs-CRP), interleukins1β (IL-1β) and IL-17. Diagnostic myocardial markers such as N-terminal pro B-type natriuretic peptide (NT-proBNP), and soluble suppression of tumorigenicity 2 (sST2) also showed substantial elevation. Following treatment discontinuation and glucocorticoid administration, both ROCK2 levels and inflammatory indicators declined. CONCLUSIONS Elevated ROCK2 protein levels could serve as a promising biomarker for ICIs-associated myocarditis. ROCK2 detection, combined with traditional markers, may enhance diagnostic accuracy. Further studies are warranted to explore ROCK2 inhibition as a potential therapeutic strategy for managing ICIs-associated myocarditis.
Collapse
Affiliation(s)
- Caie Li
- Department of Cardiology, The Second Hospital of Lanzhou University, Lanzhou 730030, China
| | - Yucheng Jin
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Jie Ma
- Department of Cardiology, The Second Hospital of Lanzhou University, Lanzhou 730030, China
| | - Runmin Sun
- Department of Cardiology, The Second Hospital of Lanzhou University, Lanzhou 730030, China
| | - Jiawang Huang
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Yali Qi
- Department of Oncology, The Second Hospital of Lanzhou University, Lanzhou 730030, China
| | - Qiongying Wang
- Department of Cardiology, The Second Hospital of Lanzhou University, Lanzhou 730030, China
| | - Jing Yu
- Department of Cardiology, The Second Hospital of Lanzhou University, Lanzhou 730030, China.
| |
Collapse
|
5
|
Qi Y, Ge H, Sun X, Wei Y, Zhai J, Qian H, Mo H, Ma F. Systemic immune characteristics predicting toxicity to immune checkpoint inhibitors in patients with advanced breast cancer. J Autoimmun 2025; 153:103423. [PMID: 40267835 DOI: 10.1016/j.jaut.2025.103423] [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: 01/30/2025] [Revised: 04/12/2025] [Accepted: 04/14/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are among the most promising treatment options for cancer. However, frequent and sometimes life-threatening immune-related adverse events (irAEs) are associated with ICI treatment. Therefore, it is imperative to establish a model for predicting the risk of irAEs to identify high-risk groups, enable more accurate clinical risk‒benefit analysis for ICI treatment and decrease the incidence of irAEs. However, no ideal model for predicting irAEs has been applied in clinical practice. The aim of this study was to analyze the systemic immune characteristics of patients with irAEs and establish a model for predicting the risk of irAEs. METHODS We conducted a study to monitor irAEs in patients with advanced breast cancer undergoing immunotherapy during and following the treatment course. Peripheral blood mononuclear cells (PBMCs) were collected before and after two cycles of therapy. Mass cytometry time-of-flight (CyTOF) was employed to identify baseline and posttreatment immune cell subpopulations, and the relationships between the proportions of cells in these subpopulations and the occurrence of irAEs were explored. Additionally, we conducted subgroup analyses stratified by the anatomic location and time of onset of irAEs. Furthermore, we developed a logistic regression model to predict the risk of irAEs and validated this model using two independent validation cohorts from the Gene Expression Omnibus (GEO) database (accession numbers GSE189125 and GSE186143). RESULTS By analyzing 106 blood samples and samples from two independent validation cohorts (n = 16 and 60 patients), we found that high proportions of CXCR3+CCR6+CD4+ T cells and CD38+CD86+CXCR3+CCR6+CD8+ T cells and a low proportion of CXCR3lowCD56dim natural killer (NK) cells at baseline were significantly correlated with the incidence of irAEs (P = 0.0029, P < 0.001, and P = 0.0017, respectively). In the subgroup analysis, we observed consistent results in patients with immune-related pneumonitis (ir-pneumonitis) and immune-related thyroiditis (ir-thyroiditis). In the early irAE group, the baseline proportion of CXCR3+CCR6+CD4+ T cells was greater than that in the late irAE group (P = 0.011). An analysis of PBMCs before and after ICI treatment revealed thatthe dynamic changes in the proportions of naïve CD4+ T cells and CXCR3lowCD56dim NK cells were closely related to irAE occurrence. Finally, we ultimately developed a model for predicting the risk of irAEs, which yielded an area under the receiver operating characteristic curve (AUROC) of 0.79 in the training cohort and an AUROC of 0.75 in the single-cell validation cohort (GSE189125). CONCLUSIONS These findings indicate that different populations of immune cells are associated with different irAEs and that characterization of these cells may be used as biomarkers to predict the risk of specific toxicities. This will facilitate the management of irAEs and may lead to a reduction in the incidence of irAEs.
Collapse
Affiliation(s)
- Yalong Qi
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Hewei Ge
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Xiaoying Sun
- Department of Medical Oncology, Cancer Hospital of HuanXing ChaoYang District, Beijing, China.
| | - Yuhan Wei
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Jingtong Zhai
- Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China.
| | - Haili Qian
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Hongnan Mo
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Fei Ma
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
6
|
Septianda I, Alsagaff MY, Pikir BS, Ashariati A, Fuadi MR, Zahra F. Effect of cyclophospamide, doxorubicin, vincristine and prednisone/prednisolone (CHOP) chemotherapy regimens on left ventricular systolic function and cardiac structural abnormalities in non-Hodgkin lymphoma patients. J Egypt Natl Canc Inst 2025; 37:29. [PMID: 40310515 DOI: 10.1186/s43046-025-00288-w] [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: 01/02/2025] [Accepted: 04/12/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND The cyclophospamide, doxorubicin, vincristine, and prednisone/prednisolone (CHOP) regimen, a standard treatment for aggressive non-Hodgkin lymphoma (NHL), is effective but linked to chemotherapy-induced cardiotoxicity (CDIC), such as heart failure and left ventricular dysfunction. Anthracyclines like doxorubicin are key contributors to CDIC. This study examines left ventricular dysfunction and structural abnormalities in NHL patients receiving CHOP therapy with cumulative doxorubicin doses ≥ 250 mg/m2, aiming to improve early CDIC detection and guide timely cardioprotective interventions. METHODS This prospective cohort study was conducted at Dr. Soetomo Regional General Hospital, Surabaya, from June to October 2023. We included NHL patients aged 18-60 years at any stage, treated with CHOP regimens containing cumulative doxorubicin doses ≥ 250 mg/m2, who completed at least four chemotherapy cycles, had baseline left ventricular ejection fraction (LVEF) ≥ 50%, and good echocardiographic window quality. Data were collected from medical records, biochemical tests, and echocardiography. Wilcoxon and paired T-tests were used for statistical analyses. The primary outcome was cardiac function, evaluated by reductions in LVEF, increased left ventricular strain, new structural abnormalities, and elevated high-sensitivity (HS) troponin and NT-proBNP levels post-chemotherapy. RESULTS A total of 32 patients, aged 46.62 ± 16.64 years, were included, with the colli region and stage 2 NHL being the most common. Significant differences were observed in all parameters between pre- and post-chemotherapy. LVEF decreased by - 2.40% ± 1.79 (p < 0.001), while global longitudinal strain (GLS) declined by 2.25 ± 1.36 (p < 0.001). HS troponin I levels showed a significant increase of 17.27 ± 36.29 (p < 0.001), and NT-proBNP levels rose by 321.23 ± 85.34 (p < 0.001). The study also identified a higher risk of cardiotoxicity in patients over 50 years of age. CONCLUSION The CHOP regimen poses significant risks of subclinical cardiac dysfunction and structural abnormalities in NHL patients, with cumulative doxorubicin dosage being a key contributor. Early detection using sensitive biomarkers like GLS and NT-proBNP is crucial for identifying cardiotoxicity and enabling timely interventions.
Collapse
Affiliation(s)
- Imanita Septianda
- Faculty of Medicine, Airlangga University, Surabaya, Indonesia.
- Dr. Soetomo General Academic Hospital, Surabaya, Indonesia.
| | - Mochamad Yusuf Alsagaff
- Faculty of Medicine, Airlangga University, Surabaya, Indonesia
- Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Budi Susetyo Pikir
- Faculty of Medicine, Airlangga University, Surabaya, Indonesia
- Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Ami Ashariati
- Faculty of Medicine, Airlangga University, Surabaya, Indonesia
- Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Muhamad Robiul Fuadi
- Faculty of Medicine, Airlangga University, Surabaya, Indonesia
- Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Fatimah Zahra
- Faculty of Medicine, Airlangga University, Surabaya, Indonesia
- Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| |
Collapse
|
7
|
Carrizales-Sepúlveda EF, Hernández-Rangel E, Ordaz-Farías A. A Deeper Dive Into the Myocardium for the Detection of Cancer Therapy-Related Cardiac Dysfunction: Radiomics in Cardio-Oncology. Echocardiography 2025; 42:e70174. [PMID: 40298932 DOI: 10.1111/echo.70174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/30/2025] Open
Affiliation(s)
- Edgar Francisco Carrizales-Sepúlveda
- Heart Failure Unit, Cardiology Service, Hospital Universitario "Dr. José E. González", Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, México
| | - Eduardo Hernández-Rangel
- Multimodal Cardiac Imaging Laboratory, Cardiology Service, Hospital Universitario "Dr. José E. González", Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, México
| | - Alejandro Ordaz-Farías
- Echocardiography Laboratory, Cardiology Service, Hospital Universitario "Dr. José E. González", Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, México
| |
Collapse
|
8
|
Wang J, Kong F, Wang Y, Yu J, Liu Y, Wu W, Liu Y, Gao P, Cheng Z, Cheng K, Deng H, Lai J, Fan J, Zhang L, Fang Q, Chen T, Yang D. Pacing therapy for immune checkpoint inhibitors-associated atrioventricular block: a single-center cohort study. BMC Cardiovasc Disord 2025; 25:319. [PMID: 40275132 PMCID: PMC12023490 DOI: 10.1186/s12872-025-04764-y] [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: 12/13/2024] [Accepted: 04/14/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND ICI-associated myocarditis is an uncommon yet potentially fatal condition, particularly when concomitant with atrioventricular block (AVB) necessitating pacing. The role of pacing therapy for ICI-associated AVB remains unknown. OBJECTIVES The aim of this study is to investigate the efficacy and safety of pacing therapy for ICI-associated AVB. METHODS Patients with ICI-associated myocarditis admitted to Peking Union Medical College Hospital from May 1st 2019 to April 30th 2024 were consecutively screened and the patients with AVB requiring pacing therapy were retrospectively included. Baseline clinical characteristics and initial temporary pacing therapy were evaluated. Follow-up assessments were conducted to evaluate the survival rate and the recovery of atrioventricular conduction. RESULTS A total of 43 patients with ICI-associated myocarditis were screened. Among them, a total of 11 (11/43, 25.6%) patients (mean age 64.5 ± 8.6 years, female 18.2%) were diagnosed with advanced or complete AVB and subsequently underwent pacing therapy. Short-term (within 90-days after procedure) survival rate was 72.7% (8/11). Atrioventricular conduction recovered in 4 (4/11, 36.4%) patients, without AVB recurrence after temporary pacemaker removal. For safety endpoints, right ventricular (RV) pacing parameters including pacing threshold, sensing amplitude and impedance were acceptable and no procedure-related complications occurred except RV temporary active fixation lead dislodgement in 1 patient (1/11, 9.1%). No pacing system related-infection occurred. CONCLUSIONS Pacing therapy for ICI-associated AVB demonstrates both safety and efficacy. ICI-associated AVB shows a high rate of recovery. Temporary pacemaker with active fixation lead may be a reasonable option for the initial pacing therapy.
Collapse
Affiliation(s)
- Jiaqi Wang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Fanyi Kong
- Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yifan Wang
- Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiaqi Yu
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Yingxian Liu
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Wei Wu
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Yongtai Liu
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Peng Gao
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Zhongwei Cheng
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Kang'an Cheng
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Hua Deng
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Jinzhi Lai
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Jingbo Fan
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Lihua Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Quan Fang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Taibo Chen
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China.
| | - Deyan Yang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
9
|
Patel S, Dave K, Garcia MJ, Gongora CA, Travin MI, Zhang L. Multimodal Imaging of Immune Checkpoint Inhibitor Myocarditis. J Clin Med 2025; 14:2850. [PMID: 40283680 PMCID: PMC12028134 DOI: 10.3390/jcm14082850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 04/17/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) have dramatically changed the landscape of cancer treatment and are increasingly used either as monotherapy or in combination with other ICIs, chemotherapy, and molecularly targeted agents. ICI myocarditis is a rare but potentially fatal irAE associated with the use of ICI characterized by T-cell mediated cardiomyocyte death. Diagnosing ICI myocarditis can be intricate as its atypical presentations. Multimodal imaging plays a crucial role in the diagnosis and risk stratification of ICI myocarditis. Current management strategies for ICI myocarditis include corticosteroids and immunosuppressants. Multidisciplinary collaboration is vital in these cases-combining oncology expertise with cardiology insights.
Collapse
Affiliation(s)
- Shreyans Patel
- Division of Cardiology, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; (M.J.G.); (C.A.G.); (L.Z.)
| | - Kartikeya Dave
- Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA;
| | - Mario J. Garcia
- Division of Cardiology, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; (M.J.G.); (C.A.G.); (L.Z.)
| | - Carlos A. Gongora
- Division of Cardiology, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; (M.J.G.); (C.A.G.); (L.Z.)
| | - Mark I. Travin
- Division of Nuclear Medicine, Department of Radiology, Montefiore Medical Center and the Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA;
| | - Lili Zhang
- Division of Cardiology, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; (M.J.G.); (C.A.G.); (L.Z.)
| |
Collapse
|
10
|
Ryan TD, Bates JE, Kinahan KE, Leger KJ, Mulrooney DA, Narayan HK, Ness K, Okwuosa TM, Rainusso NC, Steinberger J, Armenian SH. Cardiovascular Toxicity in Patients Treated for Childhood Cancer: A Scientific Statement From the American Heart Association. Circulation 2025; 151:e926-e943. [PMID: 40104841 DOI: 10.1161/cir.0000000000001308] [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: 03/20/2025]
Abstract
The field of cardio-oncology has expanded over the past 2 decades to address the ever-increasing issues related to cardiovascular disease in patients with cancer and survivors. There is increasing recognition that nearly all cancer treatments pose some short- or long-term risk for development of cardiovascular disease and that pediatric patients with cancer may be especially vulnerable to cardiovascular disease because of young age at treatment and expected long life span afterward. Anthracycline chemotherapy and chest-directed radiotherapy are the most well-studied cardiotoxic therapies, and dose reduction, use of cardioprotection for anthracyclines, and modern radiotherapy approaches have contributed to improved cardiovascular outcomes for survivors. Newer treatments such as small-molecule inhibitors, antibody-based cytotoxic therapy, and immunotherapy have expanded options for previously difficult-to-treat cancers but have also revealed new cardiotoxic profiles. Application of effective surveillance strategies in patients with cancer and survivors has been a focus of practitioners and researchers, whereas the prevention and treatment of extant cardiovascular disease is still developing. Incorporation of new strategies in an equitable manner and appropriate transition from pediatric to adult care will greatly influence long-term health-related outcomes in the growing population of childhood cancer survivors at risk for cardiovascular disease.
Collapse
|
11
|
Biondi F, Madonna R. The Potential Role of GLP1-RAs Against Anticancer-Drug Cardiotoxicity: A Scoping Review. J Clin Med 2025; 14:2705. [PMID: 40283534 PMCID: PMC12027986 DOI: 10.3390/jcm14082705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 04/05/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025] Open
Abstract
Background: GLP1 receptor agonists (GLP1-RAs) have become a central component in the treatment of type 2 diabetes mellitus (T2DM) and are gaining prominence in the cardiovascular field. Semaglutide and other GLP1-RA molecules possess cardioprotective properties. Cardiotoxicity, a term used to refer to cardiovascular disease caused by anticancer treatment, is a collection of common and severe conditions. Its pharmacological prevention or mitigation is a clinical unmet need as options are few and limited to some specific clinical settings. GLP1-RAs have a promising pharmacological profile given their activity on a number of pathophysiological targets and signaling pathways including oxidative stress, autophagy, and STAT3 activation. Interestingly, abnormalities in some of the GLP-1-modulated pathways have been linked to cardiotoxicity. This scoping review aims to map the extent and assess the main characteristics of research on the role of GLP1-RAs in the prevention and/or mitigation of anticancer-related cardiotoxicity. Methods: The selection process led to the inclusion of thirteen studies chosen from reports retrieved through the search string: ("semaglutide" OR "exenatide" OR "liraglutide" OR "dulaglutide" OR "tirzepatide" OR "GLP1 receptor agonist" OR "GLP1RA" OR "GLP1-RA" OR "GLP1" OR "Glucagon-like Peptide-1 Agonists") AND ("cardioncology" OR "cardiotoxicity" OR "chemotherapy" OR "anti-cancer treatment" OR "anti-cancer therapy"). The study complied with the PRISMA guidelines on scoping reviews. Results: Two studies were clinical and conducted on registries, eight used animal models, two were conducted on cell cultures, and one was conducted on both animal models and cell cultures. Evidence in favor of cardioprotection and a number of putative mechanisms emerged. Conclusions: Evidence on GLP1-RAs' effect on cardiotoxicity is limited in both quantity and quality and suffers from poor study standardization. However, most included studies documented a rigorously defined cardioprotective effect and demonstrated changes in several pathophysiologically relevant targets and pathways, including NF-κB, IL-6, reactive oxygen species, and caspase-3. Further clinical studies are warranted.
