51
|
Bonaca MP, Lang NN, Chen A, Amiri-Kordestani L, Lipka L, Zwiewka M, Strnadova C, Klaar S, Dent S, Janicijevic TK, Herrmann J, Barac A, de Boer RA, Deswal A, Schou M, Neilan TG, van der Meer P, Moslehi J, Kondapalli L, Ky B, Fernandez TL, Cornell RF, Flaig TW, Hsia J, Sharon E, de Azambuja E, Seltzer J, Januzzi JL, Petrie MC. Cardiovascular Safety in Oncology Clinical Trials: JACC: CardioOncology Primer. JACC CardioOncol 2025; 7:83-95. [PMID: 39967209 DOI: 10.1016/j.jaccao.2024.09.014] [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: 01/03/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 02/20/2025] Open
Abstract
The development of novel treatments has improved cancer outcomes but may result in cardiovascular toxicities. Traditional approaches to clinical trial safety evaluation have limitations in their ability to detect signals of cardiovascular risk. Mechanisms to increase power and specificity to clarify cardiovascular safety are required. However, implications include increased costs and slower development. The Cardiovascular Safety Research Consortium facilitated stakeholder discussions with representation from academia, industry, and regulators. A think tank was assembled with the aim of providing recommendations for improved collection and reporting of cardiovascular safety signals in oncology trials. Two working groups were formed. The first focuses on incorporation of consensus definitions of cardiovascular disease into the Common Terminology Criteria for Adverse Events used in oncology trial reporting. The second group considers methods for ascertainment and adjudication of cardiovascular events in cancer trials. The overarching aim of this primer is to improve understanding of the potential cardiovascular toxicities of cancer therapies.
Collapse
Affiliation(s)
- Marc P Bonaca
- CPC Clinical Research, Cardiology & Vascular medicine, University of Colorado Anschutz Medical School, Aurora, Colorado, USA.
| | - Ninian N Lang
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom. https://twitter.com/ninianlang
| | - Alice Chen
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland, USA
| | - Laleh Amiri-Kordestani
- Center for Drug Evaluation and Research (CDER), and Oncology Center of Excellence (OCE), U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | | | - Michal Zwiewka
- Paul-Ehrlich-Institut, Bundesinstitut für Impfstoffe und biomedizinische Arzneimittel, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | | | | | - Susan Dent
- Duke Cancer Institute, Duke University, Durham, North Carolina, USA. https://twitter.com/sdent_cardioonc
| | | | - Joerg Herrmann
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA. https://twitter.com/mayocvonc
| | - Ana Barac
- Inova Schar Cancer Institute and Inova Heart and Vascular Institute, Annandale, Virginia, USA. https://twitter.com/AnaBaracCardio
| | - Rudolf A de Boer
- Erasmus MC, Cardiovascular Institute, Thorax Center, Department of Cardiology, Rotterdam, the Netherlands
| | - Anita Deswal
- Department of Cardiology, University of Texas, MD Anderson Cancer Center, Houston, Texas, USA. https://twitter.com/anita_deswal
| | - Morten Schou
- Herlev-Gentofte Hospital, University of Copenhagen, Herlev-Gentofte, Denmark. https://twitter.com/mortschou
| | - Tomas G Neilan
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA. https://twitter.com/TomasNeilan
| | - Peter van der Meer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Javid Moslehi
- Section of Cardio-Oncology & Immunology, Division of Cardiology and the Cardiovascular Research Institute, University of California-San Francisco, San Francisco, California, USA
| | | | - Bonnie Ky
- Perelman School of Medicine the University of Pennsylvania, Thalheimer Center for Cardio-Oncology, Philadelphia, Pennsylvania, USA
| | - Teresa Lopez Fernandez
- Cardiology Department, Institute for Health Research, La Paz University Hospital, Madrid, Spain; Cardiology Department, Quirón Pozuelo University Hospital, Madrid, Spain. https://twitter.com/TeresaLpezFdez1
| | | | - Thomas W Flaig
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Judith Hsia
- CPC Clinical Research, Cardiology & Vascular medicine, University of Colorado Anschutz Medical School, Aurora, Colorado, USA
| | - Elad Sharon
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA. https://twitter.com/EladSharonMD
| | - Evandro de Azambuja
- Institute Jules Bordet and l'Université Libre de Bruxelles (L.U.B), Brussels, Belgium. https://twitter.com/E_de_Azambuja
| | | | - James L Januzzi
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School; Heart Failure and Biomarker Trials, Baim Institute for Clinical Research, Boston, Massachusetts, USA. https://twitter.com/jjheart_doc
| | - Mark C Petrie
- University of Glasgow, Glasgow, United Kingdom. https://twitter.com/markcpetrie20
| |
Collapse
|
52
|
Stein-Merlob AF, Yang EH. Addressing the Growing Intersectionality of Cancer and Heart Disease: The Rise of Cardio-Oncology. Cardiol Clin 2025; 43:xiii-xv. [PMID: 39551566 DOI: 10.1016/j.ccl.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Affiliation(s)
- Ashley F Stein-Merlob
- UCLA Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California at Los Angeles, 650 Charles E Young Drive South, A2-237 CHS, Los Angeles, CA 90095-1679, USA.
| | - Eric H Yang
- UCLA Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California at Los Angeles, 650 Charles E Young Drive South, A2-237 CHS, Los Angeles, CA 90095-1679, USA.
| |
Collapse
|
53
|
Linschoten M, Geels J, van Werkhoven E, Visser-Wisselaar H, Chamuleau MED, Teske AJ, Robbers L, Oerlemans S, Crommelin H, Breems-de Ridder M, Schut A, Asselbergs FW, van Rhenen A. Rationale and design of the HOVON 170 DLBCL-ANTICIPATE trial: preventing anthracycline-induced cardiac dysfunction with dexrazoxane. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2025; 11:8. [PMID: 39875951 PMCID: PMC11773844 DOI: 10.1186/s40959-025-00303-y] [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: 01/09/2025] [Indexed: 01/30/2025]
Abstract
BACKGROUND Dexrazoxane has been studied for its ability to prevent anthracycline-induced cardiac dysfunction (AICD) in several trials but its use in clinical practice remains limited. This is related to the low to moderate quality of the generated evidence, safety concerns and restricted prescribing indications. Additional randomized trials are needed before this drug can be routinely integrated into cardio-oncology clinical practice. OBJECTIVES To describe the rationale and design of the HOVON 170 DLBCL - ANTICIPATE trial. This trial aims to establish the efficacy and safety of dexrazoxane for the primary prevention of AICD in patients diagnosed with Diffuse Large B-Cell Lymphoma (DLBCL) treated with six cycles R-CHOP21 chemo-immunotherapy. METHODS This is a multicenter, parallel-group, open-label, phase III trial, randomizing 324 patients between either no cardioprotective treatment or dexrazoxane from the first R-CHOP cycle. The primary and co-primary endpoints are the incidence of AICD within 12 months of registration and the percentage of patients with complete metabolic remission at the end-of-treatment PET-CT respectively. The trial is registered at the EU Clinical Trials Register (EU-CT number 2023-505377-32) and ClinicalTrials.gov (NCT06220032). RESULTS The medical research ethics committee approved the trial in May 2024. Recruitment has started in September 2024 and is expected to last for three years. CONCLUSIONS This trial is poised to contribute crucial evidence concerning the efficacy and safety on the use of dexrazoxane in the primary prevention of AICD. The trial is anticipated to address critical knowledge gaps and offer important insights into the value of dexrazoxane in cardio-oncology practice.
Collapse
Affiliation(s)
- Marijke Linschoten
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
- Netherlands Heart Institute, Utrecht, The Netherlands.
| | - Jesse Geels
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Erik van Werkhoven
- HOVON Foundation, Rotterdam, The Netherlands
- Department of Hematology, Erasmus Medical Center Cancer Institute, Rotterdam, The Netherlands
| | | | - Martine E D Chamuleau
- Department of Hematology, Amsterdam University Medical Center, Location Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Arco J Teske
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lourens Robbers
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Simone Oerlemans
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, The Netherlands
| | - Heleen Crommelin
- Department of Clinical Pharmacy, Division Laboratories, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Astrid Schut
- The Dutch Network for Cardiovascular Research (WCN), Utrecht, The Netherlands
| | - Folkert W Asselbergs
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Institute of Health Informatics, University College London, London, UK
- Biomedical Research Center, The National Institute for Health Research University College London Hospitals, University College London, London, UK
| | - Anna van Rhenen
- Department of Hematology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
54
|
Itzhaki Ben Zadok O, Simitsis P, Nohria A. Recovery of Left Ventricular Ejection Fraction in Patients With Anthracycline-Induced Cardiomyopathy: A Contemporary Cohort Study. J Card Fail 2025:S1071-9164(25)00009-0. [PMID: 39842707 DOI: 10.1016/j.cardfail.2024.12.012] [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: 08/15/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/24/2025]
Abstract
BACKGROUND Data on left ventricular ejection fraction (LVEF) recovery in patients with anthracycline-induced cardiomyopathy (AIC) are limited. OBJECTIVES To evaluate LVEF recovery rate, its predictors and its association with cardiovascular outcomes in a contemporary and diverse cohort with AIC. METHODS This retrospective study analyzed patients diagnosed with AIC from 2010-2023 at 2 U.S. university hospitals and an affiliated cancer center. LVEF recovery, defined as ≥ 10% improvement in LVEF to a value ≥ 50% within 3 years of AIC detection, was assessed by using Cox proportional-hazards accounting for competing risks. The association between LVEF recovery and the composite of heart failure (HF) hospitalizations, mechanical circulatory support, heart transplantation, or cardiovascular death was assessed by using Cox regression analysis with LVEF recovery as a time-dependent factor. RESULTS Among 167 patients with anthracycline-induced cardiomyopathy (AIC) (median age 67 [Q1, Q3: 53, 74] years, 53% female), the majority had lymphoma (55%) or breast cancer (23%). The median time from first anthracycline exposure to AIC detection was 631 (219, 3569) days, and the median LVEF was 38% (29%, 45%). At the detection of AIC, 69% had symptomatic HF. LVEF recovered in 38% (n = 63) at a median of 349 (137, 691) days from AIC detection. Age ≥ 60 years at anthracycline exposure, non-white race, diabetes mellitus, longer interval between anthracycline exposure and AIC detection, and LV dilation were associated with a lower likelihood of recovery, while statin use and AIC detection after 2022 were associated with a higher likelihood of recovery. LVEF recovery was not associated with cardiovascular outcomes. CONCLUSION In this contemporary and diverse AIC cohort, 38% achieved LVEF recovery. Routine screening for AIC and statin therapy may improve recovery rates.
Collapse
Affiliation(s)
- Osnat Itzhaki Ben Zadok
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA; Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| | - Panagiotis Simitsis
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Anju Nohria
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
55
|
Rivero-Santana B, Saldaña-García J, Caro-Codón J, Zamora P, Moliner P, Martínez Monzonis A, Zatarain E, Álvarez-Ortega C, Gómez-Prieto P, Pernas S, Rodriguez I, Buño Soto A, Cadenas R, Palacios Ozores P, Pérez Ramírez S, Merino Salvador M, Valbuena S, Fernández Gasso L, Juárez V, Severo A, Terol B, de Soto Álvarez T, Rodríguez O, Brion M, González-Costello J, Canales Albendea M, González-Juanatey JR, Moreno R, López-Sendón J, López-Fernández T. Anthracycline-induced cardiovascular toxicity: validation of the Heart Failure Association and International Cardio-Oncology Society risk score. Eur Heart J 2025; 46:273-284. [PMID: 39106857 DOI: 10.1093/eurheartj/ehae496] [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: 02/16/2024] [Revised: 04/24/2024] [Accepted: 07/18/2024] [Indexed: 08/09/2024] Open
Abstract
BACKGROUND AND AIMS Baseline cardiovascular toxicity risk stratification is critical in cardio-oncology. The Heart Failure Association (HFA) and International Cardio-Oncology Society (ICOS) score aims to assess this risk but lacks real-life validation. This study validates the HFA-ICOS score for anthracycline-induced cardiovascular toxicity. METHODS Anthracycline-treated patients in the CARDIOTOX registry (NCT02039622) were stratified by the HFA-ICOS score. The primary endpoint was symptomatic or moderate to severe asymptomatic cancer therapy-related cardiac dysfunction (CTRCD), with all-cause mortality and cardiovascular mortality as secondary endpoints. RESULTS The analysis included 1066 patients (mean age 54 ± 14 years; 81.9% women; 24.5% ≥65 years). According to the HFA-ICOS criteria, 571 patients (53.6%) were classified as low risk, 333 (31.2%) as moderate risk, 152 (14.3%) as high risk, and 10 (0.9%) as very high risk. Median follow-up was 54.8 months (interquartile range 24.6-81.8). A total of 197 patients (18.4%) died, and 718 (67.3%) developed CTRCD (symptomatic: n = 45; moderate to severe asymptomatic: n = 24; and mild asymptomatic: n = 649). Incidence rates of symptomatic or moderate to severe symptomatic CTRCD and all-cause mortality significantly increased with HFA-ICOS score [hazard ratio 28.74, 95% confidence interval (CI) 9.33-88.5; P < .001, and hazard ratio 7.43, 95% CI 3.21-17.2; P < .001) for very high-risk patients. The predictive model demonstrated good calibration (Brier score 0.04, 95% CI 0.03-0.05) and discrimination (area under the curve 0.78, 95% CI 0.70-0.82; Uno's C-statistic 0.78, 95% CI 0.71-0.84) for predicting symptomatic or severe/moderate asymptomatic CTRCD at 12 months. CONCLUSIONS The HFA-ICOS score effectively categorizes patients by cardiovascular toxicity risk and demonstrates strong predictive ability for high-risk anthracycline-related cardiovascular toxicity and all-cause mortality.