Collapse
Affiliation(s)
- Filippo Biondi
- Department of Pathology, Cardiology Division, University of Pisa, Via Paradisa, 56124 Pisa, Italy
| | - Rosalinda Madonna
- Department of Pathology, Cardiology Division, University of Pisa, Via Paradisa, 56124 Pisa, Italy
| |
Collapse
|
12
|
Pereyra Pietri M, Farina JM, Scalia IG, Roarke M, Mahmoud AK, Masson R, Wasef B, Tagle-Cornell C, Kenyon CR, Abbas MT, Baba Ali N, Awad K, Kamel MA, Said EF, O'Shea M, Barry T, Narayanasamy H, Ray JC, El Masry H, Larsen CM, Herrmann J, Arsanjani R, Ayoub C. Characterization of Cardiovascular Events and Prognosis in Immune Checkpoint Inhibitor-Related Myocarditis. Mayo Clin Proc 2025:S0025-6196(24)00693-1. [PMID: 40202477 DOI: 10.1016/j.mayocp.2024.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/31/2024] [Accepted: 12/19/2024] [Indexed: 04/10/2025]
Abstract
OBJECTIVE To evaluate the incidence, timing, and characteristics of immune checkpoint inhibitor-related myocarditis (ICIrM) and associated cardiovascular events at 3-year follow-up. METHODS All patients treated with immune checkpoint inhibitors (ICIs) at a multicenter institution from 2011 to 2022 were retrospectively reviewed for ICIrM. A propensity score-matched control group was identified from patients treated with ICIs without development of myocarditis (ratio of 1:4). Baseline characteristics, cardiovascular events, and mortality outcomes were manually curated during extended 3-year follow-up. Major adverse cardiovascular events (MACE) were defined as transient ischemic attack/stroke, heart failure, and myocardial infarction. RESULTS Of 5423 patients treated with ICIs, ICIrM occurred in 59 (1.1%), and 236 propensity score-matched patients who received ICIs without myocarditis were identified as controls. Mean age was 68.5 ± 12.3 years; 65.4% were male. Median time to development of ICIrM was 44 days (interquartile range, 28 to 102 days), with median troponin value of 364 ng/L (interquartile range, 115 to 1224 ng/L). Patients with ICIrM had increased risk of cardiac death (hazard ratio [HR], 34.0; 95% CI, 7.8 to 148.0; P<.001), MACE (HR, 5.0; 95% CI, 3.1 to 8.1; P<.001), ventricular tachycardia (HR, 12.3; 95% CI, 1.3 to 118.4; P=.03), and complete heart block (HR, 2.3; 95% CI, 1.0 to 5.1; P=.046); these occurred predominantly within 120 days after diagnosis of ICIrM. Triple-M syndrome (myocarditis, myasthenia, and myositis) occurred in 12 (20.3%), with increased risk for all-cause mortality (HR, 2.1; 95% CI, 1.0 to 4.1; P=.04) but not for cardiac death or MACE. CONCLUSION Immune checkpoint inhibitor-related myocarditis is associated with increased cardiovascular events that are further characterized on extended follow-up, with most occurring in the first 4 months after diagnosis.
Collapse
Affiliation(s)
| | - Juan M Farina
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ
| | - Isabel G Scalia
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ
| | - Michael Roarke
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ
| | - Ahmed K Mahmoud
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ
| | - Rajeev Masson
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ
| | - Beman Wasef
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ
| | | | | | | | - Nima Baba Ali
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ
| | - Kamal Awad
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ
| | - Moaz A Kamel
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ
| | - Ebram F Said
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ
| | - Michael O'Shea
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ
| | - Timothy Barry
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ
| | | | - Jordan C Ray
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL
| | - Hicham El Masry
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ
| | | | - Joerg Herrmann
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Reza Arsanjani
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ
| | - Chadi Ayoub
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ.
| |
Collapse
|
13
|
Dobbin SJH, Mangion K, Berry C, Roditi G, Basak S, McClure JD, Brooksbank K, Sonecki P, Sourbron S, Evans J, White J, Welsh P, Butler E, Venugopal B, Touyz RM, Jones RJ, Petrie MC, Lang NN. Vascular endothelial growth factor inhibitor-induced cardiotoxicity: prospective multimodality assessment incorporating cardiovascular magnetic resonance imaging. Heart 2025:heartjnl-2024-325535. [PMID: 40180444 DOI: 10.1136/heartjnl-2024-325535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 03/19/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Vascular endothelial growth factor inhibitors (VEGFIs) are effective anticancer agents, but are associated with cancer therapy-related cardiac dysfunction (CTRCD) and hypertension. The timing, frequency and magnitude of these toxicities are poorly defined. The objective of this study is therefore to investigate the incidence, time course and mechanisms of VEGFI-associated CTRCD and hypertension. METHODS Patients commencing VEGFI underwent blood pressure (BP) monitoring, echocardiography and cardiac biomarker measurement at baseline and prospectively over 24 weeks. Serial adenosine stress perfusion cardiovascular MRI (CMR) was performed in a substudy. CTRCD was defined as left ventricular ejection fraction (LVEF) decline by ≥10 percentage points from baseline to a value <50%. RESULTS 78 patients participated (68% men; age 63±11 years). 15 patients (19%) developed CTRCD, and it was evident at 4 weeks in 93% of cases. Overall, LVEF was 4.2% (95% CI: -6.2% to -2.3%, p<0.001) lower than baseline at 4 weeks. At 4 weeks, N-terminal pro-brain natriuretic peptide, but not troponin, was higher in patients with CTRCD. 62 (77%) patients developed hypertension. Home systolic and diastolic BP increased by 7.2 mm Hg (4.7-9.8, p<0.001) and 4.8 mm Hg (3.1-6.5, p<0.001), respectively, at 1 week. There was no association between change in LVEF and BP.CMR-derived LVEF, T1 relaxation times and resting myocardial blood flow (n=46) were 5.2% (-7.3% to -3.1%, p<0.001), 27 ms (-40 to -14, p<0.001) and 14.7 mL/100mL/min (-24.2 to -5.1, p=0.004), respectively, lower at 4 weeks. CONCLUSION VEGFI-associated CTRCD is frequent and occurs early. This finding has implications for prioritising early cardiac imaging follow-up after commencing treatment. Underlying mechanisms include myocardial and microvascular effects that are at least partly independent of hypertension.
Collapse
Affiliation(s)
- Stephen J H Dobbin
- University of Glasgow BHF Glasgow Cardiovascular Research Centre, Glasgow, UK
| | - Kenneth Mangion
- University of Glasgow BHF Glasgow Cardiovascular Research Centre, Glasgow, UK
| | - Colin Berry
- University of Glasgow BHF Glasgow Cardiovascular Research Centre, Glasgow, UK
- Cardiology, Golden Jubilee National Hospital, Clydebank, UK
| | - Giles Roditi
- Department of Radiology, Glasgow Royal Infirmary, Glasgow, UK
| | | | - John D McClure
- University of Glasgow BHF Glasgow Cardiovascular Research Centre, Glasgow, UK
| | - Katriona Brooksbank
- University of Glasgow BHF Glasgow Cardiovascular Research Centre, Glasgow, UK
| | | | - Steven Sourbron
- Department of Infection, Immunity and Cardiovascular Disease, The University of Sheffield-Western Bank Campus, Sheffield, UK
| | - Jeff Evans
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Jeff White
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Paul Welsh
- University of Glasgow BHF Glasgow Cardiovascular Research Centre, Glasgow, UK
| | - Elaine Butler
- University of Glasgow BHF Glasgow Cardiovascular Research Centre, Glasgow, UK
| | | | - Rhian M Touyz
- Research Institute of McGill University Health Centre, McGill University, Montreal, Quebec, Canada
| | - Robert J Jones
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Mark C Petrie
- University of Glasgow BHF Glasgow Cardiovascular Research Centre, Glasgow, UK
| | - Ninian N Lang
- University of Glasgow BHF Glasgow Cardiovascular Research Centre, Glasgow, UK
| |
Collapse
|
14
|
Jeon MH, DiSipio T, Wilson L, Garvey G, Diaz A. Assessment and stratification of cardiovascular disease risk in people diagnosed with breast cancer: A scoping review. Cancer Treat Rev 2025; 135:102903. [PMID: 40054314 DOI: 10.1016/j.ctrv.2025.102903] [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: 12/17/2024] [Revised: 01/31/2025] [Accepted: 02/16/2025] [Indexed: 04/08/2025]
Abstract
BACKGROUND Breast cancer patients are at increased risk of cardiovascular disease, which often are associated with cardiotoxic breast cancer treatment or overlapping risk factors between the two diseases. Pre-treatment cardiovascular risk assessment can enable accurate risk stratification and prevention of cardiovascular disease. Several tools have been suggested, described or used in research to assess baseline (pre-treatment) risk to determine appropriate cardiovascular disease care before, during and after cancer treatment. This scoping review aims to identify and describe key features of baseline cardiovascular disease risk assessment tools for breast cancer patients. METHODS PubMed, Embase and Google Scholar were searched for articles published January 2013 - March 2024 to identify publications reporting cardiovascular disease risk assessment tools in breast cancer patients. Publications included research articles (observational and experimental studies) and position/policy, commentary and review papers. Eligibility was assessed and key data were extracted independently by two reviewers. Conflicts were discussed and resolved with the authorship team. RESULTS A total 144 articles were identified. Of these, 57 reported original data for the development, validation or recommendations of cardiovascular disease risk assessment tools and 87 reported the use of such tools. From these articles, 13 tools were identified that assessed the risk of cardiovascular disease broadly (n = 3) or death due to cardiovascular disease (n = 1) or specifically of cardiotoxicity or heart failure (n = 8) or venous thromboembolism (n = 1) in people diagnosed with breast cancer. Fourteen tools assessed cardiovascular disease risk in people diagnosed with mixed cancer types, including breast cancer. The planned development of four tools and/or surveillance pathways were described in protocol papers. Among all these tools identified (n = 31), seven tools (among these, four tools assessed people diagnosed with breast cancer only) went through external validation and performed poorly or moderately in stratifying cancer patients effectively into risk categories. Risk factors included in the assessment tools were age, breast cancer treatment type and pre-existing cardiovascular disease. While clinical guidelines and recommendations about baseline cardiovascular disease risk assessment were identified, these were either for cancer patients broadly or for cancer treatment types, and not specifically for people diagnosed with breast cancer. CONCLUSION Several tools to assess baseline cardiovascular disease in people diagnosed with breast cancer were identified but only seven tools had gone through a validation process, and none were found to be very effective in differentiating people by baseline cardiovascular disease risk. Further work is needed to optimise the effectiveness of baseline cardiovascular disease risk assessments for breast cancer patients to enable appropriate stratification and monitoring of risk before, during and after treatment to improve cardiovascular health and outcomes.
Collapse
Affiliation(s)
- Mi Hye Jeon
- School of Public Health, The University of Queensland, Herston 4030, Australia.
| | - Tracey DiSipio
- School of Public Health, The University of Queensland, Herston 4030, Australia.
| | - Louise Wilson
- School of Public Health, The University of Queensland, Herston 4030, Australia.
| | - Gail Garvey
- School of Public Health, The University of Queensland, Herston 4030, Australia.
| | - Abbey Diaz
- School of Public Health, The University of Queensland, Herston 4030, Australia; Yardhura Walani National Centre for Aboriginal and Torres Strait Islander Wellbeing Research, The Australian National University, Canberra 0200, Australia.
| |
Collapse
|
15
|
Li C, Ma J, Wang Q, Ma L, Han J, Qi Y, Pei X, Yu J. Apatinib-Induced Hypertension Correlates with Improved Prognosis in Solid Tumor Patients. Cardiovasc Toxicol 2025; 25:570-581. [PMID: 40048128 DOI: 10.1007/s12012-025-09980-8] [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: 12/07/2024] [Accepted: 02/21/2025] [Indexed: 03/15/2025]
Abstract
This study evaluated the occurrence of apatinib-induced hypertension and its impact on the prognosis of patients with solid tumors. A retrospective cohort study with prospective follow-up was conducted on 769 patients treated with apatinib from 2014 to 2021 across three hospitals. Patients were categorized into hypertension and non-hypertension groups. The primary outcome was overall survival (OS), with progression-free survival (PFS) as a secondary outcome. Apatinib-induced hypertension occurred in 33.3% of patients and was associated with significantly longer OS (HR 0.40, 95% CI [0.37-0.48], p < 0.0001) and PFS (HR 0.41, 95% CI [0.35-0.49], p < 0.001). Subgroup analysis confirmed these findings in all cancer types, except for PFS in non-small cell lung cancer. Hypertension may serve as a predictive biomarker for improved anti-tumor efficacy.
Collapse
Affiliation(s)
- Caie Li
- Center of Hypertension, Department of Cardiology, The Second Hospital of Lanzhou University, Lanzhou, 730030, China
| | - Jie Ma
- Center of Hypertension, Department of Cardiology, The Second Hospital of Lanzhou University, Lanzhou, 730030, China
| | - Qiongying Wang
- Center of Hypertension, Department of Cardiology, The Second Hospital of Lanzhou University, Lanzhou, 730030, China
| | - Liping Ma
- Department of Cardiology, The First People's Hospital of Tianshui, Tianshui, 741000, China
| | - Juncheng Han
- Center of Hypertension, Department of Cardiology, The Second Hospital of Lanzhou University, Lanzhou, 730030, China
- Department of Cardiology, The First People's Hospital of Lanzhou City, Lanzhou, China
| | - Yali Qi
- Department of Oncology, The Second Hospital of Lanzhou University, Lanzhou, 730030, China
| | - Xiaxia Pei
- Department of Oncology, The Second Hospital of Lanzhou University, Lanzhou, 730030, China
| | - Jing Yu
- Center of Hypertension, Department of Cardiology, The Second Hospital of Lanzhou University, Lanzhou, 730030, China.
| |
Collapse
|
16
|
Ali A, Koutroumpakis E, Song J, Booser D, Barcenas CH, Tripathy D, Barac A, Palaskas NL, Deswal A. Risk Stratification for Trastuzumab-Induced Cardiac Dysfunction and Potential Implications for Surveillance. JACC CardioOncol 2025; 7:203-215. [PMID: 40246379 PMCID: PMC12046757 DOI: 10.1016/j.jaccao.2024.12.007] [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: 06/12/2024] [Revised: 12/09/2024] [Accepted: 12/16/2024] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Although patient factors and sequential anthracycline use contribute to risk for cancer therapy-related cardiac dysfunction (CTRCD) with HER2-directed cancer therapy, frequent (every 3 months) left ventricular ejection fraction (LVEF) surveillance is recommended irrespective of baseline risk. OBJECTIVES The aim of this study was to examine the incidence of trastuzumab-associated CTRCD in a contemporary cohort with HER2-positive breast cancer and assess the performance of a risk assessment tool to identify patients at low risk for CTRCD to guide risk-based surveillance strategies. METHODS A retrospective cohort of patients with HER2-positive breast cancer treated with trastuzumab at a tertiary cancer center was examined. Patients were categorized as low, medium, and high or very high risk for CTRCD by Heart Failure Association/International Cardio-Oncology Society risk assessment. RESULTS Of 496 patients treated with trastuzumab, 29.8% also received anthracyclines. Over a median follow-up period of 51 months, 8.7% developed CTRCD, but only 1.6% had associated heart failure (HF). CTRCD rates were 3.6%, 12.8%, and 32.1% in low-risk, medium-risk, and high or very high risk groups, respectively. HF incidence was 0.4% in the low-risk group and 2.1% in the medium-risk group, with no HF in patients at low- or medium-risk who received trastuzumab without anthracyclines. HF was observed in 11% of high-risk patients. The risk assessment had a negative predictive value for CTRCD in low vs moderate- or high-risk patients of 96.4% (95% CI: 93.5%-98.3%). CONCLUSIONS The findings support the exploration of a prospective personalized risk-based approach to cardiac LVEF surveillance during trastuzumab therapy. Less frequent LVEF monitoring in low-risk patients may optimize resource use and reduce patient burden without compromising safety.
Collapse
Affiliation(s)
- Abdelrahman Ali
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA. https://twitter.com/Abdelcards
| | - Efstratios Koutroumpakis
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA. https://twitter.com/EKoutroumpakis
| | - Juhee Song
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Daniel Booser
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Carlos H Barcenas
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Debu Tripathy
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA. https://twitter.com/DrDebuTripathy
| | - Ana Barac
- Inova Schar Heart and Vascular, Inova Schar Cancer, Fairfax, Virginia, USA. https://twitter.com/AnaBaracCardio
| | - Nicolas L Palaskas
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA. https://twitter.com/PalaskasN
| | - Anita Deswal
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
| |
Collapse
|
17
|
Jensen G, Wang X, Kuempel J, Palaskas N, Chen Z, Yu W, Chen Y, Mohammad H, Luo W, Chang J. Immune checkpoint inhibitor-associated myocarditis: a historical and comprehensive review. Am J Physiol Heart Circ Physiol 2025; 328:H734-H751. [PMID: 39925096 DOI: 10.1152/ajpheart.00687.2024] [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: 10/04/2024] [Revised: 11/13/2024] [Accepted: 01/03/2025] [Indexed: 02/11/2025]
Abstract
The most fatal side effect associated with revolutionary immune checkpoint inhibitor (ICI) cancer therapies is myocarditis, a rare and devastating complication with a mortality rate approaching 40%. This review comprehensively examines the limited knowledge surrounding this recently recognized condition, emphasizing the absence of evidence-based therapeutic strategies, diagnostic modalities, and reliable biomarkers that hinder effective management. It explores advancements in preclinical models that are uncovering disease mechanisms and enabling the identification of therapeutic targets. These efforts have informed the design of early clinical trials aimed at reducing mortality. With the growing prevalence of ICI therapies in oncology, addressing critical gaps, such as long-term outcomes and risk stratification, has become increasingly urgent. By synthesizing current evidence, this work seeks to enhance understanding and guide the development of strategies to improve patient outcomes and ensure the continued safe use of ICIs in cancer care.
Collapse
Affiliation(s)
- Garrett Jensen
- Institute for Biosciences and Technology, Center for Genomics and Precision Medicine, Texas A&M University, Houston, Texas, United States
| | - Xinjie Wang
- Institute for Biosciences and Technology, Center for Genomics and Precision Medicine, Texas A&M University, Houston, Texas, United States
| | - Jacob Kuempel
- Institute for Biosciences and Technology, Center for Genomics and Precision Medicine, Texas A&M University, Houston, Texas, United States
| | - Nicolas Palaskas
- Department of Cardiology, MD Anderson Cancer Center, Houston, Texas, United States
| | - Zhishi Chen
- Institute for Biosciences and Technology, Center for Genomics and Precision Medicine, Texas A&M University, Houston, Texas, United States
| | - Wei Yu
- Institute for Biosciences and Technology, Center for Genomics and Precision Medicine, Texas A&M University, Houston, Texas, United States
| | - Yanping Chen
- Institute for Biosciences and Technology, Center for Genomics and Precision Medicine, Texas A&M University, Houston, Texas, United States
| | - Haseeb Mohammad
- Texas A&M University College of Medicine, Houston, Texas, United States
| | - Weijia Luo
- Institute for Biosciences and Technology, Center for Genomics and Precision Medicine, Texas A&M University, Houston, Texas, United States
| | - Jiang Chang
- Institute for Biosciences and Technology, Center for Genomics and Precision Medicine, Texas A&M University, Houston, Texas, United States
| |
Collapse
|
18
|
Levytska K, Naumann RW, Benfield MJ, Brown J, Casablanca Y, Lees B, Puechl AM, Crane EK. Pegylated liposomal doxorubicin does not affect cardiac function in patients treated for gynecologic malignancies. Gynecol Oncol Rep 2025; 58:101727. [PMID: 40201899 PMCID: PMC11978316 DOI: 10.1016/j.gore.2025.101727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/13/2025] [Accepted: 03/15/2025] [Indexed: 04/10/2025] Open
Abstract
Objective Although pegylated liposomal doxorubicin (PLD) has a more favorable side-effect profile compared to doxorubicin, the FDA label for PLD includes a warning listing cardiotoxicity. Our objective was to evaluate predictors of pre- and post-treatment cardiac testing and quantify the effect of PLD on cardiac function in patients treated for gynecologic malignancies. Methods Retrospective chart review of gynecologic oncology patients who received PLD over a 10-year period at a single institution. Cardiac studies were aligned to PLD treatment and ejection fractions (EF) were compared pre- and post-treatment. Results A total of 453 patients who had received PLD were identified; 216 (48 %) had pre-PLD treatment cardiac function testing. Predictors of pre-chemotherapy testing were diabetes (p = 0.015), higher ECOG score (p = 0.004), and cardiac disease (p = 0.032). Eighty-three (18.3 %) patients had pre- and post-PLD treatment cardiac function testing. Predictors of pre- and post- testing were number of cycles of PLD (p < 0.0001) and total dose of PLD (p < 0.0001). Seventy-five (90 %) patients had no change in EF (defined as < 10 %), while 2 (2.4 %) had improvement in EF > 10 %, and 6 (7.2 %) had a decrease in EF > 10 %. Initial EF in patients with > 10 % decrease was higher than in those without change or improvement (p = 0.0004). One (1.2 %) patient had a clinically significant decrease in EF (32.5 %) resulting in interruption of treatment. Conclusion Risk of cardiac toxicity from administration of PLD for patients undergoing treatment for gynecologic cancers appears to be low. Selective screening of cardiac function should be employed for these patients.
Collapse
Affiliation(s)
- Khrystyna Levytska
- Division of Gynecologic Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - R. Wendel Naumann
- Division of Gynecologic Oncology, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Miranda J. Benfield
- Division of Gynecologic Oncology, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Jubilee Brown
- Division of Gynecologic Oncology, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Yovanni Casablanca
- Division of Gynecologic Oncology, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Brittany Lees
- Division of Gynecologic Oncology, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Allison M. Puechl
- Division of Gynecologic Oncology, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Erin K. Crane
- Division of Gynecologic Oncology, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| |
Collapse
|
19
|
Fanelli E, Picca G, Airale L, Astarita A, Mingrone G, Catarinella C, Votta S, Colomba A, Cesareo M, Leone D, Paladino A, Rabbia F, Bringhen S, Gay F, Veglio F, Milan A, Vallelonga F. Blood pressure variability as predictor of cancer therapy-related cardiovascular toxicity in patients with Multiple Myeloma. Hypertens Res 2025; 48:1554-1563. [PMID: 39843857 DOI: 10.1038/s41440-024-02084-w] [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: 09/14/2024] [Revised: 12/14/2024] [Accepted: 12/21/2024] [Indexed: 01/24/2025]
Abstract
Blood pressure (BP) variability (BPV) is an independent predictor of cardiovascular (CV) events. The role of BPV in defining risk of cancer therapy-related cardiovascular toxicity (CTR-CVT) is currently unknown. The aims of this study were: (i) to evaluate BPV in a population of patients with Multiple Myeloma, undergoing proteasome inhibitors therapy; (ii) to assess the predictive value of BPV for CTR-CVT; (iii) to analyze clusters of subjects based on BPV. One hundred twenty-four patients underwent a baseline evaluation, including Ambulatory Blood Pressure Monitoring (ABPM), PWV, and Echocardiography. BPV was assessed through ABPM-based standard deviation (SD), weighted standard deviation (wSD), coefficient of variation (CoV), average real variability (ARV), and variability independent of the mean (VIM). Individuals who developed CTR-CVT had a higher baseline BPV. Furthermore, night-time BPV was associated with CTR-CVT, independently of age, smoking, BP, diabetes, dyslipidemia, and kidney function (night-time systolic CoV: adjusted OR 1.09 [1.01-1.21]; night-time systolic VIM: adjusted OR 1.18 [1.01-1.39]). Cut-offs for these BPV parameters were identified as predictors of CTR-CVT occurrence: 10.5 for night-time systolic CoV; 7.8 and 6.4 for systolic and diastolic night-time VIM. Clustering analysis identified subgroups of subjects characterized by the highest BPV, who had a greater prevalence of events, but no differences in other CV risk determinants. Short-term BPV is an independent predictor of CTR-CVT. BPV may enhance the precision of risk stratification in cancer patients, enabling identification of individuals at higher risk who would not be recognized, if traditional prognostic indicators were the sole applied criteria. On the left panel in the figure, the distribution of blood pressure variability (BPV) in the population according to cancer therapy-related cardiovascular toxicity occurrence; in the central panel, association of blood pressure variability with events and cutoffs values; in the right panel, clustering analysis results based on BPV levels. Histogram and radar plot represent events and BPV indexes distribution in the three clusters, respectively. ARV, average real variability; BPV, Blood Pressure Variability; CTR-CVT, cancer therapy-related cardiovascular toxicity; CoV, coefficient of variation; DBP, Diastolic blood pressure; SBP, Systolic blood pressure; SD, standard deviation; VIM, variability independent of the mean; wSD, weighted standard deviation.
Collapse
Affiliation(s)
- Elvira Fanelli
- Division of Internal Medicine, Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy.
- Emergency Medicine Unit, Ospedale San Giovanni Bosco, Turin, Italy.
- Division of Internal Medicine, Hypertension Unit, A.O.U. Città della Salute e della Scienza di Torino, Department of Medical Sciences, University of Turin, Turin, Italy.
| | - Giulia Picca
- Division of Internal Medicine, Hypertension Unit, A.O.U. Città della Salute e della Scienza di Torino, Department of Medical Sciences, University of Turin, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Lorenzo Airale
- Division of Internal Medicine, Hypertension Unit, A.O.U. Città della Salute e della Scienza di Torino, Department of Medical Sciences, University of Turin, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Anna Astarita
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Giulia Mingrone
- Division of Internal Medicine, Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Cinzia Catarinella
- Division of Internal Medicine, Hypertension Unit, A.O.U. Città della Salute e della Scienza di Torino, Department of Medical Sciences, University of Turin, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Simona Votta
- Division of Internal Medicine, Hypertension Unit, A.O.U. Città della Salute e della Scienza di Torino, Department of Medical Sciences, University of Turin, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Anna Colomba
- Division of Internal Medicine, Hypertension Unit, A.O.U. Città della Salute e della Scienza di Torino, Department of Medical Sciences, University of Turin, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Marco Cesareo
- Division of Internal Medicine, Hypertension Unit, A.O.U. Città della Salute e della Scienza di Torino, Department of Medical Sciences, University of Turin, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Dario Leone
- Division of Internal Medicine, Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Arianna Paladino
- Division of Internal Medicine, Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Franco Rabbia
- Division of Internal Medicine, Hypertension Unit, A.O.U. Città della Salute e della Scienza di Torino, Department of Medical Sciences, University of Turin, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Sara Bringhen
- SSD Clinical Trial in Oncoematologia e Mieloma Multiplo, Division of Hematology, AOU Città della Salute e della Scienza di Torino, Turin, Italy
| | - Francesca Gay
- SSD Clinical Trial in Oncoematologia e Mieloma Multiplo, Division of Hematology, AOU Città della Salute e della Scienza di Torino, Turin, Italy
- Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Franco Veglio
- Division of Internal Medicine, Hypertension Unit, A.O.U. Città della Salute e della Scienza di Torino, Department of Medical Sciences, University of Turin, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Alberto Milan
- Division of Internal Medicine, Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Fabrizio Vallelonga
- Division of Internal Medicine, Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
20
|
Itzhaki Ben Zadok O, O'Hare MJ, Nohria A. Immune Checkpoint Inhibitor-Related Myocarditis With or Without Concomitant Myopathy: Clinical Findings and Cardiovascular Outcomes. JACC CardioOncol 2025; 7:252-264. [PMID: 40246383 PMCID: PMC12046767 DOI: 10.1016/j.jaccao.2025.02.005] [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: 01/27/2025] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Data on cardiovascular outcomes in patients with both immune checkpoint inhibitor-induced immune-related myocarditis (irMyocarditis) and immune-related myopathy (irMyopathy) are limited. OBJECTIVES The aim of this study was to describe clinical characteristics and cardiovascular outcomes in patients with isolated irMyocarditis vs those with concomitant irMyocarditis and irMyopathy. METHODS A retrospective cohort study was conducted among patients diagnosed with irMyocarditis at Massachusetts General Brigham between 2015 and 2023. Clinical, laboratory, and imaging characteristics were evaluated, and cardiovascular outcomes were compared between patients with and those without concomitant irMyopathy. The outcomes assessed included acute heart failure requiring diuresis, significant arrhythmias (ventricular arrhythmias and high-degree atrioventricular block), and cardiovascular and all-cause mortality during the index hospitalization. RESULTS Among 101 patients with irMyocarditis, 32 (31.7%) had concomitant irMyopathy. Patients with irMyocarditis and irMyopathy had higher high-sensitivity troponin T (median 716 ng/L vs 75 ng/L; P < 0.001) and creatine kinase levels (median 3441 U/L vs 232 U/L; P < 0.001) and were more likely to present with significant arrhythmias (HR: 2.12; 95% CI: 1.13-3.97; P = 0.019). Conversely, patients with isolated irMyocarditis had higher N-terminal prohormone of brain natriuretic peptide levels (median 2043 pg/mL vs 606 pg/mL; P = 0.007), lower left ventricular ejection fractions (median 56% vs 65%; P = 0.008), and a higher likelihood of acute decompensated heart failure (HR: 5.88; 95% CI: 1.45-25; P = 0.013). Cardiovascular and all-cause death during admission were numerically higher in patients with concomitant irMyopathy but were not significantly different between the 2 groups. CONCLUSIONS Patients with irMyocarditis and irMyopathy and those with isolated irMyocarditis have distinct biomarker profiles and cardiovascular complications. These differences should be confirmed in larger prospective cohorts to guide tailored management strategies.
Collapse
Affiliation(s)
- Osnat Itzhaki Ben Zadok
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Meabh J O'Hare
- Division of Neuromuscular Medicine, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Anju Nohria
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
21
|
Pereyra Pietri M, Farina JM, Scalia IG, Mahmoud AK, Roarke M, Wasef B, Tagle-Cornell C, Kenyon CR, Abbas MT, Ali NB, Awad KA, Javadi N, Bismee NN, Larsen CM, Herrmann J, Arsanjani R, Ayoub C. Comparison of the diagnostic and prognostic value of criteria for immune checkpoint inhibitor related myocarditis. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2025; 11:30. [PMID: 40149000 PMCID: PMC11948924 DOI: 10.1186/s40959-025-00327-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 03/14/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Myocarditis is a dreaded complication of immune-checkpoint inhibitor (ICI) therapy but challenging to diagnose. There are no published data comparing the two leading diagnostic criteria for ICI-related myocarditis (ICIrM) and their association with cardiovascular events. METHODS In this retrospective cohort study, we reviewed all patients who underwent ICI therapy and had cardiac troponin assessment for possible myocarditis across three tertiary institutions from 2011 to 2022. ICIrM was adjudicated by the Bonaca et al. criteria and the ESC-ICOS guidelines. A propensity matched control group was identified of patients treated with ICI without developing myocarditis. Baseline characteristics and long-term outcomes, including cardiac death, MACE (myocardial infarction, TIA/stroke, heart failure), and arrhythmias data were curated, and patients diagnosed with ICIrM by each criteria were compared to controls for cardiovascular events. RESULTS A total of 59 patients (mean age was 73.1 ± 10.2 years, 60.1% male) were identified as having a diagnosis of ICIrM by Bonaca criteria (16 definite, 13 probable and 30 possible myocarditis). Forty-seven of these patients met the ESC-ICOS guidelines criteria, and all patients meeting either set of ICIrM criteria were treated with steroid therapy. At 3-year follow up, patients diagnosed with ICIrM by the Bonaca criteria had a high risk of cardiac mortality (HR 17.84, 95%CI 2.36-134.62, p = 0.005), MACE (HR 4.90, 95%CI 2.40-10.02, p < 0.001) and arrhythmias (HR 3.33, 95%CI 1.78-6.21, p < 0.001) when compared to matched controls. ICIrM by ESC-ICOS criteria was similarly predictive of cardiac mortality, MACE, and arrhythmias (HR 15.01, 95%CI 1.96-114.76, p = 0.009, HR 5.18, 95%CI 2.33-11.53, p < 0.001, and HR 3.41, 95%CI 1.73-6.70, p < 0.001 respectively). CONCLUSION The ESC-ICOS guidelines were more restrictive than the Bonaca et al. criteria for the diagnosis of ICIrM but similar in terms of prognostic value.