Collapse
Affiliation(s)
- Borja Rivero-Santana
- Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, C/Paseo de la Castellana n° 261, 28046 Madrid, Spain
| | - Jesús Saldaña-García
- Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, C/Paseo de la Castellana n° 261, 28046 Madrid, Spain
| | - Juan Caro-Codón
- Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, C/Paseo de la Castellana n° 261, 28046 Madrid, Spain
| | - Pilar Zamora
- Oncology Department, La Paz University Hospital, IdiPAZ Research Institute, CIBER ONC, Madrid, Spain
| | - Pedro Moliner
- Cardiology Department, Bellvitge University Hospital, Bio-Heart Cardiovascular Diseases Research Group, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, CIBER CV, Barcelona, Spain
| | - Amparo Martínez Monzonis
- Cardiology Department, Clinic University Hospital, IDIS Research Institute, CIBERCV, Santiago de Compostela, Spain
| | - Eduardo Zatarain
- Cardiology Department, Hospital General Universitario Gregorio Marañón, CIBER-CV (ISCIII), IISGM, Complutense University, Madrid, Spain
| | - Carlos Álvarez-Ortega
- Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, C/Paseo de la Castellana n° 261, 28046 Madrid, Spain
| | - Pilar Gómez-Prieto
- Hematology Department, La Paz University Hospital, IdiPAZ Research Institute, Madrid, Spain
| | - Sonia Pernas
- Medical Oncology Department, Institut Catala d'Oncologia-H.U.Bellvitge-IDIBELL, L'Hospitalet, Barcelona, Spain
| | - Isabel Rodriguez
- Radiation Oncology Department, La Paz University Hospital, IdiPAZ Research Institute, Madrid, Spain
| | - Antonio Buño Soto
- Department of Laboratory Medicine, La Paz University Hospital, IdiPAZ Research Institute, Madrid, Spain
| | - Rosalía Cadenas
- Cardiology Department, Infanta Sofía University Hospital, European University of Madrid, Madrid, Spain
| | - Patricia Palacios Ozores
- Oncology Department, Oncology Translational Research Group, Clinic University Hospital, IDIS Research Institute, Santiago de Compostela, Spain
| | | | - María Merino Salvador
- Medical Oncology Department, Infanta Sofía University Hospital, Infanta Sofía University Hospital, Henares University Hospital Foundation for Biomedical Research and Innovation (FIIB HUIS HHEN), Madrid, Spain
| | - Silvia Valbuena
- Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, C/Paseo de la Castellana n° 261, 28046 Madrid, Spain
| | - Lucía Fernández Gasso
- Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, C/Paseo de la Castellana n° 261, 28046 Madrid, Spain
| | - Victor Juárez
- Cardiology Department, Hospital Universitario 12 de Octubre, CIBER CV, Madrid, Spain
| | - Andrea Severo
- Cardiology Department, Hospital Universitario 12 de Octubre, CIBER CV, Madrid, Spain
| | - Belén Terol
- Cardiology Department, Hospital Universitario Quironsalud, C. Diego de Velázquez, 1, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - Teresa de Soto Álvarez
- Hematology Department, La Paz University Hospital, IdiPAZ Research Institute, Madrid, Spain
| | - Olaia Rodríguez
- Department of Laboratory Medicine, La Paz University Hospital, IdiPAZ Research Institute, Madrid, Spain
| | - María Brion
- Cardiology Department, Clinic University Hospital, IDIS Research Institute, CIBERCV, Santiago de Compostela, Spain
| | - José González-Costello
- Cardiology Department, Bellvitge University Hospital, Bio-Heart Cardiovascular Diseases Research Group, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, CIBER CV, Barcelona, Spain
| | | | - José R González-Juanatey
- Cardiology Department, Clinic University Hospital, IDIS Research Institute, CIBERCV, Santiago de Compostela, Spain
| | - Raúl Moreno
- Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, C/Paseo de la Castellana n° 261, 28046 Madrid, Spain
| | | | - Teresa López-Fernández
- Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, C/Paseo de la Castellana n° 261, 28046 Madrid, Spain
- Cardiology Department, Hospital Universitario Quironsalud, C. Diego de Velázquez, 1, 28223 Pozuelo de Alarcón, Madrid, Spain
| |
Collapse
|
56
|
Sartorio A, Cristin L, Pont CD, Farzaneh-Far A, Romano S. Global longitudinal strain as an early marker of cardiac damage after cardiotoxic medications, a state of the art review. Prog Cardiovasc Dis 2025:S0033-0620(25)00001-5. [PMID: 39798593 DOI: 10.1016/j.pcad.2025.01.001] [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: 01/07/2025] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
Ejection fraction (EF) is the principal parameter used clinically to assess cardiac function and provides prognostic information. However, significant myocardial damage can be present despite preserved EF. Recently, the measurement of left ventricle (LV) deformation by global longitudinal strain (GLS) has been introduced as a novel early marker of cardiac dysfunction. Cardiotoxicity is a frequent side effect of several drugs most notably those used in the treatment of cancer. Although oncology drugs remain the best known cardiotoxic medications, many other drugs can potentially affect LV function. The early recognition of LV dysfunction due to cardiotoxicity is important and of increasing clinical relevance particularly with the rapid pace of development of new drugs. The aim of our review is to provide an overview of the current literature regarding utility of GLS to assess drug-induced myocardial damage. We propose that GLS is a sensitive early marker of myocardial dysfunction associated with the use of certain medications with high risk of cardiotoxicity. Thus, the use of this technique can potentially alert the clinician to myocardial toxicity before reductions in EF are seen.
Collapse
Affiliation(s)
- Andrea Sartorio
- Department of Internal Medicine, Section of Internal Medicine C, University of Verona, Italy
| | - Luca Cristin
- Department of Internal Medicine, Section of Internal Medicine C, University of Verona, Italy
| | - Chiara Dal Pont
- Department of Internal Medicine, Section of Internal Medicine C, University of Verona, Italy
| | - Afshin Farzaneh-Far
- Division of Cardiology, Department of Medicine, Duke University, Durham, NC, USA
| | - Simone Romano
- Department of Internal Medicine, Section of Internal Medicine C, University of Verona, Italy.
| |
Collapse
|
57
|
Wu H, Huang Y, Yang M, Lu Z, Deng W, Wang Y, Xiao Q, Gu Y. Quantitative assessment of early changes in myocardial extracellular volume during postoperative adjuvant chemotherapy in patients with breast cancer via dual-layer spectral detector computed tomography: a cohort study. Quant Imaging Med Surg 2025; 15:404-416. [PMID: 39839008 PMCID: PMC11744108 DOI: 10.21037/qims-24-777] [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: 04/16/2024] [Accepted: 11/01/2024] [Indexed: 01/23/2025]
Abstract
Background The major of anticancer therapies induce a wide spectrum of cardiotoxic effects. Early identification of anticancer treatment-associated cardiotoxicity is critical to informing decisions on subsequent interventions. Myocardial extracellular volume (ECV) has been proposed as a useful parameter for quantifying the early cardiotoxicity of cancer-related treatment. This study used dual-layer spectral detector computed tomography (CT) technology to simultaneously assess cardiac function and myocardial ECV, characterizing the early changes in parameters during breast cancer therapy. Methods A single-center cohort study was conducted that prospectively enrolled 40 women with breast cancer (mean age 47.5±10.8 years) who underwent postoperative adjuvant chemotherapy between January 12, 2022, and November 2, 2023, with available data from baseline to 3 months after chemotherapy of cardiac computed tomography (CCT), ultrasound cardiography (UCG), electrocardiography, and serum biomarkers. Midventricular and global ECVs of the left ventricle were measured based on an iodine map of the late enhancement phase of dual-layer spectral detector CT. Changes in cardiac function parameters, ECVs, and cardiac biomarkers from baseline to the 3-month follow-up were analyzed. Correlation coefficients between the changes in cardiac function parameters and ECVs were calculated. Results Between baseline and 3 months, there was no significant change in left ventricular ejection fraction (LVEF) on UCG (67.1%±3.8% vs. 66.3%±4.3%, P=0.29) or LVEF on CCT (65.4%±5.9% vs. 64.3%±7.4%, P=0.28). Heart rate increased over 3 months of follow-up (75.2±11.5 vs. 81.7±12.3 bpm; P<0.01). After normalization to body surface area (BSA), cardiac output on CCT/BSA ratio (CCT-CO indexed) (3.5±0.6 vs. 3.8±0.6 L/(min·m2); P=0.01) and left ventricular late (active) filling volume/BSA ratio (LVLFV indexed) (13.5±3.7 vs. 15.8±4.2 mL/m2; P<0.01) significantly increased, while there was a significant decrease at the 3-month follow-up in left ventricular early (passive) filling volume/BSA ratio (LVEFV indexed) (33.3±6.6 vs. 30.6±8.2 mL/m2; P=0.01) and LVEFV/LVLFV ratio (2.7±1.1 vs. 2.1±0.9; P<0.01). Midventricular and global ECVs were elevated at 3 months, significantly so for the midanterior ECV (24.0%±4.5% vs. 25.6%±3.1%; P=0.04), midaverage ECV (25.6%±2.5% vs. 27.0%±2.9%; P=0.01) and global ECV (25.4%±2.4% vs. 27.3%±2.7%; P<0.01). Although changes in ECVs were not associated with changes in LVEFs, global ECV changes were moderately correlated with changes in left ventricular end-diastolic volume/BSA ratio (CCT-LVEDV indexed) (r=0.52; P<0.001), left ventricular stroke volume/BSA ratio (CCT-LVSV indexed) (r=0.56; P<0.001), CCT-CO indexed (r=0.40; P=0.01), and LVEFV indexed (r=0.41; P=0.009). Conclusions CCT-derived ECV was used to evaluate myocardial changes in the early stage of chemotherapy before LVEF significantly decreased. The increases in ECV were not correlated with LVEF. The changes in myocardial ECVs were moderately correlated with cardiac function parameters. ECV may be a useful biomarker for detecting cardiotoxicity in patients with breast cancer in the early stage of anticancer therapy.
Collapse
Affiliation(s)
- Honglin Wu
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan Huang
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Meng Yang
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zhiheng Lu
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Weiwei Deng
- Clinical & Technical Support Department, Philips Healthcare, Shanghai, China
| | - Yu Wang
- Clinical & Technical Support Department, Philips Healthcare, Shanghai, China
| | - Qin Xiao
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yajia Gu
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
58
|
Du YX, Li X, Ji SW, Niu N. Hypertension toxicity of VEGFR-TKIs in cancer treatment: incidence, mechanisms, and management strategies. Arch Toxicol 2025; 99:67-81. [PMID: 39347999 DOI: 10.1007/s00204-024-03874-4] [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: 08/08/2024] [Accepted: 09/19/2024] [Indexed: 10/01/2024]
Abstract
Vascular endothelial growth factor receptor tyrosine kinase inhibitors (VEGFR-TKIs) are a class of targeted anticancer agents that include pazopanib, sunitinib, axitinib, and others. Currently, VEGFR-TKIs are widely used in the clinical treatment of various tumors, which can prolong patients' survival and even cure tumors. However, the use of VEGFR-TKIs is frequently associated with the occurrence of cardiovascular adverse events, with hypertension being the most prevalent. Hypertension and its complications can significantly impact the prognosis of patients, potentially jeopardizing their lives and resulting in the reduction or even cessation of treatment in severe cases. This review addresses the incidence of hypertension due to VEGFR-TKIs, mechanisms of toxicity, management strategies, and future research directions. In addition, hypertension due to VEGFR-TKIs may be associated with salt sensitivity, and possible mechanisms of hypertensive side effects are vasodilator imbalance, decreased capillary density, renal injury, impaired endothelial function due to oxidative stress, decreased lymphatic vascular density, and "off-target effect". A comprehensive understanding of hypertension toxicity due to cancer treatment with VEGFR-TKIs, can enhance clinical practice, thereby improving the prognostic outcomes of VEGFR-TKIs in oncology patients.
Collapse
Affiliation(s)
- Yan-Xi Du
- School of Clinical Medicine, North Sichuan Medical College, Nanchong, 637000, China
| | - Xu Li
- School of Pharmacy, North Sichuan Medical College, Nanchong, 637000, China
| | - Si-Wen Ji
- Office of Academic Affairs, North Sichuan Medical College, Nanchong, 637000, China
| | - Na Niu
- School of Pharmacy, North Sichuan Medical College, Nanchong, 637000, China.
| |
Collapse
|
59
|
Liang Y, Zeng L, Zhou R, Feng M, Liu L, Chen K, Huang J, Liang H, He B, Zhang B, Ying Y, Chen Y, Guan T, Yi M. Cardiovascular Disease and Other Competing Causes of Death in Older Kidney Cancer Patients. Rev Cardiovasc Med 2025; 26:25277. [PMID: 39867178 PMCID: PMC11759974 DOI: 10.31083/rcm25277] [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: 06/17/2024] [Revised: 08/29/2024] [Accepted: 09/05/2024] [Indexed: 01/28/2025] Open
Abstract
Background To study the risk of cardiovascular disease (CVD) and other competing causes of death in older kidney cancer patients. Methods Data on older patients (aged 65 and above) diagnosed with kidney cancer between 1975 and 2018 were extracted from the Surveillance, Epidemiology, and End Results (SEER) database. We delved into the distribution of CVD and other competing causes of death across the entire cohort and in various patient subgroups. The competing risk analysis was used to produce cumulative mortality curves based on cumulative mortality for the primary outcomes by follow-up period. Utilizing the standardized mortality ratios (SMRs) and absolute excess risks (AERs), we contrasted the risk of CVD and other competing causes of death in older kidney cancer patients to that observed in the general population. Results The analysis included 29,349 older kidney cancer patients, of which included 4563 CVD deaths. As survival time extended, the proportion of non-cancer deaths increased in older kidney cancer patients, with CVD accounting for the largest share of non-cancer deaths. At 10-15 years after diagnosis, cumulative non-cancer mortality exceeded primary kidney cancer as the predominant cause of death, and cumulative CVD mortality is higher among all non-cancer causes. Older kidney cancer patients exhibited a greater risk of CVD and other non-cancer deaths than their counterparts in the general older population did (SMR: 1.38-2.81; AER: 1.1-143.69). Conclusions As survival time increases, the risk of non-cancer death in older kidney cancer patients gradually surpassed that of primary cancer, and CVD death accounted for the majority of non-cancer deaths. Among older kidney cancer patients, the risk of CVD mortality was higher than in the general population. Managing non-cancer deaths, especially CVD deaths, should be a focus in the care of older kidney cancer patients.
Collapse
Affiliation(s)
- Yinglan Liang
- Department of Anesthesiology, The Second Clinical College of Guangzhou Medical University, 510180 Guangzhou, Guangdong, China
- Cardiovascular Medicine and Cardio-Oncology Group, Medical Exploration and Translation Team, 510000 Guangzhou, Guangdong, China
| | - Liangjia Zeng
- Cardiovascular Medicine and Cardio-Oncology Group, Medical Exploration and Translation Team, 510000 Guangzhou, Guangdong, China
- Department of Clinical Medicine, The Nanshan Clinical College of Guangzhou Medical University, 510180 Guangzhou, Guangdong, China
| | - Ruoyun Zhou
- Cardiovascular Medicine and Cardio-Oncology Group, Medical Exploration and Translation Team, 510000 Guangzhou, Guangdong, China
- Department of Clinical Medicine, The Third Clinical College of Guangzhou Medical University, 510180 Guangzhou, Guangdong, China
| | - Manting Feng
- Cardiovascular Medicine and Cardio-Oncology Group, Medical Exploration and Translation Team, 510000 Guangzhou, Guangdong, China
- Department of Clinical Medicine, The Second Clinical College of Guangzhou Medical University, 510180 Guangzhou, Guangdong, China
| | - Linglong Liu
- Department of Anesthesiology, The Second Clinical College of Guangzhou Medical University, 510180 Guangzhou, Guangdong, China
- Cardiovascular Medicine and Cardio-Oncology Group, Medical Exploration and Translation Team, 510000 Guangzhou, Guangdong, China
| | - Kexin Chen
- Cardiovascular Medicine and Cardio-Oncology Group, Medical Exploration and Translation Team, 510000 Guangzhou, Guangdong, China
- Department of Clinical Medicine, The Nanshan Clinical College of Guangzhou Medical University, 510180 Guangzhou, Guangdong, China
| | - Jinqi Huang
- Cardiovascular Medicine and Cardio-Oncology Group, Medical Exploration and Translation Team, 510000 Guangzhou, Guangdong, China
- Department of Clinical Medicine, The First Clinical College of Guangzhou Medical University, 510180 Guangzhou, Guangdong, China
| | - Haowen Liang
- Cardiovascular Medicine and Cardio-Oncology Group, Medical Exploration and Translation Team, 510000 Guangzhou, Guangdong, China
- Department of Clinical Medicine, The Third Clinical College of Guangzhou Medical University, 510180 Guangzhou, Guangdong, China
| | - Baixin He
- Cardiovascular Medicine and Cardio-Oncology Group, Medical Exploration and Translation Team, 510000 Guangzhou, Guangdong, China
- Department of Clinical Medicine, The Second Clinical College of Guangzhou Medical University, 510180 Guangzhou, Guangdong, China
| | - Binghua Zhang
- Cardiovascular Medicine and Cardio-Oncology Group, Medical Exploration and Translation Team, 510000 Guangzhou, Guangdong, China
- Department of Clinical Medicine, The Sixth Clinical College of Guangzhou Medical University, 510180 Guangzhou, Guangdong, China
| | - Yican Ying
- Cardiovascular Medicine and Cardio-Oncology Group, Medical Exploration and Translation Team, 510000 Guangzhou, Guangdong, China
- Department of Clinical Medicine, The Second Clinical College of Guangzhou Medical University, 510180 Guangzhou, Guangdong, China
| | - Yuerong Chen
- Minimally Invasive Tumor Therapies Center, Guangdong Second Provincial General Hospital, 510317 Guangzhou, Guangdong, China
| | - Tianwang Guan
- Guangdong Engineering Research Center of Boron Neutron Therapy and Application in Malignant Tumors, Dongguan Key Laboratory of Precision Diagnosis and Treatment for Tumors, Dongguan Engineering Research Center for Innovative Boron Drugs and Novel Radioimmune Drugs, Cancer Center, the 10th Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou 510280, China
| | - Min Yi
- Department of Endocrinology, The Second Affiliated Hospital of Guangzhou Medical University, 510260 Guangzhou, Guangdong, China
| |
Collapse
|
60
|
El-Taji O, Clarke NW. Balancing Efficacy and Cardiotoxicity in Prostate Cancer Therapy: A Call for Precision in Treatment Strategies. Eur Urol 2025; 87:27-28. [PMID: 39674665 DOI: 10.1016/j.eururo.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 10/07/2024] [Indexed: 12/16/2024]
Affiliation(s)
- Omar El-Taji
- The Christie and Salford Royal Hospitals, Manchester, UK
| | - Noel W Clarke
- The Christie and Salford Royal Hospitals, Manchester, UK.
| |
Collapse
|
61
|
Gao S, Zhu H, Chang X, Cao X, Wang Z, Chu X, Zhang L, Wang X, Lu J. Cardiovascular death risk in patients with solid tumors: a population-based study in the United States. Eur J Cancer Prev 2025; 34:11-23. [PMID: 39230031 DOI: 10.1097/cej.0000000000000921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
BACKGROUND Previous studies have focused on the risk of cardiovascular disease (CVD)-specific death in hematological cancers and in some single anatomical tumor sites, there remains a paucity of data on systematic analyses in solid tumors. OBJECTIVE The objective of this study is to evaluate the distribution, risk, and trends of CVD-specific death in patients with solid tumors. METHODS A total of 2 679 293 patients with solid tumors diagnosed between 1975 and 2019 were screened from the Surveillance, Epidemiology and End Results (SEER) program across 15 anatomical sites. Standardized mortality ratios (SMRs) and absolute excess risks (AERs) were used to describe the intensity of CVD-specific death, competing risk regression models were used to assess the risk of CVD-specific death, and restricted cubic spline analyses were employed to investigate the potential linear or nonlinear relationship between age and CVD death. RESULTS CVD-specific death in patients with solid tumors accounted for 48.95% of non-cancer deaths. Compared with the general population, patients with solid tumors had higher SMR and AER of CVD death (SMR: 1.15; AER: 21.12), heart disease-related death (SMR: 1.13; AER: 13.96), and cerebrovascular disease-related death (SMR: 1.20; AER: 4.85). Additionally, the SMR exhibited a decreasing trend with increasing time to diagnosis. Furthermore, a nonlinear relationship was observed between age and CVD-specific death in patients with solid tumors of different systems. CONCLUSION CVD-specific death accounted for a large proportion of the cause of non-cancer deaths. Patients with solid tumors exhibit an elevated risk of CVD-specific death. Screening for CVD death and optimizing risk management in patients with solid tumors throughout anticancer treatment may be beneficial in preventing CVD death.
Collapse
Affiliation(s)
- Shuaijie Gao
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University
| | - Hao Zhu
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University
| | - Xinyu Chang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University
| | - Xiting Cao
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University
| | - Zhenwei Wang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University
| | - Xiaoxuan Chu
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University
| | - Lu Zhang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University
| | - Xinhua Wang
- Oncology Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jie Lu
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University
| |
Collapse
|
62
|
Clasen SC, Miller M. How We Monitor Cardiac Health in Breast Cancer Survivors. J Clin Oncol 2025; 43:4-9. [PMID: 39226483 PMCID: PMC11663137 DOI: 10.1200/jco.24.00757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/05/2024] [Accepted: 07/09/2024] [Indexed: 09/05/2024] Open
Abstract
The Oncology Grand Rounds series is designed to place original reports published in the Journal into clinical context. A case presentation is followed by a description of diagnostic and management challenges, a review of the relevant literature, and a summary of the authors' suggested management approaches. The goal of this series is to help readers better understand how to apply the results of key studies, including those published in Journal of Clinical Oncology, to patients seen in their own clinical practice.
Collapse
Affiliation(s)
- Suparna C. Clasen
- Division of Cardiovascular Medicine, Department of Medicine, Indiana University School of Medicine, 1800 N. Capitol Ave, 71, Indianapolis, IN 46202
| | - Meagan Miller
- Division of Hematology/Oncology, Department of Medicine Indiana University School of Medicine 535 Barnhill Drive, Indianapolis, IN 46202
| |
Collapse
|
63
|
Ayoub C, Appari L, Pereyra M, Farina JM, Chao CJ, Scalia IG, Mahmoud AK, Abbas MT, Baba NA, Jeong J, Lester SJ, Patel BN, Arsanjani R, Banerjee I. Multimodal Fusion Artificial Intelligence Model to Predict Risk for MACE and Myocarditis in Cancer Patients Receiving Immune Checkpoint Inhibitor Therapy. JACC. ADVANCES 2025; 4:101435. [PMID: 39759436 PMCID: PMC11699614 DOI: 10.1016/j.jacadv.2024.101435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 01/07/2025]
Abstract
Background Immune checkpoint inhibitor (ICI) therapy has dramatically improved the prognosis for some cancers but can be associated with myocarditis, adverse cardiovascular events, and mortality. Objectives The aim of this study was to develop an artificial intelligence (AI) model to predict the increased likelihood for the development of ICI-related myocarditis and adverse cardiovascular events. Methods Cancer patients treated with ICI at a tertiary institution from 2011 to 2022 were reviewed. Baseline characteristics, laboratory values, electrocardiograms, and cardiovascular clinical outcomes were extracted. A composite outcome of ICI-related myocarditis and major adverse cardiovascular events (transient ischemic attack/stroke, new diagnosis of heart failure, myocardial infarction, and cardiac death) was used to develop a multimodal joint fusion AI model by combining baseline tabular data with electrocardiogram (ECG) in a single end-to-end model. ECG data were parsed using 1-D convolution and tubular data using multilayer perceptron. Results Of 2,258 cancer patients who had ICI therapy and troponin measurement (mean age 68.5 ± 11.5 years, 59.7% male), the composite of cardiovascular clinical adverse events, including ICI-related myocarditis and major adverse cardiovascular events, occurred in 264 (11.7%) unique patients, with 428 events overall (including 59 [3%] ICI-related myocarditis events and 59 [3%] cardiac deaths). The proposed joint fusion model outperformed individual ECG and baseline electronic medical record data and laboratory value models with an area under the operating characteristics curve of 0.72 (0.64 true positive rate and 0.98 negative predictive value). Conclusion A multimodal fusion AI model to predict myocarditis and adverse cardiovascular events in cancer patients starting ICI therapy had good prognostic performance. It may have clinical utility in identifying at-risk patients who may benefit from closer surveillance.