Collapse
Affiliation(s)
- Milagros Pereyra Pietri
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Juan M Farina
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Isabel G Scalia
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Ahmed K Mahmoud
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Michael Roarke
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Beman Wasef
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Cecilia Tagle-Cornell
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Courtney R Kenyon
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Mohammed Tiseer Abbas
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Nima Baba Ali
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Kamal A Awad
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Niloofar Javadi
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Nadera N Bismee
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Carolyn M Larsen
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Joerg Herrmann
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Reza Arsanjani
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Chadi Ayoub
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA.
| |
Collapse
|
22
|
Wan Z, Wang C, Luo S, Zhu J, He H, Hao K. Bridging the Gap Between hiPSC-CMs Cardiotoxicity Assessment and Clinical LVEF Decline Risk: A Case Study of 21 Tyrosine Kinase Inhibitors. Pharmaceuticals (Basel) 2025; 18:450. [PMID: 40283889 PMCID: PMC12030206 DOI: 10.3390/ph18040450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 03/15/2025] [Accepted: 03/19/2025] [Indexed: 04/29/2025] Open
Abstract
Objectives: There is growing concern over tyrosine kinase inhibitor (TKI)-induced cardiotoxicity, particularly regarding left ventricular dysfunction and heart failure in clinical treatment. These adverse effects often lead to treatment discontinuation, severely impacting patient outcomes. Therefore, there is an urgent need for more precise risk assessment methods. This study aimed to assess the cardiotoxicity of TKIs, refine in vitro to in vivo extrapolation (IVIVE) methodologies to improve predictive accuracy, and identify critical in vitro parameters for assessment. Methods: By leveraging high-throughput cardiotoxicity screening with human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), a mechanism-based toxicodynamic (TD) model for TKIs was constructed. A QSP-PK-TD model was developed by integrating pharmacokinetic (PK) and quantitative systems pharmacology (QSP) models. This model incorporates critical drug exposure factors, such as plasma protein binding, tissue-plasma partitioning, and drug distribution heterogeneity to enhance extrapolation accuracy. Results: The QSP-PK-TD model validated the reliability of IVIVE and identified the area under the curve of drug effects on mitochondrial membrane potential (AEMMP) and cardiomyocyte contractility (AEAAC) as key in vitro parameters for assessing TKI-induced cardiotoxicity. Incorporating critical drug exposure factors obviously improved qualitative and quantitative extrapolation accuracy. Conclusions: This study established a framework for predicting in vivo cardiotoxicity from in vitro parameters, enabling efficient translation of preclinical data into clinical risk assessment. These findings provide valuable insights for drug development and regulatory decision-making, offering a powerful tool for evaluating TKI-induced cardiotoxicity.
Collapse
Affiliation(s)
- Zhijie Wan
- State Key Laboratory of Natural Medicine, Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Chenyu Wang
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Shizheng Luo
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Jinwei Zhu
- State Key Laboratory of Natural Medicine, Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Hua He
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Kun Hao
- State Key Laboratory of Natural Medicine, Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
23
|
Mattioli AV, Bucciarelli V, Gallina S. The role of physical exercise in cancer therapy-related CV toxicity. Future Cardiol 2025; 21:131-134. [PMID: 39779253 PMCID: PMC11875471 DOI: 10.1080/14796678.2025.2451530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/07/2025] [Indexed: 01/11/2025] Open
Affiliation(s)
- Anna Vittoria Mattioli
- Department of Quality of Life Sciences, University of Bologna-Alma Mater Studiorum, Bologna, Italy
| | - Valentina Bucciarelli
- Cardiovascular Sciences Department, Azienda Ospedaliero-Universitaria delle Marche, Ancona, Italy
| | - Sabina Gallina
- Department of Neuroscience, Imaging and Clinical Sciences, “G. d’Annunzio” University of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
24
|
Travers S, Alexandre J, Baldassarre LA, Salem JE, Mirabel M. Diagnosis of cancer therapy-related cardiovascular toxicities: A multimodality integrative approach and future developments. Arch Cardiovasc Dis 2025; 118:185-198. [PMID: 39947997 DOI: 10.1016/j.acvd.2024.12.012] [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: 11/07/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 03/14/2025]
Abstract
Diagnosing cancer therapy-related cardiovascular toxicities may be a challenge. The interplay between cancer and cardiovascular diseases, beyond shared cardiovascular and cancer risk factors, and the increasingly convoluted cancer therapy schemes have complicated cardio-oncology. Biomarkers used in cardio-oncology include serum, imaging and rhythm modalities to ensure proper diagnosis and prognostic stratification of cardiovascular toxicities. For now, troponin and natriuretic peptides, multimodal cardiovascular imaging (led by transthoracic echocardiography combined with cardiac magnetic resonance or computed tomography angiography) and electrocardiography (12-lead or Holter monitor) are cornerstones in cardio-oncology. However, the imputability of cancer therapies is sometimes difficult to assess, and more refined biomarkers are currently being studied to increase diagnostic accuracy. Advances reside partly in pathophysiology-based serum biomarkers, improved cardiovascular imaging through new technical developments and remote monitoring for rhythm disorders. A multiparametric omics approach, enhanced by deep-learning techniques, should open a new era for biomarkers in cardio-oncology in the years to come.
Collapse
Affiliation(s)
- Simon Travers
- INSERM UMR-S 1180, Université Paris-Saclay, 91400 Orsay, France; Laboratoire de Biochimie, DMU BioPhyGen, Hôpital Européen Georges Pompidou, AP-HP, 75015 Paris, France.
| | - Joachim Alexandre
- INSERM U1086 ANTICIPE, Biology-Research Building, UNICAEN, Normandie University Group, 14000 Caen, France; Department of Pharmacology, Biology-Research Building, PICARO Cardio-Oncology Programme, Caen-Normandy University Hospital, 14000 Caen, France.
| | - Lauren A Baldassarre
- Cardiovascular Medicine, Yale School of Medicine, 06510 New Haven CT, United States of America.
| | - Joe Elie Salem
- CIC-1901, Department of Pharmacology, Hôpital Pitié-Salpêtrière, AP-HP, Sorbonne Université, INSERM, 75013 Paris, France.
| | - Mariana Mirabel
- Cardiology Department, Institut Mutualiste Montsouris, 75014 Paris, France.
| |
Collapse
|
25
|
Guan T, Monteiro O, Chen D, Luo Z, Chi K, Li Z, Liang Y, Lu Z, Jiang Y, Yang J, Lin W, Yi M, Zhang K, Ou C. Long-term and short-term cardiovascular disease mortality among patients of 21 non-metastatic cancers. J Adv Res 2025; 69:215-224. [PMID: 38537701 PMCID: PMC11954795 DOI: 10.1016/j.jare.2024.03.017] [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: 01/21/2024] [Revised: 03/05/2024] [Accepted: 03/23/2024] [Indexed: 04/04/2024] Open
Abstract
INTRODUCTION Previous studies on cardiovascular disease (CVD) death risk in cancer patients mostly focused on overall cancer, age subgroups and single cancers. OBJECTIVES To assess the CVD death risk in non-metastatic cancer patients at 21 cancer sites. METHODS A total of 1,672,561 non-metastatic cancer patients from Surveillance, Epidemiology, and End Results (SEER) datebase (1975-2018) were included in this population-based study, with a median follow-up of 12·7 years. The risk of CVD deaths was assessed using proportions, competing-risk regression, absolute excess risks (AERs), and standardized mortality ratios (SMRs). RESULTS In patients with localized cancers, the proportion of CVD death and cumulative mortality from CVD in the high-competing risk group (14 of 21 unique cancers) surpassed that of primary neoplasm after cancer diagnosis. The SMRs and AERs of CVD were found higher in patients with non-metastatic cancer than the general US population (SMR 1·96 [95 %CI, 1·95-1·97]-19·85[95 %CI, 16·69-23·44]; AER 5·77-210·48), heart disease (SMR 1·94[95 %CI, 1·93-1·95]-19·25[95 %CI, 15·76-23·29]; AER 4·36-159·10) and cerebrovascular disease (SMR 2·05[95 %CI, 2·02-2·08]-24·71[95 %CI, 16·28-35·96]; AER 1·01-37·44) deaths. In the high-competing risk group, CVD-related SMR in patients with localized stage cancer increased with survival time but followed a reverse-dipper pattern in the low-competing risk group (7 of 21 cancers). The high-competing risk group had higher CVD-related death risks than the low-competing risk group. CONCLUSION The CVD death risk in patients with non-metastatic cancer varied by cancer stage, site and survival time. The risk of CVD mortality is higher in 14 out of 21 localized cancers (high-competing cancers). Targeted strategies for CVD management in non-metastatic cancer patients are needed.
Collapse
Affiliation(s)
- Tianwang Guan
- Cancer Center, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan 523059, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou 510515, China
| | - Olivia Monteiro
- Faculty of Medicine, Medical Sciences Division, Macau University of Science and Technology, Avenida da Harmonia, Praia Park, Coloane, Macao 999078, China
| | - Dongting Chen
- Department of Clinical Medicine, Clinical Medical School, Guangzhou Medical University, Guangzhou 510180, China
| | - Zehao Luo
- Department of Clinical Medicine, Clinical Medical School, Guangzhou Medical University, Guangzhou 510180, China
| | - Kaiyi Chi
- Department of Clinical Medicine, Clinical Medical School, Guangzhou Medical University, Guangzhou 510180, China; Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Zhihao Li
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Yinglan Liang
- Department of Clinical Medicine, Clinical Medical School, Guangzhou Medical University, Guangzhou 510180, China
| | - Zhenxing Lu
- The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan 523059, China
| | - Yanting Jiang
- Department of Clinical Medicine, Clinical Medical School, Guangzhou Medical University, Guangzhou 510180, China
| | - Jinming Yang
- Department of Clinical Medicine, Clinical Medical School, Guangzhou Medical University, Guangzhou 510180, China
| | - Wenrui Lin
- Department of Clinical Medicine, Clinical Medical School, Guangzhou Medical University, Guangzhou 510180, China
| | - Min Yi
- Department of Endocrinology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510180, China
| | - Kang Zhang
- Faculty of Medicine, Medical Sciences Division, Macau University of Science and Technology, Avenida da Harmonia, Praia Park, Coloane, Macao 999078, China; The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan 523059, China.
| | - Caiwen Ou
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou 510515, China; The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan 523059, China.
| |
Collapse
|
26
|
Zhang D, Xiong X, Ding H, He X, Li H, Yao Y, Ma R, Liu T. Effectiveness of exercise-based interventions in preventing cancer therapy-related cardiac dysfunction in patients with breast cancer: A systematic review and network meta-analysis. Int J Nurs Stud 2025; 163:104997. [PMID: 39961652 DOI: 10.1016/j.ijnurstu.2025.104997] [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: 09/05/2024] [Revised: 12/07/2024] [Accepted: 01/06/2025] [Indexed: 04/23/2025]
Abstract
BACKGROUND Despite advances in cancer treatment that have improved survival rates among patients with breast cancer, they are at high risk of developing cancer therapy-related cardiac dysfunction, which typically manifests as heart failure. Although exercise improves cardiorespiratory fitness in these patients, its effectiveness in preventing cancer therapy-related cardiac dysfunction remains unclear. OBJECTIVE To assess the effectiveness of exercise-based interventions using cardiac function parameters and to identify the optimal exercise modality for preventing cancer therapy-related cardiac dysfunction. DESIGN A systematic review and network meta-analysis. METHODS A comprehensive search was conducted across PubMed, Embase, Scopus, Web of Science, and the Cochrane Library databases, covering all records from their inception through August 6, 2024. Studies that used exercise-based interventions, either exercise alone or with other interventions, were included. Those with insufficient data for the primary and secondary outcomes were excluded. Quality appraisal was evaluated using the risk of bias tool (RoB-2). All statistical analyses were conducted using the meta (version 7.0-0) and gemtc (version 1.0-2) packages in R software (version 4.3.3). For continuous outcomes, pairwise and network meta-analysis were employed to estimate mean differences (MDs) and 95 % confidence intervals (CI). The Surface Under the Cumulative Ranking Curve (SUCRA) was employed to rank treatments. The study protocol has been registered on PROSPERO (ID: CRD42024501160). RESULTS In total, 13 randomized controlled trials involving 1122 participants were included in the review. There was low-to-high risk of bias across thirteen studies. Compared to usual care, exercise-based interventions significantly improved left ventricular ejection fraction (MD, 1.68; 95 % CI, 0.59-2.77) and global longitudinal strain (MD, 1.40; 95 % CI, 0.59-2.21). Based on the ranking probabilities, combined aerobic and resistance exercise was the most efficacious method for improving left ventricular ejection fraction (four studies; SUCRA, 0.96), followed by exercise-based cardio-oncology rehabilitation (two studies; SUCRA, 0.45) and aerobic exercise (four studies; SUCRA, 0.42). In terms of improving global longitudinal strain, combined aerobic and resistance exercise also ranked highest (three studies; SUCRA, 0.88). However, exercise-based cardio-oncology rehabilitation (two studies; SUCRA, 0.47) and aerobic exercise (one study; SUCRA, 0.45) were less effective. CONCLUSIONS This network meta-analysis showed very low certainty for the prospective efficacy of exercise-based interventions, especially the combined aerobic and resistance exercise, in preventing cancer therapy-related cardiac dysfunction. Further rigorous studies are required to confirm the effectiveness of exercise-based interventions in preventing cancer therapy-related cardiac dysfunction among patients with breast cancer.
Collapse
Affiliation(s)
- Dandan Zhang
- Sun Yat-sen University, School of Nursing, Guangzhou, Guangdong, China
| | - Xingyu Xiong
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hexiao Ding
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Xiaole He
- Nursing Department, Huizhou Central People's Hospital, Huizhou, Guangdong, China
| | - Huan Li
- Department of Thyroid and Breast Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuzhi Yao
- Department of Thyroid and Breast Surgery Center, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong, China
| | - Ruisi Ma
- School of Physical Education, Jinan University, Guangzhou, Guangdong, China
| | - Ting Liu
- Sun Yat-sen University, School of Nursing, Guangzhou, Guangdong, China.
| |
Collapse
|
27
|
Bello MO, Wadid M, Malode A, Patel V, Shah A, Vyas A, Ahmad HA, Tarun T, Dani S, Ahmad J, Zarwan C, Ganatra S. Atrial Fibrillation in Patients with Breast Cancer: A Literature Review. Cardiol Ther 2025; 14:1-15. [PMID: 39714744 PMCID: PMC11893935 DOI: 10.1007/s40119-024-00394-1] [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: 09/13/2024] [Accepted: 12/04/2024] [Indexed: 12/24/2024] Open
Abstract
In addition to traditional risk factors, patients with breast cancer are at an increased risk of atrial fibrillation due to cancer itself and certain cancer therapies. Atrial fibrillation in these patients adds to their morbidity and mortality. The precise mechanisms leading to the increased atrial fibrillation in patients with breast cancer are not well understood. The main goal of atrial fibrillation management in this population is to facilitate uninterrupted cancer treatment while addressing the arrhythmia and other cardiovascular sequelae of cancer treatment. Rhythm control is often challenging to implement in patients with breast cancer during active antineoplastic therapy because of the need for uninterrupted anticoagulation, potential drug-drug interactions between cancer treatments and antiarrhythmic medications, and the increased likelihood of atrial fibrillation recurrence. Prevention of thromboembolism and anticoagulation can also be challenging in patients with breast cancer as a result of the increased risk of cancer-related procoagulant state and coagulopathies. The integration of a cardio-oncology team and a multidisciplinary approach are crucial for better outcomes. The therapeutic interventions should be tailored toward individual patients' profiles through a shared decision-making approach. The precise mechanisms leading to the increased atrial fibrillation in patients with breast cancer are not well understood, highlighting the gaps in our knowledge. More research is required to reduce these gaps, refine risk stratification, and optimize treatment strategies in these patients.