Collapse
Affiliation(s)
- Chadi Ayoub
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, Arizona, USA
| | - Lalith Appari
- Department of Radiology, Mayo Clinic, Phoenix, Arizona, USA
| | - Milagros Pereyra
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, Arizona, USA
| | - Juan M. Farina
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, Arizona, USA
| | - Chieh-Ju Chao
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Isabel G. Scalia
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, Arizona, USA
| | - Ahmed K. Mahmoud
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, Arizona, USA
| | | | - Nima Ali Baba
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, Arizona, USA
| | - Jiwoong Jeong
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, Arizona, USA
| | - Steven J. Lester
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, Arizona, USA
| | | | - Reza Arsanjani
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, Arizona, USA
| | - Imon Banerjee
- Department of Radiology, Mayo Clinic, Phoenix, Arizona, USA
| |
Collapse
|
64
|
Aziz MK, Molony D, Monlezun D, Holder T, Brunckhorst O, Higgason N, Roland J, Magill R, Fatakdawala M, Iacobucci A, Mody-Bailey N, Owen C, Zarker A, Thames E, Swaby J, Xiao D, Choi L, Desai S, Galan J, Deng B, Hartshorne T, Nichols A, Zhang A, Imber J, Song J, Jones W, Rivas A, Sanchez D, Guhan M, Gandaglia G, Ranganath S, Jacob J, Howell S, Plana J, van den Bergh R, Roberts M, Sommer SG, Oldenburg J, Ploussard G, Tilki D, Schoots I, Briers E, Stranne J, Rouviere O, van Oort I, Oprea-Lager D, De Santis M, Cornford P, Koutroumpakis E, Ziaolhagh A, Ali A, Wamique Yusuf S, Iliescu C, Canfield S. Prostate Cancer Therapy Cardiotoxicity Map (PROXMAP) for Advanced Disease States: A Systematic Review and Network Meta-analysis with Bayesian Modeling of Treatment Histories. Eur Urol 2025; 87:15-26. [PMID: 39299896 DOI: 10.1016/j.eururo.2024.08.031] [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: 05/10/2024] [Revised: 07/22/2024] [Accepted: 08/23/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND AND OBJECTIVE Recommendations of first-line therapies for metastatic hormone-sensitive (mHSPC), nonmetastatic castrate-resistant (M0CRPC), and metastatic castrate-resistant (mCRPC) prostate cancer do not account for cardiotoxicity due to a lack of clear prior evidence. This manuscript assesses cardiotoxicity of these therapies. METHODS We searched Ovid Medline, Elsevier Embase, and the Cochrane Library for randomized clinical trials (RCTs) from database inception to January 14, 2024. Network meta-analyses of first-line mHSPC, M0CRPC, and mCRPC therapies were constructed for the five cardiotoxicity metrics defined by the International Cardio-Oncology Society: heart failure, myocarditis, vascular toxicity, hypertension, and arrhythmias. Additional Bayesian network meta-analyses also accounted for prior treatment history. KEY FINDINGS AND LIMITATIONS Thirteen RCTs (16 292 patients) were included. For mHSPC, androgen deprivation therapy (ADT) plus docetaxel (DTX) plus abiraterone acetate (AA) with prednisone (P) demonstrated a significant increase in hypertension and arrhythmias versus ADT + DTX (risk ratio [RR] 2.85, 95% confidence interval [CI] 1.67-4.89, and RR 2.01, 95% CI 1.17-3.44, respectively); however, no corresponding differences were observed between ADT + DTX plus darolutamide (DAR) and ADT + DTX (RR 1.55, 95% CI 0.73-3.30, and RR 0.94, 95% CI 0.63-1.40, respectively). For mCRPC assuming a history of mHSPC treatment, ADT + AA + P plus olaparib (OLA) demonstrated a statistically significant decrease in hypertension versus ADT + AA + P (RR 0.20, 95% CI 0.16-0.26). M0CRPC results were unremarkable. CONCLUSIONS AND CLINICAL IMPLICATIONS For mHSPC, ADT + DTX + DAR demonstrates less cardiotoxicity than ADT + DTX + AA + P due to a lower risk of hypertension and arrhythmias from decreased mineralocorticoid excess. In addition, OLA counterintuitively offers decreased hypertension when superimposed on ADT + AA + P for mCRPC treatment after prior androgen deprivation from mHSPC therapy.
Collapse
Affiliation(s)
- Moez Karim Aziz
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA; Department of Internal Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Donald Molony
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Dominique Monlezun
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Travis Holder
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Oliver Brunckhorst
- MRC Centre for Transplantation, Guy's Hospital Campus, King's College London, King's Health Partners, London, UK
| | - Noel Higgason
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jerry Roland
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Resa Magill
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Mariya Fatakdawala
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Alexander Iacobucci
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Neal Mody-Bailey
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Chris Owen
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Andrew Zarker
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Emma Thames
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Justin Swaby
- Department of Internal Medicine, University of Georgia, Augusta, GA, USA
| | - Daniel Xiao
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Lily Choi
- Department of Internal Medicine, University of the Incarnate Word, San Antonio, TX, USA
| | - Shubh Desai
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jacob Galan
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Brett Deng
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Taylor Hartshorne
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Alexis Nichols
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Allan Zhang
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jared Imber
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jeffrey Song
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX, USA
| | - William Jones
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Alexis Rivas
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Darren Sanchez
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Maya Guhan
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX, USA
| | | | - Shreyas Ranganath
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jerril Jacob
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Skyler Howell
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Juan Plana
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Roderick van den Bergh
- Prostate Cancer Guidelines Panel, European Association of Urology, Arnhem, The Netherlands
| | - Matthew Roberts
- Prostate Cancer Guidelines Panel, European Association of Urology, Arnhem, The Netherlands
| | - Silke Gillessen Sommer
- Prostate Cancer Guidelines Panel, European Association of Urology, Arnhem, The Netherlands
| | - Jan Oldenburg
- Prostate Cancer Guidelines Panel, European Association of Urology, Arnhem, The Netherlands
| | - Guillaume Ploussard
- Prostate Cancer Guidelines Panel, European Association of Urology, Arnhem, The Netherlands
| | - Derya Tilki
- Prostate Cancer Guidelines Panel, European Association of Urology, Arnhem, The Netherlands
| | - Ivo Schoots
- Prostate Cancer Guidelines Panel, European Association of Urology, Arnhem, The Netherlands
| | - Erik Briers
- Prostate Cancer Guidelines Panel, European Association of Urology, Arnhem, The Netherlands
| | - Johan Stranne
- Prostate Cancer Guidelines Panel, European Association of Urology, Arnhem, The Netherlands
| | - Olivier Rouviere
- Prostate Cancer Guidelines Panel, European Association of Urology, Arnhem, The Netherlands
| | - Inge van Oort
- Prostate Cancer Guidelines Panel, European Association of Urology, Arnhem, The Netherlands
| | - Daniela Oprea-Lager
- Prostate Cancer Guidelines Panel, European Association of Urology, Arnhem, The Netherlands
| | - Maria De Santis
- Prostate Cancer Guidelines Panel, European Association of Urology, Arnhem, The Netherlands
| | - Philip Cornford
- Prostate Cancer Guidelines Panel, European Association of Urology, Arnhem, The Netherlands
| | | | - Ali Ziaolhagh
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Abdelrahman Ali
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Syed Wamique Yusuf
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cezar Iliescu
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Steven Canfield
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
65
|
Bostany G, Chen Y, Francisco L, Dai C, Meng Q, Sparks J, Sessions M, Nabell L, Stringer-Reasor E, Khoury K, Lenneman C, Keene K, Armenian S, Landier W, Bhatia S. Cardiac Dysfunction Among Breast Cancer Survivors: Role of Cardiotoxic Therapy and Cardiovascular Risk Factors. J Clin Oncol 2025; 43:32-45. [PMID: 38833638 DOI: 10.1200/jco.23.01779] [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: 08/16/2023] [Revised: 03/15/2024] [Accepted: 04/04/2024] [Indexed: 06/06/2024] Open
Abstract
PURPOSE Cardiac dysfunction is the leading cause of mortality among 10-year breast cancer survivors. Limited information regarding long-term risks of cardiac dysfunction after cardiotoxic therapy (anthracyclines, trastuzumab/pertuzumab, radiation) has precluded development of surveillance guidelines for the survivors. METHODS Patients with breast cancer who completed cardiotoxic therapy underwent echocardiographic screening every 2 years. New-onset cardiac dysfunction was defined as left ventricular ejection fraction (LVEF) <50% after cardiotoxic therapy initiation and included early- and late-onset cardiac dysfunction. RESULTS We evaluated 2,808 echocardiograms in 829 breast cancer survivors; the median age at breast cancer diagnosis was 54.2 years (range, 20.3-86.3); the median follow-up was 8.6 years (1.8-39.8); 39.7% received anthracyclines, 16% received trastuzumab/pertuzumab, 6.2% received both anthracyclines and trastuzumab/pertuzumab, and 38.1% received radiation alone. The cumulative incidence of cardiac dysfunction increased from 1.8% at 2 years to 15.3% at 15 years from cardiotoxic therapy initiation. Multivariable Cox regression analysis identified the following risk factors: non-Hispanic Black race (hazard ratio [HR], 2.15 [95% CI], 1.37 to 3.38), cardiotoxic therapies (anthracyclines: HR, 2.35 [95% CI, 1.25 to 4.4]; anthracyclines and trastuzumab/pertuzumab: HR, 3.92 [95% CI, 1.74 to 8.85]; reference: left breast radiation alone), selective estrogen receptor modulators (HR, 2.0 [95% CI, 1.2 to 3.33]), and precancer hypertension (HR, 3.16 [95% CI, 1.63 to 6.1]). Late-onset cardiac dysfunction was most prevalent among anthracycline- and radiation-exposed patients; early-onset cardiac dysfunction was most prevalent among patients exposed to anthracyclines and trastuzumab/pertuzumab; equal prevalence of both early- and late-onset cardiac dysfunction was observed in trastuzumab-/pertuzumab-exposed patients. Adjusted longitudinal analyses revealed an annual decline in LVEF by 0.29% (P = .009) over 20 years from breast cancer diagnosis. CONCLUSION These findings provide evidence to support echocardiographic surveillance for several years after cardiotoxic therapy and also suggest a need to examine the efficacy of management of cardiovascular risk factors to mitigate risk.
Collapse
Affiliation(s)
- Geoffrey Bostany
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham (UAB) Heersink School of Medicine (SOM), Birmingham, AL
| | - Yanjun Chen
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham (UAB) Heersink School of Medicine (SOM), Birmingham, AL
| | - Liton Francisco
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham (UAB) Heersink School of Medicine (SOM), Birmingham, AL
| | - Chen Dai
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham (UAB) Heersink School of Medicine (SOM), Birmingham, AL
| | - Qingrui Meng
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham (UAB) Heersink School of Medicine (SOM), Birmingham, AL
| | - Jessica Sparks
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham (UAB) Heersink School of Medicine (SOM), Birmingham, AL
| | - Min Sessions
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham (UAB) Heersink School of Medicine (SOM), Birmingham, AL
| | - Lisle Nabell
- Division of Hematology/Oncology, UAB, Birmingham, AL
| | | | - Katia Khoury
- Division of Hematology/Oncology, UAB, Birmingham, AL
| | | | | | - Saro Armenian
- Department of Pediatric Oncology, City of Hope, Duarte, CA
| | - Wendy Landier
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham (UAB) Heersink School of Medicine (SOM), Birmingham, AL
- Division of Pediatric Hematology/Oncology, UAB, Birmingham, AL
| | - Smita Bhatia
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham (UAB) Heersink School of Medicine (SOM), Birmingham, AL
- Division of Pediatric Hematology/Oncology, UAB, Birmingham, AL
| |
Collapse
|
66
|
Oikonomou EK, Sangha V, Dhingra LS, Aminorroaya A, Coppi A, Krumholz HM, Baldassarre LA, Khera R. Artificial Intelligence-Enhanced Risk Stratification of Cancer Therapeutics-Related Cardiac Dysfunction Using Electrocardiographic Images. Circ Cardiovasc Qual Outcomes 2025; 18:e011504. [PMID: 39221857 PMCID: PMC11745701 DOI: 10.1161/circoutcomes.124.011504] [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: 08/05/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Risk stratification strategies for cancer therapeutics-related cardiac dysfunction (CTRCD) rely on serial monitoring by specialized imaging, limiting their scalability. We aimed to examine an application of artificial intelligence (AI) to ECG images as a surrogate for imaging risk biomarkers and its association with early CTRCD. METHODS Across a US-based health system (2013-2023), we identified 1550 patients (aged, 60 [interquartile range, 51-69] years, 1223 [78.9%] women) without cardiomyopathy who received anthracyclines or trastuzumab for breast cancer or non-Hodgkin lymphoma and had ECG performed ≤12 months before treatment. We deployed a validated AI model of left ventricular systolic dysfunction to baseline ECG images and defined low-, intermediate-, and high-risk groups based on AI-ECG left ventricular systolic dysfunction probabilities of <0.01, 0.01 to 0.1, and ≥0.1 (positive screen), respectively. We explored the association with early CTRCD (new cardiomyopathy, heart failure, or left ventricular ejection fraction <50%), or left ventricular ejection fraction <40%, up to 12 months after treatment. In a mechanistic analysis, we assessed the association between global longitudinal strain and AI-ECG left ventricular systolic dysfunction probabilities in studies performed within 15 days of each other. RESULTS Among 1550 patients without known cardiomyopathy (median follow-up, 14.1 [interquartile range, 13.4-17.1] months), 83 (5.4%), 562 (36.3%), and 905 (58.4%) were classified as high, intermediate, and low risk, respectively, by baseline AI-ECG. A high-risk versus low-risk AI-ECG screen (≥0.1 versus <0.01) was associated with a 3.4-fold and 13.5-fold higher incidence of CTRCD (adjusted hazard ratio, 3.35 [95% CI, 2.25-4.99]) and left ventricular ejection fraction <40% (adjusted hazard ratio, 13.52 [95% CI, 5.06-36.10]), respectively. Post hoc analyses supported longitudinal increases in AI-ECG probabilities within 6 to 12 months of a CTRCD event. Among 1428 temporally linked echocardiograms and ECGs, AI-ECG left ventricular systolic dysfunction probabilities were associated with worse global longitudinal strain (global longitudinal strain, -19% [interquartile range, -21% to -17%] for probabilities <0.1, to -15% [interquartile range, -15% to -9%] for ≥0.5 [P<0.001]). CONCLUSIONS AI applied to baseline ECG images can stratify the risk of early CTRCD associated with anthracycline or trastuzumab exposure in the setting of breast cancer and non-Hodgkin lymphoma therapy.