Collapse
Affiliation(s)
- Mozidat Olamide Bello
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA, 01805, USA
| | - Mark Wadid
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA, 01805, USA
| | - Aishwarya Malode
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA, 01805, USA
| | - Vahin Patel
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA, 01805, USA
| | - Anuj Shah
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA, 01805, USA
| | - Ankit Vyas
- Department of Vascular Medicine, Ochsner Clinic Foundation, New Orleans, LA, USA
| | | | - Tushar Tarun
- Division of Cardiovascular Medicine, Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Sourbha Dani
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA, 01805, USA
| | - Javaria Ahmad
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA, 01805, USA
| | - Corrine Zarwan
- Division of Hematology/Oncology, Department of Medicine, Lahey Hospital and Medical Center, Burlington, MA, USA
| | - Sarju Ganatra
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA, 01805, USA.
| |
Collapse
|
28
|
Walls GM, Bergom C, Mitchell JD, Rentschler SL, Hugo GD, Samson PP, Robinson CG. Cardiotoxicity following thoracic radiotherapy for lung cancer. Br J Cancer 2025; 132:311-325. [PMID: 39506136 PMCID: PMC11833127 DOI: 10.1038/s41416-024-02888-0] [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: 05/26/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 11/08/2024] Open
Abstract
Radiotherapy is the standard of care treatment for unresectable NSCLC, combined with concurrent chemotherapy and adjuvant immunotherapy. Despite technological advances in radiotherapy planning and delivery, the risk of damage to surrounding thoracic tissues remains high. Cardiac problems, including arrhythmia, heart failure and ischaemic events, occur in 20% of patients with lung cancer who undergo radiotherapy. As survival rates improve incrementally for this cohort, minimising the cardiovascular morbidity of RT is increasingly important. Problematically, the reporting of cardiac endpoints has been poor in thoracic radiotherapy clinical trials, and retrospective studies have been limited by the lack of standardisation of nomenclature and endpoints. How baseline cardiovascular profile and cardiac substructure radiation dose distribution impact the risk of cardiotoxicity is incompletely understood. As Thoracic Oncology departments seek to expand the indications for radiotherapy, and as the patient cohort becomes older and more comorbid, there is a pressing need for cardiotoxicity to be comprehensively characterised with sophisticated oncology, physics and cardio-oncology evaluations. This review synthesises the evidence base for cardiotoxicity in conventional radiotherapy, focusing on lung cancer, including current data, unmet clinical needs, and future scientific directions.
Collapse
Affiliation(s)
- Gerard M Walls
- Department of Radiation Oncology, Washington University in St Louis, Saint Louis, MO, USA.
- Patrick Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland, USA.
| | - Carmen Bergom
- Department of Radiation Oncology, Washington University in St Louis, Saint Louis, MO, USA
- Siteman Cancer Center, Washington University Medical Campus, Saint Louis, MO, USA
| | - Joshua D Mitchell
- Cardio-Oncology Center of Excellence, Washington University in St Louis, St Louis, MO, USA
| | - Stacey L Rentschler
- Department of Developmental Biology, Washington University in St Louis, St. Louis, MO, USA
- Center for Cardiovascular Research, Department of Medicine, Cardiovascular Division, Washington University in St Louis, St. Louis, MO, USA
| | - Geoffrey D Hugo
- Department of Radiation Oncology, Washington University in St Louis, Saint Louis, MO, USA
- Siteman Cancer Center, Washington University Medical Campus, Saint Louis, MO, USA
| | - Pamela P Samson
- Department of Radiation Oncology, Washington University in St Louis, Saint Louis, MO, USA
- Siteman Cancer Center, Washington University Medical Campus, Saint Louis, MO, USA
| | - Clifford G Robinson
- Department of Radiation Oncology, Washington University in St Louis, Saint Louis, MO, USA
- Siteman Cancer Center, Washington University Medical Campus, Saint Louis, MO, USA
| |
Collapse
|
29
|
Xiong W, Li B, Pan J, Li D, Yuan H, Wan X, Zhang Y, Fu L, Zhang J, Lei M, Chang ACY. Mitochondrial Amount Determines Doxorubicin-Induced Cardiotoxicity in Cardiomyocytes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412017. [PMID: 39921259 PMCID: PMC11948046 DOI: 10.1002/advs.202412017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/30/2024] [Indexed: 02/10/2025]
Abstract
Doxorubicin, an anthracycline commonly used for treating cancer patients, is known for its cardiotoxic side-effects. Although dose-dependent, but susceptibility remains variable among patients, and childhood-exposure-adult-onset remains challenging. Besides topoisomerase toxicity, Doxorubicin is also toxic to the mitochondria yet the underlying late onset mechanism remains elusive. Here, it is observed that the mitochondrial copy number in PBMCs of patients treated with anthracycline chemotherapy is negatively correlated with the change in plasma BNP levels after treatment. Isogenic hiPSC-CMs are generated with high, norm, and low mitochondrial copy numbers using mitochondrial transplantation and the YFP-Parkin system. Remarkably, lower mitochondria copy number translates to lower IC50, suggesting increased susceptibility. Mitochondria supplementation by intramyocardial injection prevents doxorubicin induced heart failure. Mechanistically, doxorubicin treatment leads to mPTP opening and mitochondrial DNA (mtDNA) leakage. This mtDNA leakage event activates the cGAS-STING pathway and drives inflammation and myocardial senescence. Cardiomyocyte-specific knockout of Sting (Myh6-Cre/Stingflox/flox; StingCKO) and over expression of mitochondrial tagged DNase1 in mice partially rescue doxorubicin-induced cardiac dysfunction. In conclusion, the work establishes a negative correlation between cardiomyocyte mitochondrial copy number and doxorubicin toxicity. Molecularly, it is demonstrated that mtDNA leakage activates cGAS-STING pathway and accelerates myocardial dysfunction. These insights offer new co-administration strategies for cancer patients.
Collapse
Affiliation(s)
- Weiyao Xiong
- Department of CardiologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
- Shanghai Institute of Precision MedicineShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200125China
| | - Bin Li
- Department of CardiologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
- Shanghai Institute of Precision MedicineShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200125China
| | - Jianan Pan
- Department of CardiologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
| | - Dongjiu Li
- Department of CardiologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
| | - Haihua Yuan
- Department of OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
| | - Xin Wan
- Department of CardiologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
- Shanghai Institute of Precision MedicineShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200125China
| | - Yanjie Zhang
- Department of OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
| | - Lijun Fu
- Department of CardiologyShanghai Children's Medical CentreShanghai Jiao Tong University School of MedicineShanghai200127China
| | - Junfeng Zhang
- Department of CardiologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
| | - Ming Lei
- Shanghai Institute of Precision MedicineShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200125China
| | - Alex Chia Yu Chang
- Department of CardiologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
- Shanghai Institute of Precision MedicineShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200125China
| |
Collapse
|
30
|
Adams SC, Rivera-Theurel F, Scott JM, Nadler MB, Foulkes S, Leong D, Nilsen T, Porter C, Haykowsky M, Abdel-Qadir H, Hull SC, Iyengar NM, Dieli-Conwright CM, Dent SF, Howden EJ. Cardio-oncology rehabilitation and exercise: evidence, priorities, and research standards from the ICOS-CORE working group. Eur Heart J 2025:ehaf100. [PMID: 40036781 DOI: 10.1093/eurheartj/ehaf100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/06/2025] Open
Abstract
The aim of this whitepaper is to review the current state of the literature on the effects of cardio-oncology rehabilitation and exercise (CORE) programmes and provide a roadmap for improving the evidence-based to support the implementation of CORE. There is an urgent need to reinforce and extend the evidence informing the cardiovascular care of cancer survivors. CORE is an attractive model that is potentially scalable to improve the cardiovascular health of cancer survivors as it leverages many of the existing frameworks developed through decades of delivery of cardiac rehabilitation. However, there are several challenges within this burgeoning field, including limited evidence of the efficacy of this approach in patients with cancer. In this paper, a multidisciplinary team of international experts highlights priorities for future research in this field and recommends standards for the conduct of research.
Collapse
Affiliation(s)
| | - Fernando Rivera-Theurel
- Ted Rogers Cardiotoxicity Prevention Program, University Health Network, Toronto, ON, Canada
- Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Cardiovascular Prevention and Rehabilitation Program, Toronto Rehabilitation Institute, University Health Network, Toronto, ON, Canada
| | - Jessica M Scott
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michelle B Nadler
- Division of Medical Oncology and Hematology, Department of Medicine, University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada
| | - Stephen Foulkes
- Faculty of Nursing, College of Health Sciences, University of Alberta, Edmonton, AB, Canada
| | - Darryl Leong
- The Population Health Research Institute and Department of Medicine, McMaster University and Hamilton Health Sciences, Hamilton, ON, Canada
| | - Tormod Nilsen
- Department of Physical Performance, Norwegian School of Sports Sciences, Oslo, Norway
| | - Charles Porter
- Department of Cardiovascular Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Mark Haykowsky
- Faculty of Nursing, College of Health Sciences, University of Alberta, Edmonton, AB, Canada
| | - Husam Abdel-Qadir
- Ted Rogers Cardiotoxicity Prevention Program, University Health Network, Toronto, ON, Canada
- Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Women's College Hospital and Peter Munk Cardiac Centre, Toronto, ON, Canada
| | - Sarah C Hull
- Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT, USA
- Program for Biomedical Ethics, Yale School of Medicine, New Haven, CT, USA
| | - Neil M Iyengar
- Breast Medicine Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Christina M Dieli-Conwright
- Division of Population Sciences, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Harvard University, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Susan F Dent
- Wilmot Cancer Institute, Department of Medicine, University of Rochester, Rochester, NY, USA
| | - Erin J Howden
- Cardiometabolic Health and Exercise Physiology Laboratory, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| |
Collapse
|
31
|
Lin R, Tian A, Wang Y, Wang X, Yuan X, Yu J, Li G, Xie W. The predictive value of changes in 18F-FDG PET/CT cardiac uptake patterns and metabolic parameters for anthracycline based chemotherapy induced cardiac toxicity in lymphoma patients. PLoS One 2025; 20:e0319442. [PMID: 40014616 DOI: 10.1371/journal.pone.0319442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 01/31/2025] [Indexed: 03/01/2025] Open
Abstract
OBJECTIVE This study aims to examine alterations in positron emission tomography with 2-deoxy-2-[fluorine-18]fluoro-D-glucose integrated with computed tomography (18F-FDG PET/CT) heart uptake patterns and metabolic factors before and after anthracycline-based chemotherapy in lymphoma patients, and to investigate the added benefit of oncological 18F-FDG PET/CT in chemotherapy-induced heart damage. MATERIALS AND METHODS Between July 2017 and December 2022, lymphoma patients diagnosed at the Second Affiliated Hospital of Dalian Medical University who underwent 6 cycles of anthracycline-based chemotherapy and had baseline and 6-cycle oncological 18F-FDG PET/CT scans were included. A total of 366 patients with complete data sets were enrolled. Relevant parameters including blood tests, lipid profile, cardiac biomarkers, lactate dehydrogenase (LDH), erythrocyte sedimentation rate (ESR), albumin (ALB), β2-microglobulin (β2-MG), and cardiac ultrasound findings were collected. Patients were monitored from the initiation of chemotherapy until January 2024, and the occurrence of cancer therapy-related cardiovascular toxicity (CTR-CVT) was documented. Changes in PET/CT heart uptake patterns pre- and post-treatment, along with the presence or absence of CTR-CVT, were used to analyze alterations in left ventricular and epicardial adipose tissue metabolic parameters, as well as changes in echocardiographic parameters. Logistic regression analysis was employed to identify risk factors for CTR-CVT. RESULTS Among lymphoma patients who received 6 cycles of anthracycline-based chemotherapy, compared to their initial state, there was a notable decrease in white blood cell count (WBC), neutrophil-to-lymphocyte ratio (NLR), erythrocyte sedimentation rate (ESR), and β2-microglobulin (β2-MG) levels post-treatment. Conversely, albumin (ALB) levels and blood lipid levels significantly rose after treatment. Post-treatment, the maximum standardized uptake value (SUVmax) and mean standardized uptake value (SUVmean) of the left ventricle significantly increased, and the percentage of patients exhibiting no uptake pattern in the left ventricle significantly decreased, while those with diffuse uptake pattern notably increased. Moreover, the count of patients with abnormal cardiac uptake significantly rose post-treatment. Analyzing changes in uptake patterns, the group displaying abnormal changes exhibited an increase in left atrial diameter and a decrease in left ventricular ejection fraction compared to the group with normal changes. The SUVmax of the epicardial adipose tissue was notably higher in the abnormal change group compared to the normal change group. Based on the presence or absence of CTR-CVT, the CTR-CVT group showcased higher left atrial diameter and left ventricular end-systolic diameter, and lower left ventricular ejection fraction compared to the non-CTR-CVT group. Additionally, the SUVmax and SUVmean of the epicardial adipose tissue were higher in the CTR-CVT group than in the non-CTR-CVT group. Left atrial end-systolic diameter, left ventricular ejection fraction, SUVmax of the epicardial adipose tissue, and change in uptake pattern were identified as risk factors for CTR-CVT. CONCLUSION In lymphoma patients treated with anthracycline-based chemotherapy, alterations in 18F-FDG PET/CT cardiac uptake patterns and metabolic parameters observed during the follow-up period before and after treatment, as well as changes in epicardial adipose tissue metabolic parameters post-treatment, could serve as predictors for the occurrence of CTR-CVT.
Collapse
Affiliation(s)
- Runlong Lin
- Department of Nuclear Medicine, The Second Hospital of Dalian Medical University, Dalian, China
| | - Aijuan Tian
- Department of Nuclear Medicine, The Second Hospital of Dalian Medical University, Dalian, China
| | - Ying Wang
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Xiaomei Wang
- Department of Nuclear Medicine, The Second Hospital of Dalian Medical University, Dalian, China
| | - Xin Yuan
- Department of Nuclear Medicine, The Second Hospital of Dalian Medical University, Dalian, China
| | - Jing Yu
- Department of Nuclear Medicine, The Second Hospital of Dalian Medical University, Dalian, China
| | - Guihua Li
- Department of Health Management Center, The Second Hospital of Dalian Medical University, Dalian, China
| | - Wenli Xie
- Department of Cardiovascular Medicine, The Second Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
32
|
Chong JH, Chuah CTH, Lee CG. Revolutionising Cardio-Oncology Care with Precision Genomics. Int J Mol Sci 2025; 26:2052. [PMID: 40076674 PMCID: PMC11900203 DOI: 10.3390/ijms26052052] [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: 12/11/2024] [Revised: 02/06/2025] [Accepted: 02/11/2025] [Indexed: 03/14/2025] Open
Abstract
Cardiovascular disease is the worldwide leading cause of mortality among survivors of cancer due in part to the cardiotoxicity of anticancer therapies. This paper explores the progress in precision cardio-oncology, particularly in genetic testing and therapeutics, and its impact on cardiovascular diseases in clinical and laboratory settings. These advancements enable clinicians to better assess risk, diagnose conditions, and deliver personalised, cost-effective therapeutics. Through case studies of cancer-therapy-related cardiac dysfunction, clonal haematopoiesis of indeterminate potential, and polygenic risk scoring, we demonstrate the benefits of incorporating precision genomics in individualised care in cardio-oncology. Furthermore, leveraging real-world genomic data in clinical settings can advance our understanding of long noncoding RNAs and microRNAs, which play important regulatory roles in cardio-oncology. Additionally, employing human-induced pluripotent stem cells to stratify risk and guide prevention strategies represents a promising avenue for modelling precision cardio-oncology. While these advancements showcase the significant progress in genetic approaches, they also raise substantial ethical, legal, and societal concerns. Regulatory oversight of genetic and genomic technologies should therefore evolve suitably to keep up with rapid advancements in technology and analysis. Provider education is crucial for the appropriate use of new genetic and genomic applications, including on the existing protection available for patients regarding genetic information. This can provide confidence for diverse study groups to advance genetic studies looking to develop a comprehensive understanding and effective clinical applications for heterogeneous populations. In clinical settings, the implementation of genetic and genomic applications within electronic medical records can offer point-of-care clinical decision support, thus providing timely information to guide clinical management decisions.