Collapse
Affiliation(s)
- Evangelos K. Oikonomou
- Section of Cardiovascular Medicine, Department of Internal Medicine (E.K.O., V.S., L.S.D., A.A., H.M.K., L.A.B., R.K.), Yale School of Medicine, New Haven, CT
| | - Veer Sangha
- Section of Cardiovascular Medicine, Department of Internal Medicine (E.K.O., V.S., L.S.D., A.A., H.M.K., L.A.B., R.K.), Yale School of Medicine, New Haven, CT
- Department of Engineering Science, University of Oxford, United Kingdom (V.S.)
| | - Lovedeep S. Dhingra
- Section of Cardiovascular Medicine, Department of Internal Medicine (E.K.O., V.S., L.S.D., A.A., H.M.K., L.A.B., R.K.), Yale School of Medicine, New Haven, CT
| | - Arya Aminorroaya
- Section of Cardiovascular Medicine, Department of Internal Medicine (E.K.O., V.S., L.S.D., A.A., H.M.K., L.A.B., R.K.), Yale School of Medicine, New Haven, CT
| | - Andreas Coppi
- Center for Outcomes Research and Evaluation, Yale-New Haven Hospital, New Haven, CT (A.C., H.M.K., R.K.)
| | - Harlan M. Krumholz
- Section of Cardiovascular Medicine, Department of Internal Medicine (E.K.O., V.S., L.S.D., A.A., H.M.K., L.A.B., R.K.), Yale School of Medicine, New Haven, CT
- Center for Outcomes Research and Evaluation, Yale-New Haven Hospital, New Haven, CT (A.C., H.M.K., R.K.)
| | - Lauren A. Baldassarre
- Section of Cardiovascular Medicine, Department of Internal Medicine (E.K.O., V.S., L.S.D., A.A., H.M.K., L.A.B., R.K.), Yale School of Medicine, New Haven, CT
| | - Rohan Khera
- Section of Cardiovascular Medicine, Department of Internal Medicine (E.K.O., V.S., L.S.D., A.A., H.M.K., L.A.B., R.K.), Yale School of Medicine, New Haven, CT
- Section of Biomedical Informatics and Data Science (R.K.), Yale School of Medicine, New Haven, CT
- Center for Outcomes Research and Evaluation, Yale-New Haven Hospital, New Haven, CT (A.C., H.M.K., R.K.)
- Section of Health Informatics, Department of Biostatistics, Yale School of Public Health, New Haven, CT (R.K.)
| |
Collapse
|
67
|
Tavernese A, Cammalleri V, Mollace R, Antonelli G, Piscione M, Cocco N, Carpenito M, Dominici C, Federici M, Ussia GP. The Role of Advanced Cardiac Imaging in Monitoring Cardiovascular Complications in Patients with Extracardiac Tumors: A Descriptive Review. J Cardiovasc Dev Dis 2024; 12:9. [PMID: 39852287 PMCID: PMC11765722 DOI: 10.3390/jcdd12010009] [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: 10/31/2024] [Revised: 12/09/2024] [Accepted: 12/23/2024] [Indexed: 01/26/2025] Open
Abstract
Cardiac involvement in cancer is increasingly important in the diagnosis and follow-up of patients. A thorough cardiovascular evaluation using multimodal imaging is crucial to assess any direct cardiac involvement from oncological disease progression and to determine the cardiovascular risk of patients undergoing oncological therapies. Early detection of cardiac dysfunction, particularly due to cardiotoxicity from chemotherapy or radiotherapy, is essential to establish the disease's overall prognostic impact. Comprehensive cardiovascular imaging should be integral to the clinical management of cancer patients. Echocardiography remains highly effective for assessing cardiac function, including systolic performance and ventricular filling pressures, with speckle-tracking echocardiography offering early insights into chemotoxicity-related myocardial damage. Cardiac computed tomography (CT) provides precise anatomical detail, especially for cardiac involvement due to metastasis or adjacent mediastinal or lung tumors. Coronary assessment is also important for initial risk stratification and monitoring potential coronary artery disease progression after radiotherapy or chemotherapeutic treatment. Finally, cardiac magnetic resonance (CMR) is the gold standard for myocardial tissue characterization, aiding in the differential diagnosis of cardiac masses. CMR's mapping techniques allow for early detection of myocardial inflammation caused by cardiotoxicity. This review explores the applicability of echocardiography, cardiac CT, and CMR in cancer patients with extracardiac tumors.
Collapse
Affiliation(s)
- Annamaria Tavernese
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Roma, Italy
| | - Valeria Cammalleri
- Operative Research Unit of Emodinamica, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Roma, Italy
| | - Rocco Mollace
- Department of Experimental Medicine, Università degli Studi di Roma Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
- Cardiology Unit, Humanitas Gavazzeni, 24125 Bergamo, Italy
| | - Giorgio Antonelli
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Roma, Italy
| | - Mariagrazia Piscione
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Roma, Italy
| | - Nino Cocco
- Operative Research Unit of Emodinamica, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Roma, Italy
| | - Myriam Carpenito
- Operative Research Unit of Emodinamica, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Roma, Italy
| | - Carmelo Dominici
- Operative Research Unit of Cardiac Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Roma, Italy
| | - Massimo Federici
- Department of Experimental Medicine, Università degli Studi di Roma Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Gian Paolo Ussia
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Roma, Italy
- Operative Research Unit of Emodinamica, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Roma, Italy
| |
Collapse
|
68
|
Pi JK, Chen XT, Zhang YJ, Chen XM, Wang YC, Xu JY, Zhou JH, Yu SS, Wu SS. Insight of immune checkpoint inhibitor related myocarditis. Int Immunopharmacol 2024; 143:113559. [PMID: 39536487 DOI: 10.1016/j.intimp.2024.113559] [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: 08/20/2024] [Revised: 10/20/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
As the understanding of immune-related mechanisms in the development and progression of cancer advances, immunotherapies, notably Immune Checkpoint Inhibitors (ICIs), have become integral in comprehensive cancer treatment strategies. ICIs reactivate T-cell cytotoxicity against tumors by blocking immune suppressive signals on T cells, such as Programmed Death-1 (PD-1) and Cytotoxic T-lymphocyte Antigen-4 (CTLA-4). Despite their beneficial effects, ICIs are associated with immune-related adverse events (irAEs), manifesting as autoimmune side effects across various organ systems. A particularly alarming irAE is life-threatening myocarditis. This rare but severe side effect of ICIs leads to significant long-term cardiac complications, including arrhythmias and heart failure, and has been observed to have a mortality rate of up to 50% in affected patients. This greatly limits the clinical application of ICI-based immunotherapy. In this review, we provide a comprehensive summary of the current knowledge regarding the diagnosis and management of ICI-related myocarditis. We also discuss the utility of preclinical mouse models in understanding and addressing this critical challenge.
Collapse
Affiliation(s)
- Jin-Kui Pi
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xiao-Ting Chen
- Animal Experimental Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Yan-Jing Zhang
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xue-Mei Chen
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Yin-Chan Wang
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Jia-Yi Xu
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Jin-Han Zhou
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Shuai-Shuai Yu
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Si-Si Wu
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
| |
Collapse
|
69
|
Guan T, Jiang Y, Tu P, Ye B, Zeng L, Luo Z, Chi K, Liang H, Yang Y, Huang J, Zhang B, Tai R, Ye J, Deng Z, Ke Y, Chen H, Zhang Z, Liu Z, Ou C. Risk classification for non-cancer death in middle-aged cancer patients. J Adv Res 2024:S2090-1232(24)00616-7. [PMID: 39730025 DOI: 10.1016/j.jare.2024.12.039] [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: 08/26/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 12/29/2024] Open
Abstract
INTRODUCTION Non-cancer events are important causes of competing mortality among cancer patients. However, the risk of non-cancer death and risk classification in middle-aged cancer patients is not clear. To comprehensively analyze the risk of non-cancer deaths in 24 different cancers among middle-aged patients. METHODS Standardized mortality rate (SMR), absolute excess risk (AER), proportion of deaths, age-adjusted mortality rate (AAMR), and the competing model were used to assess the risk of non-cancer death in middle-aged cancer patients. A non-cancer death risk classification was developed for the 24 cancer types based on the competing risk of non-cancer death and the risk of non-cancer death (hazard ratio). RESULTS A total of 1,082,030 middle-aged cancer patients of 24 cancer types was identified. The risk of non-cancer death was elevated in middle-aged cancer patients compared to the general middle-aged population (SMR = 3.37, 95 % CI 3.35-3.39, AER = 99.18). The cumulative mortality was higher for non-cancer causes compared to primary cancer in 15 cancer types. The AAMR for non-cancer causes declined from 2.3 % in 1975 to 1.4 % in 2017. A risk classification was developed to classify different cancers into 6 risk categories. CONCLUSION The risk of non-cancer death was elevated in middle-aged cancer patients and varied for different cancer types. A new risk classification system was developed to estimate the risk of non-cancer deaths in different cancers, and the 24 cancer types were classified into 6 distinct categories. These results highlight the necessity for risk stratification management for non-cancer death in middle-aged cancer patients.
Collapse
Affiliation(s)
- Tianwang Guan
- Cancer Center, The 10th Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong 523059, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou 510280, China
| | - Yanting Jiang
- Department of Radiation Oncology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang, China
| | - Peinan Tu
- Cancer Center, The 10th Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong 523059, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou 510280, China
| | - Baokui Ye
- Department of Intensive Care Unit, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangzhou, China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Liangjia Zeng
- 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
| | - Haowen Liang
- Department of Clinical Medicine, Clinical Medical School, Guangzhou Medical University, Guangzhou 510180, China
| | - Yuting Yang
- Department of Clinical Medicine, Clinical Medical School, Guangzhou Medical University, Guangzhou 510180, China
| | - Jinqi Huang
- Department of Clinical Medicine, Clinical Medical School, Guangzhou Medical University, Guangzhou 510180, China
| | - Binghua Zhang
- Department of Clinical Medicine, Clinical Medical School, Guangzhou Medical University, Guangzhou 510180, China
| | - Rundong Tai
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou 510280, China; The 10th Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong 523059, China
| | - Jujian Ye
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou 510280, China; The 10th Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong 523059, China
| | - Zhilin Deng
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou 510280, China; The 10th Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong 523059, China
| | - Yushen Ke
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou 510280, China; The 10th Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong 523059, China
| | - Huiwan Chen
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou 510280, China; The 10th Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong 523059, China
| | - Zhiling Zhang
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China; Guangdong Provincial Clinical Research Center for Cancer, State Key Laboratory of Oncology in Southern China, Guangzhou, China.
| | - Zhigang Liu
- Cancer Center, The 10th Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong 523059, China.
| | - Caiwen Ou
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou 510280, China; The 10th Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong 523059, China.
| |
Collapse
|
70
|
Theofilis P, Vlachakis PK, Oikonomou E, Drakopoulou M, Karakasis P, Apostolos A, Pamporis K, Tsioufis K, Tousoulis D. Cancer Therapy-Related Cardiac Dysfunction: A Review of Current Trends in Epidemiology, Diagnosis, and Treatment. Biomedicines 2024; 12:2914. [PMID: 39767820 PMCID: PMC11673750 DOI: 10.3390/biomedicines12122914] [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: 11/23/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Cancer therapy-related cardiac dysfunction (CTRCD) has emerged as a significant concern with the rise of effective cancer treatments like anthracyclines and targeted therapies such as trastuzumab. While these therapies have improved cancer survival rates, their unintended cardiovascular side effects can lead to heart failure, cardiomyopathy, and arrhythmias. The pathophysiology of CTRCD involves oxidative stress, mitochondrial dysfunction, and calcium dysregulation, resulting in irreversible damage to cardiomyocytes. Inflammatory cytokines, disrupted growth factor signaling, and coronary atherosclerosis further contribute to this dysfunction. Advances in cardio-oncology have led to the early detection of CTRCD using cardiac biomarkers like troponins and imaging techniques such as echocardiography and cardiac magnetic resonance (CMR). These tools help identify asymptomatic patients at risk of cardiac events before the onset of clinical symptoms. Preventive strategies, including the use of cardioprotective agents like beta-blockers, angiotensin-converting enzyme inhibitors, mineralocorticoid receptor antagonists, and sodium-glucose cotransporter-2 inhibitors have shown promise in reducing the incidence of CTRCD. This review summarizes the mechanisms, detection methods, and emerging treatments for CTRCD, emphasizing the importance of interdisciplinary collaboration between oncologists and cardiologists to optimize care and improve both cancer and cardiovascular outcomes.