Collapse
Affiliation(s)
- Jun Hua Chong
- National Heart Centre Singapore, 5 Hospital Dr, Singapore 169609, Singapore
- Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Charles T. H. Chuah
- Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
- National Cancer Centre Singapore, 30 Hospital Blvd, Singapore 168583, Singapore
- Singapore General Hospital, Outram Road, Singapore 169608, Singapore
| | - Caroline G. Lee
- Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, C/O MD7, Level 2, 8 Medical Drive, Singapore 117597, Singapore
- Division of Cellular & Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, 11 Hospital Crescent, Singapore 169610, Singapore
- NUS Graduate School, National University of Singapore, 21 Lower Kent Ridge Road, Singapore 119077, Singapore
| |
Collapse
|
33
|
Romano LR, Polimeni A, Indolfi C, Curcio A. Management of Cardiac Rhythm Disorders in Cardio-oncology. Arrhythm Electrophysiol Rev 2025; 14:e05. [PMID: 40084345 PMCID: PMC11904418 DOI: 10.15420/aer.2024.20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 08/16/2024] [Indexed: 03/16/2025] Open
Abstract
Arrhythmias and cancer are two pathological conditions that often coexist due to a patient's pre-existing comorbidities, or toxicity linked to anti-neoplastic drugs, and both are often characterised by poor prognosis. Cardio-oncology is a new interdisciplinary field that focuses on the cardiovascular health of cancer patients, especially those undergoing cancer treatment. Furthermore, cardiotoxicity can cause arrhythmias through primary and secondary mechanisms. Chemotherapy drugs have been shown to directly affect molecular pathways associated with arrhythmia development, as well as indirectly through mechanisms involving ischaemia or inflammatory injury to the heart. Understanding how to prevent and to treat these electrophysiological issues in cancer is an important challenge for cardio-oncologists. This review explores the intersection between cardio-oncology and electrophysiology, the various cardiac cell types implicated in the development of arrhythmias during cancer, the interplay between arrhythmias and cancer pathogenesis, and the need for the implantation of electronic devices along with their associated risks.
Collapse
Affiliation(s)
- Letizia Rosa Romano
- Department of Pharmacy, Health and Nutritional Sciences, Università della CalabriaRende, Italy
- Division of Cardiology, Annunziata HospitalCosenza, Italy
| | - Alberto Polimeni
- Department of Pharmacy, Health and Nutritional Sciences, Università della CalabriaRende, Italy
- Division of Cardiology, Annunziata HospitalCosenza, Italy
| | - Ciro Indolfi
- Division of Cardiology, Annunziata HospitalCosenza, Italy
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia UniversityCatanzaro, Italy
| | - Antonio Curcio
- Department of Pharmacy, Health and Nutritional Sciences, Università della CalabriaRende, Italy
- Division of Cardiology, Annunziata HospitalCosenza, Italy
| |
Collapse
|
34
|
Haj-Yehia E, Michel L, Mincu RI, Rassaf T, Totzeck M. Prevention of cancer-therapy related cardiac dysfunction. Curr Heart Fail Rep 2025; 22:9. [PMID: 39969700 PMCID: PMC11839799 DOI: 10.1007/s11897-025-00697-x] [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] [Accepted: 02/10/2025] [Indexed: 02/20/2025]
Abstract
PURPOSE OF REVIEW Introduction of modern cancer therapies has led to increased survival of affected patients. With this advantage, the incidence of cancer therapy-related cardiac dysfunction (CTRCD) has increased and reasonable prevention strategies become necessary. This review outlines the major approaches to limit development and progression of CTRCD. RECENT FINDINGS A broad range of cancer therapies can provoke CTRCD ranging from mild asymptomatic forms to severe heart failure. Profound cardiological assessment of cardiovascular comorbidities before initiation of cancer therapy allows identification of cancer patients at higher risk developing CTRCD which may also require closer surveillance. Cardioprotective adjustment of cancer therapy and initiation of cardioprotective medication and lifestyle optimization prior to anti-cancer treatment additionally limit the risk of CTRCD. During therapy, regular examination of cancer patients using high-sensitive cardiological diagnostic tools as three-dimensional (3D) echocardiography and global longitudinal strain (GLS) enables early detection of mild forms of CTRCD. This allows appropriate adjustment of cancer therapy and initiation of CTRCD treatment to prevent further progression to severe forms. Cardiological risk stratification before treatment initiation, cardioprotective interventions before and during cancer therapy, along with regular monitoring of treated cancer patients, can help prevent the development of CTRCD. This maintains the antitumor effects of cancer therapy while limiting cardiotoxic side effects resulting in improved quality of life and mortality of affected cancer patients.
Collapse
Affiliation(s)
- Elias Haj-Yehia
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center Essen, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Lars Michel
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center Essen, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Raluca I Mincu
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center Essen, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center Essen, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Matthias Totzeck
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center Essen, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany.
| |
Collapse
|
35
|
Farì R, Besutti G, Pattacini P, Ligabue G, Piroli F, Mantovani F, Navazio A, Larocca M, Pinto C, Giorgi Rossi P, Tarantini L. The role of imaging in defining cardiovascular risk to help cancer patient management: a scoping review. Insights Imaging 2025; 16:37. [PMID: 39961941 PMCID: PMC11832977 DOI: 10.1186/s13244-025-01907-9] [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: 09/02/2024] [Accepted: 01/13/2025] [Indexed: 02/20/2025] Open
Abstract
OBJECTIVE This scoping review explores the potential role of cancer-staging chest CT scans in assessing cardiovascular (CV) risk in cancer patients. It aims to evaluate: (1) the correlation between non-gated chest CT and the conventional Agatston score from cardiac CT; (2) the association between coronary calcium scores from non-gated chest CT and CV risk in non-oncological patients; (3) the link between coronary calcium assessed by non-gated chest CT and CV events or endothelial damage in cancer patients. METHODS Three different searches were performed on PubMed, according to the three steps described above. Both original articles and systematic reviews were included. RESULTS Many studies in the literature have found a strong correlation between coronary calcium scores from non-gated chest CTs and the conventional Agatston scores from gated cardiac CTs. Various methodologies, including Agatston scoring, ordinal scoring, and the "extent" and "length" methods, have been successfully adapted for use with non-gated chest CTs. Studies show that non-gated scans, even those using iodinated contrast, can accurately assess coronary calcification and predict CV risk, with correlations as high as r = 0.94 when compared to cardiac CTs. In oncological settings, studies demonstrated a significant link between coronary calcium levels on non-gated chest CTs and higher CV risk, including MACE and overall mortality. CONCLUSIONS Radiological assessment of coronary calcium on non-gated CT scans shows potential for improving CV risk prediction. CRITICAL RELEVANCE STATEMENT Non-gated chest CT scans can detect endothelial damage in cancer patients, highlighting the need for standardized radiological practices to assess CV risks during routine oncological follow-up, thereby enhancing radiology's role in comprehensive cancer care. KEY POINTS Cancer therapies improve outcomes but increase cardiovascular risk, requiring balanced management. Coronary calcification on non-gated CT correlates with Agatston scores, predicting cardiovascular risk. Routinely performed CTs predict cardiovascular risk, optimizing the management of cancer patients.
Collapse
Affiliation(s)
- Roberto Farì
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy.
- Radiology Unit, Department of Diagnostic Imaging and Laboratory Medicine, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy.
| | - Giulia Besutti
- Radiology Unit, Department of Diagnostic Imaging and Laboratory Medicine, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Pierpaolo Pattacini
- Radiology Unit, Department of Diagnostic Imaging and Laboratory Medicine, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Guido Ligabue
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Francesco Piroli
- Cardiology Unit, Department of Specialized Medicine, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Francesca Mantovani
- Cardiology Unit, Department of Specialized Medicine, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Alessandro Navazio
- Cardiology Unit, Department of Specialized Medicine, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Mario Larocca
- Oncology Department, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Carmine Pinto
- Oncology Department, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Paolo Giorgi Rossi
- Epidemiology Unit, Azienda USL-IRCCS di Reggio Emilia, 42123, Reggio Emilia, Italy
| | - Luigi Tarantini
- Cardiology Unit, Department of Specialized Medicine, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| |
Collapse
|
36
|
Zhang SC, Gasho JO, Eno C, Silos KD, Pendergast F, Zhang W, Vail E, Kamrava M, Hakimian B, Mirhadi A, Mak RH, Nikolova AP, Atkins KM. Early cardio-oncology intervention in thoracic radiotherapy: prospective single-arm pilot study. COMMUNICATIONS MEDICINE 2025; 5:43. [PMID: 39962137 PMCID: PMC11833047 DOI: 10.1038/s43856-025-00761-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 02/06/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND While there is increasing recognition of the morbidity of cardiovascular disease in cancer survivors, including accelerated atherosclerosis following thoracic radiotherapy, patients are frequently under-optimized for cardiovascular risk. METHODS In this prospective single-arm cohort pilot study, patients were treated with high-dose thoracic radiotherapy and had early consultation with cardio-oncology. Twenty patients were enrolled. The primary endpoint was adherence to cardio-oncology consultation. Secondary endpoints were cardiovascular medication intervention rate and patient-reported intervention perspectives. Clonal hematopoiesis of indeterminate potential, a major cardiovascular risk marker enriched in patients with cancer and induced by radiation exposure, was measured as an exploratory endpoint. RESULTS The cohort median age is 71 years. Most patients are female (13/20), have primary lung or esophageal carcinoma (16/20), and 7/20 have pre-existing cardiovascular disease. We show that cardio-oncology consultation adherence is high (19/20) and results in cardiovascular medication optimization changes in most patients (12/19), most commonly to initiate or intensify statin therapy (8/12). 8/12 patients with a primary cardiologist prior to enrollment have medication changes recommended. Most (12/17) participants are glad to learn about their heart health during cancer treatment. Clonal hematopoiesis is detectable prior to treatment in 8/20 patients and three develop new variants after treatment (1/3 de novo). CONCLUSIONS We observe that early cardio-oncology consultation is feasible, leads to cardiovascular medication optimization in the majority (>60%) of participants, most commonly to initiate or intensify statin therapy. New clonal hematopoiesis variants are detectable early after radiotherapy and the impact on post-treatment cardiovascular risk is worthy of further study.
Collapse
Affiliation(s)
- Samuel C Zhang
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jordan O Gasho
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Celeste Eno
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Katrina D Silos
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Felicity Pendergast
- Cancer Clinical Trials Office, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Wenjuan Zhang
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Eric Vail
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Mitchell Kamrava
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Behrooz Hakimian
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Amin Mirhadi
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Raymond H Mak
- Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Katelyn M Atkins
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Department of Cardiology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
37
|
Soosaipillai A, Nardi-Agmon I, Brinc D, Fabros A, Kavsak PA, Thavendiranathan P, Di Meo A. Macrotroponin interference and association with cardiotoxicity in patients receiving cardiotoxic breast cancer therapy: a pilot study. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2025; 11:18. [PMID: 39953627 PMCID: PMC11827144 DOI: 10.1186/s40959-025-00314-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 01/29/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND Cancer therapy-related cardiac dysfunction (CTRCD) is an important adverse effect in patients receiving potential cardiotoxic cancer therapies. Interpretation of cardiac troponin results can be affected by presence of macrotroponin, which can complicate CTRCD assessment. We aimed to assess whether macrotroponin is detectable in women with ERBB2 + breast cancer receiving sequential therapy with anthracyclines and trastuzumab. METHODS A total of 20 serum samples from 12 ERBB2 + breast cancer patients (median age: 55 years, range: 30-69 years) who exhibited a significant increase in high-sensitivity cardiac troponin I (hs-cTnI) from baseline to post-anthracycline (~ 2 months after therapy initiation) and/or 3-months into trastuzumab therapy (~ 5 months after therapy initiation) and/or who had at least one hs-cTnI value above the female-specific 99th percentile (hs-cTnI > 16 ng/L) and had available banked blood for analysis were included in this pilot study. Samples were analyzed using the Abbott STAT High-Sensitive Troponin-I and Roche Elecsys Troponin T hs STAT assays. Macrotroponin was detected by treating the sample with protein G and re-measuring hs-cTn. Macrotroponin presence was defined as a hs-cTnI or hs-cTnT recovery of < 40% or 85%, respectively. RESULTS Macrotroponin was not identified after anthracycline treatment but was present in four patients 3-months into trastuzumab therapy, two of which had hs-cTnI concentrations above the 99th percentile. None of these patients exhibited a significant reduction in LVEF and/or GLS despite having significant elevations in hs-cTnI. CONCLUSIONS Clinicians should be cautious of benign hs-cTn elevations resulting from macrotroponin presence, as it can complicate CTRCD assessment.
Collapse
Affiliation(s)
- Andrea Soosaipillai
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Division of Clinical Biochemistry, Laboratory Medicine Program, Toronto General Hospital, University Health Network, 200 Elizabeth Street, Toronto, ON, M5G 2C4, Canada
| | - Inbar Nardi-Agmon
- Department of Medicine, Division of Cardiology, Ted Rogers Program in Cardiotoxicity Prevention, Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, Toronto, ON, Canada
| | - Davor Brinc
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Division of Clinical Biochemistry, Laboratory Medicine Program, Toronto General Hospital, University Health Network, 200 Elizabeth Street, Toronto, ON, M5G 2C4, Canada
| | - Anselmo Fabros
- Division of Clinical Biochemistry, Laboratory Medicine Program, Toronto General Hospital, University Health Network, 200 Elizabeth Street, Toronto, ON, M5G 2C4, Canada
| | - Peter A Kavsak
- Deparment of Pathology and Molecular Medicine, Division of Clinical Pathology, McMaster University, Hamilton, ON, Canada
| | - Paaladinesh Thavendiranathan
- Department of Medicine, Division of Cardiology, Ted Rogers Program in Cardiotoxicity Prevention, Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, Toronto, ON, Canada
- Joint Department of Medical Imaging, Toronto General Hospital, University Health Network, Toronto, ON, Canada
| | - Ashley Di Meo
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.
- Division of Clinical Biochemistry, Laboratory Medicine Program, Toronto General Hospital, University Health Network, 200 Elizabeth Street, Toronto, ON, M5G 2C4, Canada.