Collapse
Affiliation(s)
- Panagiotis Theofilis
- 1st Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.); (P.K.V.); (M.D.); (A.A.); (K.P.); (K.T.)
| | - Panayotis K. Vlachakis
- 1st Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.); (P.K.V.); (M.D.); (A.A.); (K.P.); (K.T.)
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, Thoracic Diseases General Hospital Sotiria, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Maria Drakopoulou
- 1st Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.); (P.K.V.); (M.D.); (A.A.); (K.P.); (K.T.)
| | - Paschalis Karakasis
- 2nd Department of Cardiology, Hippokration General Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece;
| | - Anastasios Apostolos
- 1st Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.); (P.K.V.); (M.D.); (A.A.); (K.P.); (K.T.)
| | - Konstantinos Pamporis
- 1st Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.); (P.K.V.); (M.D.); (A.A.); (K.P.); (K.T.)
| | - Konstantinos Tsioufis
- 1st Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.); (P.K.V.); (M.D.); (A.A.); (K.P.); (K.T.)
| | - Dimitris Tousoulis
- 1st Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.T.); (P.K.V.); (M.D.); (A.A.); (K.P.); (K.T.)
| |
Collapse
|
71
|
Li Y, Li H, Ye X, Zhu Z, Qiu Y. Novel models by machine learning to predict the risk of cardiac disease-specific death in young patients with breast cancer. Discov Oncol 2024; 15:809. [PMID: 39692943 DOI: 10.1007/s12672-024-01676-9] [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: 09/07/2024] [Accepted: 12/06/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND With the tremendous leap of various adjuvant therapies, breast cancer (BC)-related deaths have decreased significantly. Increasing attention was focused on the effect of cardiac disease on BC survivors, while limited existing population-based studies lay emphasis on the young age population. METHOD Data of BC patients aged less than 50 years was collected from the SEER database. A competing risk model was introduced to analyze the effects of clinicopathology variables on the cardiac disease-specific death (CDSD) risks of these patients. Further, an XGBoost prediction model was constructed to predict the risk of CDSD. Prediction performance was assessed using the receiver operating characteristic (ROC) analysis, area under the POC curve (AUC) values, calibration curves, decision curves, and confusion matrix, and SHapley Additive exPlanations (SHAP) were used to interpret the models. RESULTS Our competing risk analysis proved that young BC patients with older age, low household income, non-metropolitan residential environment, black race, unmarried status, HR + subtype, higher T stage (T2-4), receiving chemotherapy, and non-surgery are under higher risk of CDSD. Further, five machine learning models were constructed to predict the CDSD risks of young BC patients, among which the XGBoost models showed the highest AUC value (train set: AUC = 0.846; test set: AUC = 0.836). The confusion matrix of the XGBoost model demonstrated that the sensitivity, specificity, and correction were 0.81, 0.94, and 0.94 for the train set, and 0.82, 0.95, and 0.96 for the test set, respectively. The SHAP graph indicated that median household income, marital status, race, and age at diagnosis were the top four strongest predictors. CONCLUSION Independent CDSD risk factors for young BC patients were identified, and machine-learning prognostic models were constructed to predict their CDSD risks. Our validation results indicated that the predicted probability of our XGBoost model agrees well with the actual CDSD risks, and it can help recognize high-risk populations and therefore develop effective cardioprotection strategies. Hopefully, our findings can support the growth of the new field of cardio-oncology.
Collapse
Affiliation(s)
- Yi Li
- Department of Geriatrics, Hematology and Oncology Ward, The Second Affiliated Hospital, School of Medicine, South China University of Technology, No.1 Panfu Road, Guangzhou, China
| | - Handong Li
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xuan Ye
- Department of Breast and Thyroid Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Zhigang Zhu
- Department of Geriatrics, Hematology and Oncology Ward, The Second Affiliated Hospital, School of Medicine, South China University of Technology, No.1 Panfu Road, Guangzhou, China.
| | - Yixuan Qiu
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, No. 106 Zhongshan Second Road, Guangzhou, China.
| |
Collapse
|
72
|
Conyers R, Elliott DA. Straight to the heart: Protecting the patient's heart during chemotherapy. Cell Stem Cell 2024; 31:1715-1716. [PMID: 39642857 DOI: 10.1016/j.stem.2024.10.010] [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: 10/02/2024] [Revised: 10/09/2024] [Accepted: 10/14/2024] [Indexed: 12/09/2024]
Abstract
How do we protect the heart during chemotherapy with the anthracycline drug class? Tackling this question, Liu et al. combined pluripotent stem cell models, CRISPR genetic screens, and molecular modeling to identify indisulam as a potential cardioprotective drug in this issue of Cell Stem Cell.
Collapse
Affiliation(s)
- Rachel Conyers
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC, Australia; Children's Cancer Centre, Royal Melbourne Hospital, Parkville, VIC, Australia; Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia; The Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Parkville, VIC, Australia
| | - David A Elliott
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC, Australia; Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia; The Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Parkville, VIC, Australia; Australian Regenerative Medicine Institute, Monash University, Wellington Road, Clayton, VIC, Australia.
| |
Collapse
|
73
|
Blum SM, Zlotoff DA, Smith NP, Kernin IJ, Ramesh S, Zubiri L, Caplin J, Samanta N, Martin S, Wang M, Tirard A, Song Y, Xu KH, Barth J, Sen P, Slowikowski K, Tantivit J, Manakongtreecheep K, Arnold BY, Nasrallah M, Pinto CJ, McLoughlin D, Jackson M, Chan P, Lawless A, Michaud WA, Sharova T, Nieman LT, Gainor JF, Wu CJ, Juric D, Mino-Kenudson M, Oliveira G, Sullivan RJ, Boland GM, Stone JR, Thomas MF, Neilan TG, Reynolds KL, Villani AC. Immune responses in checkpoint myocarditis across heart, blood and tumour. Nature 2024; 636:215-223. [PMID: 39506125 DOI: 10.1038/s41586-024-08105-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 09/24/2024] [Indexed: 11/08/2024]
Abstract
Immune checkpoint inhibitors are widely used anticancer therapies1 that can cause morbid and potentially fatal immune-related adverse events such as immune-related myocarditis (irMyocarditis)2-5. The pathogenesis of irMyocarditis and its relationship to antitumour immunity remain poorly understood. Here we sought to define immune responses in heart, tumour and blood in patients with irMyocarditis by leveraging single-cell RNA sequencing coupled with T cell receptor (TCR) sequencing, microscopy and proteomics analyses of samples from 28 patients with irMyocarditis and 41 unaffected individuals. Analyses of 84,576 cardiac cells by single-cell RNA sequencing combined with multiplexed microscopy demonstrated increased frequencies and co-localization of cytotoxic T cells, conventional dendritic cells and inflammatory fibroblasts in irMyocarditis heart tissue. Analyses of 366,066 blood cells revealed decreased frequencies of plasmacytoid dendritic cells, conventional dendritic cells and B lineage cells but an increased frequency of other mononuclear phagocytes in irMyocarditis. Fifty-two heart-expanded TCR clones from eight patients did not recognize the putative cardiac autoantigens α-myosin, troponin I or troponin T. Additionally, TCRs enriched in heart tissue were largely nonoverlapping with those enriched in paired tumour tissue. The presence of heart-expanded TCRs in a cycling blood CD8 T cell population was associated with fatal irMyocarditis case status. Collectively, these findings highlight crucial biology driving irMyocarditis and identify putative biomarkers.
Collapse
Affiliation(s)
- Steven M Blum
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Mass General Cancer Center, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Daniel A Zlotoff
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Neal P Smith
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Isabela J Kernin
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Swetha Ramesh
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Leyre Zubiri
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Mass General Cancer Center, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Joshua Caplin
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Nandini Samanta
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Sidney Martin
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Mike Wang
- Mass General Cancer Center, Boston, MA, USA
| | - Alice Tirard
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Yuhui Song
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Katherine H Xu
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Jaimie Barth
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Pritha Sen
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Transplant, Oncology and Immunocompromised Host Program, Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Division of Rheumatology, North Shore Physicians Group, Department of Medicine, Mass General Brigham Healthcare Center, Lynn, MA, USA
| | - Kamil Slowikowski
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Jessica Tantivit
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Kasidet Manakongtreecheep
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Benjamin Y Arnold
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Mazen Nasrallah
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Division of Rheumatology, North Shore Physicians Group, Department of Medicine, Mass General Brigham Healthcare Center, Lynn, MA, USA
| | - Christopher J Pinto
- Mass General Cancer Center, Boston, MA, USA
- Clinical Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Daniel McLoughlin
- Mass General Cancer Center, Boston, MA, USA
- Clinical Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Monica Jackson
- Mass General Cancer Center, Boston, MA, USA
- Clinical Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - PuiYee Chan
- Mass General Cancer Center, Boston, MA, USA
- Clinical Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Aleigha Lawless
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - William A Michaud
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Tatyana Sharova
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Linda T Nieman
- Harvard Medical School, Boston, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Justin F Gainor
- Mass General Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Catherine J Wu
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Dejan Juric
- Mass General Cancer Center, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Mari Mino-Kenudson
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Giacomo Oliveira
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ryan J Sullivan
- Mass General Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Genevieve M Boland
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - James R Stone
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Molly F Thomas
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Division of Gastroenterology, Department of Medicine, Oregon Health and Sciences University, Portland, OR, USA
- Department of Cell, Developmental, and Cancer Biology, Oregon Health and Sciences University, Portland, OR, USA
| | - Tomas G Neilan
- Harvard Medical School, Boston, MA, USA
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Kerry L Reynolds
- Mass General Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Alexandra-Chloé Villani
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
74
|
Leong DP, Waliany S, Abdel-Qadir H, Atkins KM, Neilan TG, Lang NN, Liu JE, Blaes AH, Mian HS, Moore HN, Hajjar LA, Morgans AK, Ellis PM, Dent S. Cardiovascular Considerations During Cancer Therapy: Gaps in Evidence and JACC: CardioOncology Expert Panel Recommendations. JACC CardioOncol 2024; 6:815-834. [PMID: 39801647 PMCID: PMC11711816 DOI: 10.1016/j.jaccao.2024.06.005] [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: 09/07/2023] [Revised: 06/13/2024] [Accepted: 06/27/2024] [Indexed: 01/03/2025] Open
Abstract
The administration of certain cancer therapies can be associated with the development of cardiovascular toxicity or complications. This spectrum of toxicities is broad and requires nuanced approaches for prevention, identification, and management. This expert panel summarizes the consensus of opinions of diverse health care professionals in several key areas: 1) cardioprotection involves strategies aimed at the primary prevention of cancer therapy-related cardiovascular toxicity; 2) surveillance entails monitoring for cancer therapy-related cardiovascular toxicity during cancer therapy; 3) permissive cardiotoxicity is the informed continuation of cancer therapy in the presence of cardiovascular toxicity, along with the implementation of mitigating cardiovascular treatments; and 4) special considerations include the invasive management of severe cardiovascular disease in patients receiving treatments for advanced cancer and the exploration of drug-drug interactions in cardio-oncology. In this expert panel, we also highlight gaps in evidence in an effort to continue to advance science in the cardiovascular care of our patients undergoing cancer therapy.