| |
Collapse
|
38
|
Man X, Wang H, Chen C, Cong X, Sun L, Sun X, Chen C, Zhang J, Yang L. Efficacy of high-dose steroids versus low-dose steroids in the treatment of immune checkpoint inhibitor-associated myocarditis: a case series and systematic review. Front Immunol 2025; 16:1455347. [PMID: 40013153 PMCID: PMC11860070 DOI: 10.3389/fimmu.2025.1455347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 01/27/2025] [Indexed: 02/28/2025] Open
Abstract
Background Immune checkpoint inhibitor-associated myocarditis (ICI-M) is a rare yet potentially fatal complication of immunotherapy, with no standardized treatment protocol due to limited data. The use of varying steroid doses has resulted in inconsistent outcomes. Methods We retrospectively identified patients diagnosed with ICI-M at our institution between January 2020 and February 2024. Additionally, we conducted a comprehensive literature review using PubMed, Embase, and the Cochrane Library to facilitate a comparative analysis of clinical responses. The primary aim was to compare clinical outcomes and therapeutic responses between patients treated with high-dose versus low-dose methylprednisolone. Results Patients receiving an initial high-dose intravenous methylprednisolone (1 g/day) exhibited a more rapid reduction in myocardial injury markers, including troponin I/T (cTnI/T), creatine kinase (CK), and N-terminal pro b-type natriuretic peptide (NT-proBNP), compared to those receiving lower doses. This group also demonstrated lower incidences of biomarker rebound and maintained lower levels over time. Additionally, the clinical treatment process was more straightforward in the high-dose group, with treatment efficacy surpassing that observed in patients who received an initial methylprednisolone (mPSL) dose of less than 1 g/day. Regarding prognosis, the incidence of major adverse cardiovascular events (MACE) and cardiovascular mortality was significantly lower in the high-dose group compared to the low-dose group. Conclusions In patients with immune checkpoint inhibitor-associated myocarditis, the prompt administration of high-dose corticosteroid pulse therapy (1 g/day) is strongly associated with improved clinical outcomes. This intervention rapidly lowers myocardial injury biomarkers (cTnI/T, CK, NT-proBNP) while minimizing the risk of biomarker rebound, thus optimizing clinical management. Notably, it significantly reduces the incidence of major adverse cardiovascular events (MACE), thereby enhancing patient prognosis. The duration of therapy should be tailored based on clinical response. In cases of steroid resistance, combination therapies may provide additional benefit.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Lei Yang
- Cancer Center, The First Hospital of Jilin University,
Changchun, Jilin, China
| |
Collapse
|
39
|
Dowsing B, Dehbi HM, Chung R, Pedra J, Worn O, Artico J, Schmid P, Roylance R, Kellman P, Moon JC, Crake T, Westwood M, Ghosh A, Andres MS, Nazir MS, Lyon AR, Chen D, Walker M, Manisty CH. HER-SAFE study design: an open-label, randomised controlled trial to investigate the safety of withdrawal of pharmacological treatment for recovered HER2-targeted therapy-related cardiac dysfunction. BMJ Open 2025; 15:e091917. [PMID: 39909533 PMCID: PMC11800297 DOI: 10.1136/bmjopen-2024-091917] [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: 08/01/2024] [Accepted: 01/20/2025] [Indexed: 02/07/2025] Open
Abstract
INTRODUCTION A quarter of breast cancers show human epidermal growth factor-2 (HER2) overexpression, where targeted therapy dramatically improves survival. However, cancer therapy-related cardiac dysfunction (CTRCD) occurs in up to 15% of patients. With the interruption of HER2 therapy, if necessary, and the initiation of heart failure therapy (HFT), HER2 CTRCD recovers in over 80% of cases. The need to continue HFT in 'recovered' HER2 CTRCD following completion of HER2 therapy is unclear and there are potential significant impacts on patient's quality of life (QoL). The Randomised Controlled Trial for the Safety of Withdrawal of Pharmacological Treatment for Recovered HER2 Targeted Therapy Related Cardiac Dysfunction (HER-SAFE) aims to evaluate whether HFT can be safely withdrawn in non-high cardiovascular (CV) risk patients with 'recovered' HER2 CTRCD. METHODS AND ANALYSIS This is a multicentre, open-label randomised controlled trial investigating whether withdrawal of HFT is non-inferior to continuation in non-high CV risk, breast cancer survivors with recovered HER2 CTRCD after cancer treatment completion. The primary endpoint is the incidence of guideline-defined cardiac dysfunction or clinical heart failure. Secondary endpoints include changes in cardiac blood biomarkers, cardiovascular magnetic resonance (CMR)-derived strain and tissue mapping and heart failure symptom questionnaires. The study will recruit 90 participants who will undergo serial clinical assessment over 12 months with advanced cardiovascular imaging (CMR scans with automated analysis at baseline, 6 and 12 months), cardiac biomarker measurement (six time points over 12 months), plus complete heart failure QoL and medication disutility questionnaires. This is the first multicentre study to address this significant clinical issue. ETHICS AND DISSEMINATION This study was approved by the research ethics committee (London-London Bridge, 23/LO/0152). The results will be disseminated in peer-reviewed scientific journals. TRIAL REGISTRATION NUMBER NCT05880160.
Collapse
Affiliation(s)
- Benjamin Dowsing
- Institute of Cardiovascular Science, University College London, London, UK
- Saint Bartholomew's Hospital Barts Heart Centre, London, UK
| | | | - Robin Chung
- Saint Bartholomew's Hospital Barts Heart Centre, London, UK
- Whittington Health NHS Trust, London, UK
| | - Joanna Pedra
- Saint Bartholomew's Hospital Barts Heart Centre, London, UK
| | - Orla Worn
- Saint Bartholomew's Hospital Barts Heart Centre, London, UK
| | - Jessica Artico
- Institute of Cardiovascular Science, University College London, London, UK
- Imperial College Healthcare NHS Trust Cardiology Service, London, UK
| | - Peter Schmid
- Queen Mary University of London, London, UK
- Barts Health NHS Trust, London, UK
| | | | - Peter Kellman
- National Heart Lung and Blood Institute, Bethesda, Maryland, USA
| | - James C Moon
- Institute of Cardiovascular Science, University College London, London, UK
- Saint Bartholomew's Hospital Barts Heart Centre, London, UK
| | - Tom Crake
- Saint Bartholomew's Hospital Barts Heart Centre, London, UK
| | - Mark Westwood
- Saint Bartholomew's Hospital Barts Heart Centre, London, UK
| | - Arjun Ghosh
- Institute of Cardiovascular Science, University College London, London, UK
- Saint Bartholomew's Hospital Barts Heart Centre, London, UK
| | - Maria Sol Andres
- Cardio-Oncology Centre of Excellence, Royal Brompton Hospital, London, UK
| | - Muhummad Sohaib Nazir
- King's College London School of Biomedical Engineering & Imaging Sciences, London, UK
- Cardio-Oncology Centre of Excellence, Royal Brompton and Harefield Hospitals, London, UK
| | - Alexander R Lyon
- Cardio-Oncology Centre of Excellence, Royal Brompton Hospital, London, UK
| | - Daniel Chen
- Institute of Cardiovascular Science, University College London, London, UK
- Saint Bartholomew's Hospital Barts Heart Centre, London, UK
| | - Malcolm Walker
- Institute of Cardiovascular Science, University College London, London, UK
| | - Charlotte H Manisty
- Institute of Cardiovascular Science, University College London, London, UK
- Saint Bartholomew's Hospital Barts Heart Centre, London, UK
| |
Collapse
|
40
|
Tomsitz D, Grabmaier U, Spiro J, Nicolai L, French LE, Massberg S, Heinzerling L. Optimized monitoring for immune checkpoint inhibitor induced myocarditis using high-sensitivity troponin-T. Eur J Cancer 2025; 216:115186. [PMID: 39708617 DOI: 10.1016/j.ejca.2024.115186] [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: 11/11/2024] [Revised: 12/10/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND Immune checkpoint inhibitor (ICI)-induced Myocarditis (irMyocarditis) is a rare adverse event with a high mortality rate of 40-50 % and which is mostly not diagnosed until clinical symptoms emerge. OBJECTIVES This study aims to screen patients for irMyocarditis using high-sensitivity cardiac troponin-T (hs-TnT) before and regularly during therapy with ICI. METHODS A cohort of 280 cancer patients were prospectively screened for levels of hs-TnT at baseline and prior to every ICI infusion. In case of elevation of hs-TnT, patients were referred for further work-up. RESULTS In total, 196 patients exhibited a baseline hs-TnT ≤ 14 ng/l (99th percentile concentration for general population; group A) and 84 patients a hs-TnT > 14 ng/l at baseline (group B). An increase of hs-TnT during ICI-treatment was observed in 56 out of 196 (27.6 %) and 56 out of 84 patients (66.7 %) in group A and B. Cardiovascular assessment was performed in 11.2 % and 27.4 % of patients, respectively, and 4.1 % and 9.5 % of patients were diagnosed with irMyocarditis and treated with corticosteroids. No fatalities occurred in any of the 16 irMyocarditis patients. Defining a threshold with the highest sensitivity and maximum specificity in receiver-operating characteristics curves, identified a limit of 22 ng/l (group A) and 60 ng/l (group B) hs-TnT, associated with a sensitivity of 100 % in both and a specificity of 91.0 % and 89.6 %, respectively, to detect irMyocarditis. CONCLUSION Screening of hs-TnT can identify irMyocarditis early and lead to reduction of MACE and mortality risk through interruption of ICI-treatment and prompt therapy with corticosteroids.
Collapse
Affiliation(s)
- Dirk Tomsitz
- Department of Dermatology and Allergy, University Hospital, LMU Munich, Munich, Germany
| | - Ulrich Grabmaier
- Department of Medicine I, LMU University Hospital, LMU Munich, Munich, Germany
| | - Judith Spiro
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Leo Nicolai
- Department of Medicine I, LMU University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Lars E French
- Department of Dermatology and Allergy, University Hospital, LMU Munich, Munich, Germany; Dr. Philip Frost, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Steffen Massberg
- Department of Medicine I, LMU University Hospital, LMU Munich, Munich, Germany
| | - Lucie Heinzerling
- Department of Dermatology and Allergy, University Hospital, LMU Munich, Munich, Germany; Department of Dermatology, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and University Hospital Erlangen (UKER), Deutsches Zentrum Immuntherapie (DZI) and Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nürnberg (CCC-ER-EMN), Erlangen, Germany; SERIO Registry (www.serio-registry.org), Germany.
| |
Collapse
|
41
|
Todo M, Gatate Y, Nakano S, Kaneko G, Hagiwara M, Takahashi T, Umezawa Y, Ueda G, Ishikawa S, Makino Y, Oyama M, Shirotake S. Early detection of myocarditis caused by immune checkpoint inhibitor therapy with nivolumab and ipilimumab for advanced recurrent renal cell carcinoma. Cancer Immunol Immunother 2025; 74:97. [PMID: 39904795 PMCID: PMC11794898 DOI: 10.1007/s00262-025-03945-0] [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: 09/14/2024] [Accepted: 01/15/2025] [Indexed: 02/06/2025]
Abstract
Combination therapy with nivolumab and ipilimumab for advanced renal cell carcinoma (RCC) may cause immune-related myocarditis; however, its incidence in this cancer type and regimen remains unknown. At our institution, we measure biomarkers, such as high-sensitivity Troponin (hsTn), and perform electrocardiograms (ECGs) and echocardiography before and every month after the initiation of this therapy, and the findings obtained and patients' symptoms are continuously monitored by physicians and pharmacists. A retrospective survey was conducted on physiological and biochemical test findings and immune-related adverse events in patients with advanced RCC who received combination therapy with nivolumab and ipilimumab between October 1, 2018 and December 31, 2023. Patients suspected of having myocarditis consulted with cardiologists. Myocarditis due to this therapy was detected in 5 of the 86 patients (5.8%) assessed using the European Society of Cardiology 2022 guidelines. There were no fatal symptoms or death due to myocarditis. The median time to the onset of myocarditis was 25 days (21-86 days). The early detection of myocarditis caused by this therapy requires the monitoring of changes by periodically measuring hsTn and other cardiac markers and performing ECGs and echocardiography from the early stages of administration through to the end of treatment. In addition to checking symptoms, if these abnormalities are detected and myocarditis is suspected, prompt collaboration with cardiologists is recommended. Our management strategy of care by a onco-cardiology team may contribute to the early diagnosis and treatment of myocarditis.
Collapse
Affiliation(s)
- Maki Todo
- Department of Pharmacy, Saitama Medical University Saitama International Medical Center, 1397-1, Yamane, Hidaka, Saitama, 350-1298, Japan.
| | - Yodo Gatate
- Department of Cardiology, Saitama Medical University Saitama International Medical Center, Saitama, Japan
| | - Shintaro Nakano
- Department of Cardiology, Saitama Medical University Saitama International Medical Center, Saitama, Japan
| | - Go Kaneko
- Department of Uro-Oncology, Saitama Medical University Saitama International Medical Center, Saitama, Japan
| | - Masayuki Hagiwara
- Department of Uro-Oncology, Saitama Medical University Saitama International Medical Center, Saitama, Japan
| | - Takayuki Takahashi
- Department of Uro-Oncology, Saitama Medical University Saitama International Medical Center, Saitama, Japan
| | - Yuta Umezawa
- Department of Uro-Oncology, Saitama Medical University Saitama International Medical Center, Saitama, Japan
| | - Genji Ueda
- Department of Pharmacy, Saitama Medical University Saitama International Medical Center, 1397-1, Yamane, Hidaka, Saitama, 350-1298, Japan
| | - Shiho Ishikawa
- Department of Pharmacy, Saitama Medical University Saitama International Medical Center, 1397-1, Yamane, Hidaka, Saitama, 350-1298, Japan
| | - Yoshinori Makino
- Department of Pharmacy, Saitama Medical University Saitama International Medical Center, 1397-1, Yamane, Hidaka, Saitama, 350-1298, Japan
| | - Masafumi Oyama
- Department of Uro-Oncology, Saitama Medical University Saitama International Medical Center, Saitama, Japan
| | - Suguru Shirotake
- Department of Uro-Oncology, Saitama Medical University Saitama International Medical Center, Saitama, Japan
| |
Collapse
|
42
|
Wang RH, Chen Y, Lou YL, Lu YL, Xu HM. Characteristics, Incidence, and Management of Immune Checkpoint Inhibitors Related Cardiovascular Adverse Events in Real-World Practice-A Retrospective Study in Chinese Han Population. Ther Clin Risk Manag 2025; 21:125-135. [PMID: 39980984 PMCID: PMC11840335 DOI: 10.2147/tcrm.s477417] [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: 05/08/2024] [Accepted: 01/25/2025] [Indexed: 02/22/2025] Open
Abstract
Purpose This study aimed to elaborate on the incidence, clinical features, and management of immune checkpoint inhibitors (ICIs) related cardiovascular adverse events (CVAEs) in real-world practice. Patients and Methods We performed a retrospective chart review study on patients receiving at least one dose of ICI therapy at a Chinese tertiary hospital from March 2020 to March 2021. CVAEs were identified through clinical assessment and the Naranjo algorithm. The management and outcomes of CVAEs were monitored over a median follow-up duration of 8 months. Results Among the included 203 patients, 4.4% (9/203) developed CVAEs, including heart failure (n = 3), arrhythmia (n = 2), myocarditis (n = 2), and pericardial disease (n = 2), with a proportion (6/9) tending to be severe (grade 3 or grade 4). CVAEs were more common in older patients (mean age: 73.6 ± 9.2 years) and those with hypertension (p = 0.02) or heart failure (p = 0.01). Adherence to the American Society of Clinical Oncology (ASCO) guidelines for managing CVAEs was low (44%), with most cases showing partial resolution by the last follow-up. Conclusion We reported that the incidence of ICI-related CVAEs in the Chinese institution was higher than that in some prior studies. Adherence to guidelines for managing ICI-related CVAEs is found to be suboptimal in real-world practice and highlighted as a needed improvement.
Collapse
Affiliation(s)
- Rong-Hua Wang
- Department of Pharmacy, HuZhou Central Hospital (The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University), Huzhou, People’s Republic of China
| | - Yin Chen
- Department of Pharmacy, HuZhou Central Hospital (The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University), Huzhou, People’s Republic of China
| | - Ya-Ling Lou
- Department of Pharmacy, HuZhou Central Hospital (The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University), Huzhou, People’s Republic of China
| | - Yu-Liang Lu
- Department of Cardiology, HuZhou Central Hospital (The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University), Huzhou, People’s Republic of China
| | - Hui-Min Xu
- Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| |
Collapse
|
43
|
Balough E, Ariza A, Asnani A, Hoeger CW. Cardiotoxicity of Anthracyclines. Cardiol Clin 2025; 43:111-127. [PMID: 39551553 DOI: 10.1016/j.ccl.2024.08.002] [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
Anthracycline chemotherapy is associated with cardiotoxicity, predominantly manifesting as left ventricular systolic dysfunction within the first year of treatment. Early detection is possible through biomarkers and cardiovascular imaging before clinical symptoms develop. Comprehensive cardiovascular risk assessment is essential for all patients prior to anthracycline therapy to stratify their risk of cardiotoxicity. Preventive measures, including cardiovascular risk optimization, as well as anthracycline dose adjustments, the use of liposomal anthracyclines, and dexrazoxane in high-risk patients, are crucial to mitigate the risk of cardiotoxicity. Long-term follow-up and cardiovascular risk optimization are critical for cancer survivors to optimize cardiovascular outcomes.
Collapse
Affiliation(s)
- Elizabeth Balough
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, 185 Pilgrim Road, Baker 4, Boston, MA 02215, USA. https://twitter.com/ElizabethBaloug
| | - Abul Ariza
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; 3 Blackfan Circle, CLS-911, Boston, MA 02115, USA
| | - Aarti Asnani
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; 3 Blackfan Circle, CLS-911, Boston, MA 02115, USA. https://twitter.com/AartiAsnaniMD
| | - Christopher W Hoeger
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, 185 Pilgrim Road, Baker 4, Boston, MA 02215, USA.
| |
Collapse
|
44
|
Viñas-Mendieta AE, Gallardo-Grajeda A, López-Fernández T. Cardio-oncology: chances and challenges. Basic Res Cardiol 2025; 120:3-9. [PMID: 39348001 DOI: 10.1007/s00395-024-01080-y] [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: 06/02/2024] [Revised: 09/19/2024] [Accepted: 09/21/2024] [Indexed: 10/01/2024]
Abstract
Cardio-oncology is an emerging field that aims to ensure optimal cancer treatment while minimising cardiovascular toxicity. The management of cardiovascular toxicity is critical because it can lead to premature discontinuation of treatment, increasing the risk of cancer recurrence and mortality. The 2022 European Society of Cardiology guidelines were a milestone in advocating a patient-centred, multidisciplinary approach. Key components include risk stratification and a standardised criterion for adverse events, incorporating definitions from the International Cardio-Oncology Society. Effective risk stratification, supported by imaging and biomarkers, helps to anticipate cardiovascular problems and implement preventive measures. Future research should focus on understanding mechanisms, developing preventive strategies and implementing personalised medicine. Education and reducing disparities in care are essential to advance cardio-oncology and improve patient outcomes.