Collapse
Affiliation(s)
- Darryl P. Leong
- Population Health Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Sarah Waliany
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts, USA
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Husam Abdel-Qadir
- Women’s College Hospital, Peter Munk Cardiac Centre, University of Toronto, Toronto, Ontario, Canada
| | - Katelyn M. Atkins
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Tomas G. Neilan
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ninian N. Lang
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Jennifer E. Liu
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Anne H. Blaes
- Division of Hematology/Oncology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Hira S. Mian
- Juravinski Cancer Center, Department of Oncology, McMaster University, Hamilton, Ontario, Canada
| | - Heather N. Moore
- Department of Pharmacy, Duke University Medical Center, Durham, North Carolina, USA
| | - Ludhmila A. Hajjar
- Cardio-Oncology Department, InCor, Universidade de São Paolo, São Paolo, Brazil
| | - Alicia K. Morgans
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Peter M. Ellis
- Juravinski Cancer Center, Department of Oncology, McMaster University, Hamilton, Ontario, Canada
| | - Susan Dent
- Duke Cancer Institute, Department of Medicine, Duke University, Durham, North Carolina, USA
| |
Collapse
|
75
|
Barcia Durán JG, Das D, Gildea M, Amadori L, Gourvest M, Kaur R, Eberhardt N, Smyrnis P, Cilhoroz B, Sajja S, Rahman K, Fernandez DM, Faries P, Narula N, Vanguri R, Goldberg IJ, Fisher EA, Berger JS, Moore KJ, Giannarelli C. Immune checkpoint landscape of human atherosclerosis and influence of cardiometabolic factors. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1482-1502. [PMID: 39613875 PMCID: PMC11634783 DOI: 10.1038/s44161-024-00563-4] [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: 12/18/2023] [Accepted: 10/21/2024] [Indexed: 12/01/2024]
Abstract
Immune checkpoint inhibitor (ICI) therapies can increase the risk of cardiovascular events in survivors of cancer by worsening atherosclerosis. Here we map the expression of immune checkpoints (ICs) within human carotid and coronary atherosclerotic plaques, revealing a network of immune cell interactions that ICI treatments can unintentionally target in arteries. We identify a population of mature, regulatory CCR7+FSCN1+ dendritic cells, similar to those described in tumors, as a hub of IC-mediated signaling within plaques. Additionally, we show that type 2 diabetes and lipid-lowering therapies alter immune cell interactions through PD-1, CTLA4, LAG3 and other IC targets in clinical development, impacting plaque inflammation. This comprehensive map of the IC interactome in healthy and cardiometabolic disease states provides a framework for understanding the potential adverse and beneficial impacts of approved and investigational ICIs on atherosclerosis, setting the stage for designing ICI strategies that minimize cardiovascular disease risk in cancer survivors.
Collapse
Grants
- R35HL135799 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL084312 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- P30 CA016087 NCI NIH HHS
- 23POST1029885 American Heart Association (American Heart Association, Inc.)
- R35 HL135799 NHLBI NIH HHS
- R01 HL153712 NHLBI NIH HHS
- 20SFRN35210252 American Heart Association (American Heart Association, Inc.)
- R01HL165258 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 965509 American Heart Association (American Heart Association, Inc.)
- R01HL153712 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL165258 NHLBI NIH HHS
- R01 HL084312 NHLBI NIH HHS
- U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
Collapse
Affiliation(s)
- José Gabriel Barcia Durán
- NYU Cardiovascular Research Center, Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Dayasagar Das
- NYU Cardiovascular Research Center, Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Michael Gildea
- NYU Cardiovascular Research Center, Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Letizia Amadori
- NYU Cardiovascular Research Center, Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Morgane Gourvest
- NYU Cardiovascular Research Center, Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Ravneet Kaur
- NYU Cardiovascular Research Center, Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Natalia Eberhardt
- NYU Cardiovascular Research Center, Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Panagiotis Smyrnis
- NYU Cardiovascular Research Center, Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Burak Cilhoroz
- NYU Cardiovascular Research Center, Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Swathy Sajja
- NYU Cardiovascular Research Center, Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Karishma Rahman
- Division of Cardiology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dawn M Fernandez
- Division of Cardiology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Peter Faries
- Department of Surgery, Vascular Division, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Navneet Narula
- Department of Pathology, New York University Grossman School of Medicine, New York University Langone Health, New York, NY, USA
| | - Rami Vanguri
- Division of Precision Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Ira J Goldberg
- Division of Endocrinology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Edward A Fisher
- NYU Cardiovascular Research Center, Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA
| | - Jeffrey S Berger
- NYU Cardiovascular Research Center, Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Kathryn J Moore
- NYU Cardiovascular Research Center, Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA.
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA.
| | - Chiara Giannarelli
- NYU Cardiovascular Research Center, Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA.
- Department of Pathology, New York University Grossman School of Medicine, New York University Langone Health, New York, NY, USA.
| |
Collapse
|
76
|
Bhatti AW, Patel R, Dani SS, Khadke S, Makwana B, Lessey C, Shah J, Al-Husami Z, Yang EH, Thavendiranathan P, Neilan TG, Sadler D, Cheng RK, Dent SF, Liu J, Lopez-Fernandez T, Herrmann J, Scherrer-Crosbie M, Lenihan DJ, Hayek SS, Ky B, Deswal A, Barac A, Nohria A, Ganatra S. SGLT2i and Primary Prevention of Cancer Therapy-Related Cardiac Dysfunction in Patients With Diabetes. JACC CardioOncol 2024; 6:863-875. [PMID: 39801650 PMCID: PMC11711834 DOI: 10.1016/j.jaccao.2024.08.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/03/2024] [Indexed: 01/16/2025] Open
Abstract
Background Specific cancer treatments can lead to cancer therapy-related cardiac dysfunction (CTRCD). Sodium glucose cotransporter-2 inhibitors (SGLT2is) can potentially prevent these cardiotoxic effects. Objectives This study sought to determine whether SGLT2i use is associated with a lower incidence of CTRCD in patients with type 2 diabetes mellitus (T2DM) and cancer, exposed to potentially cardiotoxic antineoplastic agents, and without a prior documented history of cardiomyopathy or heart failure. Methods We conducted a retrospective analysis of patients aged ≥18 years within the TriNetX database with T2DM, cancer, exposure to cardiotoxic therapies, and no prior documented history of cardiomyopathy or heart failure. Patients were categorized by SGLT2i use. After propensity score matching, outcomes were compared over 12 months using Cox proportional HRs. Subgroup analyses focusing on different cancer therapy classes were performed. Results The study included 8,675 propensity-matched patients in each cohort (mean age = ∼65 years, 42% females, 71% White, ∼19% gastrointestinal malignancy, and ∼25% anthracyclines). Patients prescribed SGLT2is had a lower risk of developing CTRCD (HR: 0.76: 95% CI: 0.69-0.84). SGLT2is also reduced heart failure exacerbations (HR: 0.81; 95% CI: 0.72-0.90), all-cause mortality (HR: 0.67; 95% CI: 0.61-0.74), and all-cause hospitalizations/emergency department visits (HR: 0.93; 95% CI: 0.89-0.97). Subgroup analyses also demonstrated reduced CTRCD risk across various classes of cancer therapies in patients prescribed SGLT2is. Conclusions SGLT2i administration was associated with a significantly decreased risk of developing CTRCD in patients with T2DM and cancer.
Collapse
Affiliation(s)
- Ammar W. Bhatti
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Beth Israel Lahey Health, Burlington, Massachusetts, USA
| | - Rushin Patel
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Beth Israel Lahey Health, Burlington, Massachusetts, USA
| | - Sourbha S. Dani
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Beth Israel Lahey Health, Burlington, Massachusetts, USA
| | - Sumanth Khadke
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Beth Israel Lahey Health, Burlington, Massachusetts, USA
| | - Bhargav Makwana
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Beth Israel Lahey Health, Burlington, Massachusetts, USA
| | - Candace Lessey
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Beth Israel Lahey Health, Burlington, Massachusetts, USA
| | - Jui Shah
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Beth Israel Lahey Health, Burlington, Massachusetts, USA
| | - Zaid Al-Husami
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Beth Israel Lahey Health, Burlington, Massachusetts, USA
| | - Eric H. Yang
- University of California-Los Angeles, Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California-Los Angeles, Los Angeles, California, USA
| | - Paaladinesh Thavendiranathan
- Department of Medicine, Division of Cardiology, Ted Rogers Program in Cardiotoxicity Prevention, Peter Munk Cardiac Center, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Tomas G. Neilan
- Cardio-Oncology Program, Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Diego Sadler
- Cardio-Oncology Section, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Weston, Florida, USA
| | - Richard K. Cheng
- Division of Cardiology, University of Washington, Seattle, Washington, USA
| | - Susan F. Dent
- Duke Cancer Institute, Department of Medicine, Duke University, Durham, North Carolina
| | - Jennifer Liu
- Cardio-Oncology Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Teresa Lopez-Fernandez
- Division of Cardiology, Cardio-Oncology Unit, La Paz University Hospital, Hospital La Paz Institute for Health Research, Madrid, Spain
- Division of Cardiology, Quironsalud Madrid University Hospital, Madrid, Spain
| | - Joerg Herrmann
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Daniel J. Lenihan
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Salim S. Hayek
- Division of Cardiology, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, USA
| | - Bonnie Ky
- Thalheimer Center for Cardio-Oncology, Abramson Cancer Center and Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Anita Deswal
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ana Barac
- Inova Schar Heart and Vascular Institute, Inova Schar Cancer Institute, Fairfax, Virginia, USA
| | - Anju Nohria
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sarju Ganatra
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Beth Israel Lahey Health, Burlington, Massachusetts, USA
| |
Collapse
|
77
|
López-Fernández T, Marco I, Aznar MC, Barac A, Bergler-Klein J, Meattini I, Scott JM, Cardinale D, Dent S. Breast cancer and cardiovascular health. Eur Heart J 2024; 45:4366-4382. [PMID: 39320463 DOI: 10.1093/eurheartj/ehae637] [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: 12/29/2023] [Revised: 03/08/2024] [Accepted: 09/06/2024] [Indexed: 09/26/2024] Open
Abstract
Modern cancer therapies greatly improve clinical outcomes for both early and advanced breast cancer patients. However, these advances have raised concerns about potential short- and long-term toxicities, including cardiovascular toxicities. Therefore, understanding the common risk factors and underlying pathophysiological mechanisms contributing to cardiovascular toxicity is essential to ensure best breast cancer outcomes. While cardio-oncology has emerged as a sub-speciality to address these challenges, it is essential that all cardiologists recognize and understand the cardiovascular consequences of cancer therapy. This review aims to provide a comprehensive overview of the potential adverse cardiovascular effects associated with modern breast cancer therapies. A preventive, diagnostic, and therapeutic workflow to minimize the impact of cardiovascular toxicity on patient outcomes is presented. Key aspects of this workflow include regular monitoring of cardiovascular function, early detection and management of cancer therapy-related cardiovascular toxicities, and optimization of cardiovascular risk factor control. By highlighting the gaps in knowledge in some areas, this review aims to emphasize the critical role of cardio-oncology research in ensuring the holistic well-being of patients with breast cancer.
Collapse
Affiliation(s)
- Teresa López-Fernández
- Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, C/Paseo de la Castellana n° 261, 28046 Madrid, Spain
- Cardiology Department, Quironsalud University Hospital, C. Diego de Velázquez, 1, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - Irene Marco
- Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, C/Paseo de la Castellana n° 261, 28046 Madrid, Spain
| | - Marianne C Aznar
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Ana Barac
- Inova ScharHeart and Vascular, Inova Schar Cancer Institute, Fall Church, VA, USA
| | - Jutta Bergler-Klein
- Department of Cardiology, University Clinic of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Icro Meattini
- Department of Experimental and Clinical Biomedical Sciences 'M. Serio', Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Jessica M Scott
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daniela Cardinale
- Cardioncology Unit, European Institute of Oncology, I.R.C.C.S., Milan, Italy
| | - Susan Dent
- Wilmot Cancer Institute, Department of Medicine, University of Rochester, Rochester, NY, USA
| |
Collapse
|
78
|
Barac A, Mauro L, Harnden K. From setbacks to success: building a promising path for strain-guided cardioprotection during anthracycline treatment. Eur Heart J 2024; 45:4425-4427. [PMID: 39217602 DOI: 10.1093/eurheartj/ehae598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Affiliation(s)
- Ana Barac
- Inova Schar Heart and Vascular, Fairfax, VA, USA
- Inova Schar Cancer, Fairfax, VA, USA
| | | | | |
Collapse
|
79
|
Senechal I, Andres MS, Tong J, Ramalingam S, Nazir MS, Rosen SD, Young K, Idaikkadar P, Larkin J, Lyon AR. Risk Stratification, Screening and Treatment of BRAF/MEK Inhibitors-Associated Cardiotoxicity. Curr Oncol Rep 2024; 26:1431-1441. [PMID: 39316222 DOI: 10.1007/s11912-024-01599-2] [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: 08/13/2024] [Indexed: 09/25/2024]
Abstract
PURPOSE OF REVIEW In this review article we describe the cardiovascular adverse events associated with BRAF and MEK inhibitors as well as their pathophysiologic mechanisms and provide up to date guidance for risk stratified surveillance of patients on treatment and the optimal management of emergent cardiotoxicities. RECENT FINDINGS Combination BRAF/MEK inhibition has become an established standard treatment option for patients with a wide variety of BRAF mutant haematological and solid organ cancers, its use is most commonly associated with stage three and metastatic melanoma. The introduction of these targeted drugs has significantly improved the prognosis of previously treatment resistant cancers. It is increasingly recognised that these drugs have a number of cardiovascular toxicities including left ventricular systolic dysfunction, hypertension and QTc interval prolongation. Whilst cardiotoxicity is largely reversible and manageable with medical therapy, it does limit the effective use of these highly active agents.