Collapse
Affiliation(s)
- Adriana E Viñas-Mendieta
- Cardio-Oncology Unit, Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, C/ Paseo de la Castellana nº 261, 28046, Madrid, Spain
- Cardiology Department, Guillermo Almenara Irigoyen Hospital, Lima, Peru
| | - Andrea Gallardo-Grajeda
- Cardio-Oncology Unit, Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, C/ Paseo de la Castellana nº 261, 28046, Madrid, Spain
- Cardiology Department, National Institute of Cardiology Ignacio Chavez, Mexico City, Mexico
| | - Teresa López-Fernández
- Cardio-Oncology Unit, Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, C/ Paseo de la Castellana nº 261, 28046, Madrid, Spain.
- Cardiology Department, Quironsalud Madrid University Hospital, Madrid, Spain.
| |
Collapse
|
45
|
Bloom MW, Ferrari AM, Stojanovska J, Harnden KK, Beckford Y, Skurka K, Barac A. Cardio-Oncology Program Building: A Practical Guide. Cardiol Clin 2025; 43:195-207. [PMID: 39551558 DOI: 10.1016/j.ccl.2024.09.005] [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
The organization of a cardio-oncology clinic and overall program is designed to provide comprehensive cardiovascular care to patients who are at risk of or have developed cardiovascular sequelae during or following cancer treatments. In this article, we summarize the core components of a contemporary cardio-oncology program, including its core members (cardiologists, oncologists, clinical pharmacists, advanced practice providers, nurses, and coordinators), key services (risk assessment, treatment planning, cardiac imaging, intervention, and management), and practical integration within the health care system.
Collapse
Affiliation(s)
| | - Alana M Ferrari
- Division of Hematology/Oncology, University of Virginia Health, Charlottesville, VA, USA
| | - Jadranka Stojanovska
- Department of Radiology, NYU Langone Hospital, NYU Grossman School of Medicine, New York, NY, USA
| | | | - Yaisa Beckford
- Department of Cardiology, Inova Schar Heart and Vascular, Fairfax, VA, USA
| | - Kerry Skurka
- Cardio-Oncology Nurse/Program Consultant, International Cardio-Oncology (IC-OS) Society
| | - Ana Barac
- Department of Cardiology, Inova Schar Heart and Vascular, Fairfax, VA, USA.
| |
Collapse
|
46
|
Bloom MW, Vo JB, Rodgers JE, Ferrari AM, Nohria A, Deswal A, Cheng RK, Kittleson MM, Upshaw JN, Palaskas N, Blaes A, Brown SA, Ky B, Lenihan D, Maurer MS, Fadol A, Skurka K, Cambareri C, Chauhan C, Barac A. Cardio-Oncology and Heart Failure: a Scientific Statement From the Heart Failure Society of America. J Card Fail 2025; 31:415-455. [PMID: 39419165 DOI: 10.1016/j.cardfail.2024.08.045] [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: 04/16/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 10/19/2024]
Abstract
Heart failure and cancer remain 2 of the leading causes of morbidity and mortality, and the 2 disease entities are linked in a complex manner. Patients with cancer are at increased risk of cardiovascular complications related to the cancer therapies. The presence of cardiomyopathy or heart failure in a patient with new cancer diagnosis portends a high risk for adverse oncology and cardiovascular outcomes. With the rapid growth of cancer therapies, many of which interfere with cardiovascular homeostasis, heart failure practitioners need to be familiar with prevention, risk stratification, diagnosis, and management strategies in cardio-oncology. This Heart Failure Society of America statement addresses the complexities of heart failure care among patients with active cancer diagnoses and cancer survivors. Risk stratification, monitoring and management of cardiotoxicity are presented across stages A through D heart failure, with focused discussion on heart failure with preserved ejection fraction and special populations, such as survivors of childhood and young-adulthood cancers. We provide an overview of the shared risk factors between cancer and heart failure, highlighting heart failure as a form of cardiotoxicity associated with many different cancer therapeutics. Finally, we discuss disparities in the care of patients with cancer and cardiac disease and present a framework for a multidisciplinary-team approach and critical collaboration among heart failure, oncology, palliative care, pharmacy, and nursing teams in the management of these complex patients.
Collapse
Affiliation(s)
| | - Jacqueline B Vo
- Radiation Epidemiology Branch, Division of Cancer Epidemiology & Genetics, National Cancer Institute, Bethesda, MD
| | - Jo E Rodgers
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, Chapel Hill, NC
| | - Alana M Ferrari
- Division of Hematology/ Oncology, University of Virginia Health, Charlottesville, VA
| | - Anju Nohria
- Cardio-Oncology Program, Cardiovascular Division, Brigham and Women's Hospital, Boston, MA
| | - Anita Deswal
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Richard K Cheng
- Division of Cardiology, University of Washington, Seattle, WA
| | - Michelle M Kittleson
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | | | - Nicolas Palaskas
- Department of Cardiology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Anne Blaes
- Division of Hematology/Oncology/Transplantation, University of Minnesota, Minneapolis, MN
| | - Sherry-Ann Brown
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI; Research Collaborator, Mayo Clinic, Rochester, MN
| | - Bonnie Ky
- Division of Cardiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; Thalheimer Center for Cardio-Oncology, Abramson Cancer Center and Division of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Daniel Lenihan
- Saint Francis Healthcare, Cape Girardeau, MO and the International Cardio-Oncology Society, Tampa, FL
| | - Mathew S Maurer
- Division of Cardiology, Columbia University Irving Medical Center, New York, NY
| | | | | | - Christine Cambareri
- Clinical Oncology Pharmacist, Hospital of the University of Pennsylvania, Abramson Cancer Center, Philadelphia, PA
| | | | - Ana Barac
- Department of Cardiology, Inova Schar Heart and Vascular, Inova Schar Cancer, Falls Church, VA
| |
Collapse
|
47
|
Sayegh H, Zagouras A, Neal JW, Witteles RM, Zhu H, Waliany S. Classes of Antineoplastic Agents Associated with Increased Risk of Cancer Therapy-associated Hypertension and Management Strategies. Cardiol Clin 2025; 43:31-42. [PMID: 39551560 DOI: 10.1016/j.ccl.2024.08.006] [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
Hypertension (HTN) has been found to be the most common comorbidity in patients with cancer. In addition to increased prevalence of baseline HTN, patients with cancer may be at increased risk of HTN as a short-term or long-term adverse event from cancer therapy. Different classes of cancer therapies have been implicated in the development of HTN, including inhibitors of vascular endothelial growth factor (VEGF), Bruton tyrosine kinase inhibitors, proteasome inhibitors, androgen deprivation therapy, and others. While some of these drugs may lead to increases in blood pressure through on-target effects (eg, with VEGF inhibition), others may be associated with HTN from off-target mechanisms that are not always well understood.
Collapse
Affiliation(s)
- Hoda Sayegh
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Alexia Zagouras
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Joel W Neal
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA; Division of Oncology, Stanford University School of Medicine, Stanford, CA, USA; Stanford Cancer Institute, Stanford, CA, USA
| | - Ronald M Witteles
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA; Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Han Zhu
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA; Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA; Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Sarah Waliany
- Massachusetts General Hospital Cancer Center, Boston, MA, USA; Dana-Farber Cancer Institute, Boston, MA 02114, USA.
| |
Collapse
|
48
|
Panuccio G, Correale P, d'Apolito M, Mutti L, Giannicola R, Pirtoli L, Giordano A, Labate D, Macheda S, Carabetta N, Abdelwahed YS, Landmesser U, Tassone P, Tagliaferri P, De Rosa S, Torella D. Immuno-related cardio-vascular adverse events associated with immuno-oncological treatments: an under-estimated threat for cancer patients. Basic Res Cardiol 2025; 120:153-169. [PMID: 39225869 PMCID: PMC11790807 DOI: 10.1007/s00395-024-01077-7] [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: 05/22/2024] [Revised: 08/16/2024] [Accepted: 08/17/2024] [Indexed: 09/04/2024]
Abstract
Immunotherapy represents an emergent and heterogeneous group of anticancer treatments harnessing the human immune-surveillance system, including immune-checkpoint inhibitor monoclonal antibodies (mAbs), Chimeric Antigen Receptor T Cells (CAR-T) therapy, cancer vaccines and lymphocyte activation gene-3 (LAG-3) therapy. While remarkably effective against several malignancies, these therapies, often in combination with other cancer treatments, have showed unforeseen toxicity, including cardiovascular complications. The occurrence of immuno-mediated adverse (irAEs) events has been progressively reported in the last 10 years. These irAEs present an extended range of severity, from self-limiting to life-threatening conditions. Although recent guidelines in CardioOncology have provided important evidence in managing cancer treatments, they often encompass general approaches. However, a specific focus is required due to the particular etiology, unique risk factors, and associated side effects of immunotherapy. This review aims to deepen the understanding of the prevalence and nature of cardiovascular issues in patients undergoing immunotherapy, offering insights into strategies for risk stratification and management.
Collapse
Affiliation(s)
- Giuseppe Panuccio
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité Berlin, 12200, Berlin, Germany.
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy.
| | - Pierpaolo Correale
- Medical Oncology Unit, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, 89124, Reggio Calabria, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, 19122, USA
| | - Maria d'Apolito
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
- Medical Oncology Unit, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, 89124, Reggio Calabria, Italy
| | - Luciano Mutti
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, 19122, USA
- Department of Applied Sciences and Biotechnology, Università dell'Aquila, L'Aquila, Italy
| | - Rocco Giannicola
- Medical Oncology Unit, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, 89124, Reggio Calabria, Italy
| | - Luigi Pirtoli
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, 19122, USA
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, 19122, USA
- Department of Medical Biotechnology, University of Siena, 53100, Siena, Italy
| | - Demetrio Labate
- Unit of Intensive Care Medicine and Anesthesia, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, 89124, Reggio Calabria, Italy
| | - Sebastiano Macheda
- Unit of Intensive Care Medicine and Anesthesia, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, 89124, Reggio Calabria, Italy
| | - Nicole Carabetta
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Youssef S Abdelwahed
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité Berlin, 12200, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), 10785, Berlin, Germany
| | - Ulf Landmesser
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité Berlin, 12200, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), 10785, Berlin, Germany
- Berlin Institute of Health (BIH), 10178, Berlin, Germany
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, 19122, USA
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Salvatore De Rosa
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy.
| |
Collapse
|
49
|
Cook GJR, Alberts IL, Wagner T, Fischer BM, Nazir MS, Lilburn D. The impact of long axial field of view (LAFOV) PET on oncologic imaging. Eur J Radiol 2025; 183:111873. [PMID: 39647272 PMCID: PMC11904125 DOI: 10.1016/j.ejrad.2024.111873] [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: 08/30/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 12/10/2024]
Abstract
The development of long axial field of view (LAFOV) positron emission tomography coupled with computed tomography (PET/CT) scanners might be considered the biggest step forward in PET imaging since it became a mainstream clinical modality. Despite increased capital and maintenance costs and data storage requirements, the improvement in image quality, significantly faster acquisition times and lower radiopharmaceutical administered activities, allow a high quality and more efficient clinical service. This step change in technology overcomes some of the limitations of standard short axial field of view scanners. It allows simultaneous imaging of all body systems, and with the ability to obtain high temporal resolution data, it increases potential research applications, particularly in multisystem disease or for dosimetry measurements of novel radiopharmaceuticals. The improvements in sensitivity and signal-to-noise facilitates the use of tracers with long half-lives and low administered activity (e.g. [89Zr]-labelled monoclonal antibodies) or very short half-lives (e.g. [82Rb]), opening up applications that hitherto have been challenging. It is early in the evolution of LAFOV PET/CT and the advantages these systems offer have still to be fully realised in providing additional impact in clinical practice. In this article we describe the potential advantages of LAFOV PET technology and some of the clinical and research applications where it has been applied as well as some of the future developments that may enhance the modality further.
Collapse
Affiliation(s)
- Gary J R Cook
- King's College London & Guy's and St Thomas' PET Centre, School of Biomedical Engineering and Imaging Sciences, King's College, London SE1 7EH, UK.
| | - Ian L Alberts
- Molecular Imaging and Therapy, BC Cancer Agency, Vancouver, BC, Canada; Department of Radiology, University of British Columbia, Vancouver, BC, Canada.
| | - Thomas Wagner
- Department of Nuclear Medicine, Royal Free London NHS Trust, London NW3 1TX, UK.
| | - B Malene Fischer
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital-Rigshospitalet, 2100 Copenhagen, Denmark.
| | - Muhummad Sohaib Nazir
- Department of Cardiovascular Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK; Cardio-Oncology Centre of Excellence, Royal Brompton Hospital, Guy's and St Thomas' NHS Foundation Trust, London, UK.
| | - David Lilburn
- King's College London & Guy's and St Thomas' PET Centre, School of Biomedical Engineering and Imaging Sciences, King's College, London SE1 7EH, UK.
| |
Collapse
|
50
|
Malode A, Makwana B, Patel V, Khadke S, Parikh A, Bagga A, Dani S, Ganatra S. Cardiotoxicity and peri-operative considerations in immune checkpoint inhibitor and chimeric antigen receptor T-cell therapy: a narrative review. Anaesthesia 2025; 80 Suppl 2:25-37. [PMID: 39776062 DOI: 10.1111/anae.16493] [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] [Accepted: 10/07/2024] [Indexed: 01/11/2025]
Abstract
INTRODUCTION Immunotherapy has transformed cancer treatment, particularly with immune checkpoint inhibitors and chimeric antigen receptor T-cell therapy. Despite their efficacy, these therapies can induce cardiotoxicity, presenting significant clinical challenges. Immune checkpoint inhibitors can cause myocarditis; pericarditis; arrhythmias; and myocardial infarction through immune-mediated inflammation. Chimeric antigen receptor T-cell therapy may result in cardiovascular complications due to cytokine release syndrome, including myocardial dysfunction, endothelial damage and arrhythmias. METHODS We used PubMed, Embase and Google Scholar to search for peer-reviewed articles in September 2024 without setting any date limits. Our selection criteria encompassed studies focusing on cardiotoxicity related to immune checkpoint inhibitors or chimeric antigen receptor T-cell therapy, comprising original research, meta-analyses, clinical trials and review articles. The findings were reported narratively. RESULTS Early diagnosis of cardiotoxicity is critical but challenging due to non-specific symptoms. Diagnostic tools include ECG; cardiac biomarkers; echocardiography; cardiac magnetic resonance imaging; and endomyocardial biopsy. However, no single tool is definitive. High-dose corticosteroids are the first-line treatment for immune checkpoint inhibitor-induced myocarditis, with additional immunosuppressive therapies for refractory cases. Standard heart failure management protocols should be followed in cases of heart failure. Tocilizumab and corticosteroids are utilised for chimeric antigen receptor T-cell therapy-induced cytokine release syndrome, alongside supportive care, including fluid resuscitation and vasopressors for severe cases. DISCUSSION As the use of immunotherapy expands, understanding the mechanisms, risk factors and management strategies for cardiotoxicity is increasingly important. Collaborative efforts among oncologists, cardiologists and anaesthetists are essential to mitigate these risks, especially in peri-operative settings. Ongoing research is vital to improve the safe and effective use of immunotherapeutic drugs while minimising cardiovascular adverse effects.
Collapse
Affiliation(s)
- Aishwarya Malode
- Department of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Burlington, MA, USA
| | - Bhargav Makwana
- Department of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Burlington, MA, USA
| | - Vahin Patel
- Department of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Burlington, MA, USA
| | - Sumanth Khadke
- Department of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Burlington, MA, USA
| | - Aneri Parikh
- Department of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Burlington, MA, USA
| | - Arindam Bagga
- Department of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Burlington, MA, USA
| | - Sourbha Dani
- Department of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Burlington, MA, USA
| | - Sarju Ganatra
- Department of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Burlington, MA, USA
| |
Collapse
|