Collapse
Affiliation(s)
- Isabelle Senechal
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK.
- Centre Hospitalier Universitaire de Québec, Québec City, Québec, Canada.
| | - Maria Sol Andres
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Jieli Tong
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Sivatharshini Ramalingam
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Muhummad Sohaib Nazir
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
- School of Biomedical Engineering and Imaging Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Stuart D Rosen
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Kate Young
- Royal Marsden Hospital Foundation Trust, London, UK
| | | | - James Larkin
- Royal Marsden Hospital Foundation Trust, London, UK
| | - Alexander R Lyon
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| |
Collapse
|
80
|
Watanabe M, Fujiki S, Okura Y, Toshikawa C, Ikarashi M, Kanbayashi C, Kaneko K, Kikuchi A, Sakata E, Tsuchida K, Ozaki K, Moro K, Kubota N, Kashimura T, Moriyama M, Sato N, Tanabe N, Koyama Y, Wakai T, Saijo Y, Inomata T. Increasing survivors of anthracycline-related cardiomyopathy with breast cancer in trastuzumab era: thirty-one-year trends in a Japanese Community. Breast Cancer 2024; 31:1080-1091. [PMID: 39138789 PMCID: PMC11489246 DOI: 10.1007/s12282-024-01623-0] [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: 04/19/2024] [Accepted: 08/01/2024] [Indexed: 08/15/2024]
Abstract
BACKGROUND Trastuzumab has improved breast cancer (BC) prognosis and reduced anthracycline use. However, the characteristic changes of anthracycline-related cardiomyopathy (ARCM) in patients with BC remain unclear. We aimed to update our understanding of ARCM in the trastuzumab era. METHODS This retrospective observational cohort study included 2959 patients with BC treated with anthracyclines at three regional cancer centers in Niigata City between 1990 and 2020. Seventy-five patients (2.5%) developed ARCM and were categorized into two groups: pre- 2007 (early phase) and post-2007 (late phase), corresponding to before and during the trastuzumab era in Japan. RESULTS ARCM incidence peaked at 6% in the 1990s, then decreased and stabilized at 2% until the 2010s. Survivors of anthracycline-treated BC increased more rapidly in the late phase, with four times as many patients with ARCM compared to the end of the early phase (26 and six, respectively). Although the rate of change in accumulation from the early phase to the late phase was slight in the anthracycline-treated BC group, it was more pronounced in the ARCM group (P < 0.001). Mean anthracycline use in the late phase was significantly lower than in the early phase (307 vs. 525 mg/m2, P < 0.001). Five-year survival rates in the late phase tended to be higher than early phase (45% and 28%, respectively. P = 0.058). Human epidermal growth factor receptor type 2 (HER2) positivity with trastuzumab therapy in the late phase was an independent predictor for mortality within 10 years (hazard ratio = 0.24, 95% confidence interval: 0.10-0.56; P = 0.001). CONCLUSIONS HER2-positive patients with ARCM receiving trastuzumab therapy had a better prognosis than HER2-positive and HER2-negative patients with ARCM not receiving trastuzumab therapy, and this trend has been increasing in the trastuzumab era. These findings highlight the importance of HER2-targeted treatments in improving prognosis for BC patients with ARCM.
Collapse
Affiliation(s)
- Mitsuhiro Watanabe
- Department of Cardiovascular Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Shinya Fujiki
- Department of Cardiovascular Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yuji Okura
- Department of Onco-Cardiology, Niigata Cancer Center Hospital, 2-15-3 Kawagishi-cho, Chuo-ku, Niigata, 951-8560, Japan.
| | - Chie Toshikawa
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
- Department of Breast Surgery, Niigata City General Hospital, Niigata, Japan
| | - Mayuko Ikarashi
- Department of Breast Oncology, Niigata Cancer Center Hospital, Niigata, Japan
| | - Chizuko Kanbayashi
- Department of Breast Oncology, Niigata Cancer Center Hospital, Niigata, Japan
| | - Koji Kaneko
- Department of Breast Oncology, Niigata Cancer Center Hospital, Niigata, Japan
| | - Akira Kikuchi
- Department of Gynecology, Niigata Cancer Center Hospital, Niigata, Japan
| | - Eiko Sakata
- Department of Breast Surgery, Niigata City General Hospital, Niigata, Japan
| | - Keiichi Tsuchida
- Department of Cardiology, Niigata City General Hospital, Niigata, Japan
| | - Kazuyuki Ozaki
- Department of Cardiovascular Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
- Department of Cardiology, Niigata City General Hospital, Niigata, Japan
| | - Kazuki Moro
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Naoki Kubota
- Department of Cardiovascular Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Takeshi Kashimura
- Department of Cardiovascular Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Masato Moriyama
- Department of Pathophysiology, Faculty of Pharmacy, Niigata University of Pharmacy and Medical and Life Sciences, Niigata, Japan
| | - Nobuaki Sato
- Department of Breast Oncology, Niigata Cancer Center Hospital, Niigata, Japan
| | - Naohito Tanabe
- Department of Health and Nutrition, Faculty of Human Life Studies, University of Niigata Prefecture, Niigata, Japan
| | - Yu Koyama
- Department of Nursing, Niigata University Graduate School of Health Sciences, Niigata, Japan
| | - Toshifumi Wakai
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yasuo Saijo
- Department of Medical Oncology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Takayuki Inomata
- Department of Cardiovascular Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
81
|
Jacquemyn X, Chinni BK, Barnes BT, Rao S, Kutty S, Manlhiot C. Unsupervised machine learning identifies distinct phenotypes in cardiac complications of pediatric patients treated with anthracyclines. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:74. [PMID: 39468669 PMCID: PMC11514752 DOI: 10.1186/s40959-024-00276-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 10/17/2024] [Indexed: 10/30/2024]
Abstract
BACKGROUND Anthracyclines are essential in pediatric cancer treatment, but patients are at risk cancer therapy-related cardiac dysfunction (CTRCD). Standardized definitions by the International Cardio-Oncology Society (IC-OS) aim to enhance precision in risk assessment. OBJECTIVES Categorize distinct phenotypes among pediatric patients undergoing anthracycline chemotherapy using unsupervised machine learning. METHODS Pediatric cancer patients undergoing anthracycline chemotherapy at our institution were retrospectively included. Clinical and echocardiographic data at baseline, along with follow-up data, were collected from patient records. Unsupervised machine learning was performed, involving dimensionality reduction using principal component analysis and K-means clustering to identify different phenotypic clusters. Identified phenogroups were analyzed for associations with CTRCD, defined following contemporary IC-OS definitions, and hypertensive response. RESULTS A total of 187 patients (63.1% male, median age 15.5 years [10.4-18.7]) were included and received anthracycline chemotherapy with a median treatment duration of 0.66 years [0.35-1.92]. Median follow-up duration was 2.78 years [1.31-4.21]. Four phenogroups were identified with following distribution: Cluster 0 (32.6%, n = 61), Cluster 1 (13.9%, n = 26), Cluster 2 (24.6%, n = 46), and Cluster 3 (28.9%, n = 54). Cluster 0 showed the highest risk of moderate CTRCD (HR: 3.10 [95% CI: 1.18-8.16], P = 0.022) compared to other clusters. Cluster 3 demonstrated a protective effect against hypertensive response (HR: 0.30 [95% CI: 0.13- 0.67], P = 0.003) after excluding baseline hypertensive patients. Longitudinal assessments revealed differences in global longitudinal strain and systolic blood pressure among phenogroups. CONCLUSIONS Unsupervised machine learning identified distinct phenogroups among pediatric cancer patients undergoing anthracycline chemotherapy, offering potential for personalized risk assessment.
Collapse
Affiliation(s)
- Xander Jacquemyn
- Department of Pediatrics, The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Johns Hopkins School of Medicine, Johns Hopkins University, Johns Hopkins Hospital, 600 N. Wolfe Street, 1389 Blalock, Baltimore, MD, 21287, USA
- Department of Cardiovascular Sciences, KU Leuven & Congenital and Structural Cardiology, UZ Leuven, Leuven, Belgium
| | - Bhargava K Chinni
- Department of Pediatrics, The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Johns Hopkins School of Medicine, Johns Hopkins University, Johns Hopkins Hospital, 600 N. Wolfe Street, 1389 Blalock, Baltimore, MD, 21287, USA
| | - Benjamin T Barnes
- Department of Pediatrics, The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Johns Hopkins School of Medicine, Johns Hopkins University, Johns Hopkins Hospital, 600 N. Wolfe Street, 1389 Blalock, Baltimore, MD, 21287, USA
| | - Sruti Rao
- Department of Pediatrics, The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Johns Hopkins School of Medicine, Johns Hopkins University, Johns Hopkins Hospital, 600 N. Wolfe Street, 1389 Blalock, Baltimore, MD, 21287, USA
| | - Shelby Kutty
- Department of Pediatrics, The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Johns Hopkins School of Medicine, Johns Hopkins University, Johns Hopkins Hospital, 600 N. Wolfe Street, 1389 Blalock, Baltimore, MD, 21287, USA
| | - Cedric Manlhiot
- Department of Pediatrics, The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Johns Hopkins School of Medicine, Johns Hopkins University, Johns Hopkins Hospital, 600 N. Wolfe Street, 1389 Blalock, Baltimore, MD, 21287, USA.
| |
Collapse
|
82
|
Yuan C, Peng L, Yang D. Electrocardiogram Intricacies in a Patient With Prostate Cancer-The Heart of the Matter. JAMA Intern Med 2024:2825460. [PMID: 39466225 DOI: 10.1001/jamainternmed.2024.2282] [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: 10/29/2024]
Abstract
This case report describes the electrocardiagram findings of a patient in their 60s with chest tightness and dyspnea, left ventricular systolic impairment, and elevated troponin I levels.
Collapse
Affiliation(s)
- Cuizhen Yuan
- Department of Cardiology, Peking University People's Hospital, Beijing, China
| | - Linfeng Peng
- Department of Cardiology, Peking University People's Hospital, Beijing, China
| | - Dandan Yang
- Department of Cardiology, Peking University People's Hospital, Beijing, China
| |
Collapse
|
83
|
Guan Z, Yao T, Liu G, Liu J, Guo L, Li Z, Ma J. Peripheral biomarkers to assess risk, severity, and prognosis of immune checkpoint inhibitor-associated myocarditis: a retrospective clinical study. Front Cardiovasc Med 2024; 11:1465743. [PMID: 39512372 PMCID: PMC11540693 DOI: 10.3389/fcvm.2024.1465743] [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: 07/16/2024] [Accepted: 10/08/2024] [Indexed: 11/15/2024] Open
Abstract
Background Immune checkpoint inhibitor-associated myocarditis (ICI myocarditis) is an infrequent but potentially fatal immune-related adverse event. This study aimed to identify valuable indicators for risk prediction and evaluation of disease severity and outcomes. Methods A total of 79 patients with severe or mild ICI myocarditis and 158 controls without post-ICI immune-related adverse events were enrolled in this retrospective study. The clinical application value of a series of simple biomarkers were tested. Results Higher levels of the systemic immune-inflammation index (SII), neutrophil-to-eosinophil ratio (NER), aspartate transferase-to-albumin ratio (AAR), and lactic dehydrogenase-to-albumin ratio (LAR) at myocarditis onset were associated with severe disease conditions. In the receiver operating characteristic analysis, biomarkers areas under the curve (AUC) close to or greater than 0.8 were LAR (AUC: 0.810) and AAR (AUC: 0.806). Patients with higher SII, AAR, and LAR also exhibited poorer overall survival. The SII, NER, AAR, and LAR before the last ICI treatment increased relative to baseline in patients with ICI myocarditis, whereas no significant changes in the tested biomarkers were observed in the control group. For SII, AAR, and LAR, high ratios of the biomarker levels before the last ICI to baseline was associated with the incidence of myocarditis. Conclusions Surveillance of these economical biomarkers during ICI therapy might contribute to the risk prediction of ICI myocarditis, as well as the assessment of disease severity and prognosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jingtao Ma
- Department of Cardiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
84
|
Kuang H, Yan Q, Li Z, Lin A, Li K, Zhang J, Luo P, Yin Y. Comprehensive analysis of VEGF/VEGFR inhibitor-induced immune-mediated hypertension: integrating pharmacovigilance, clinical data, and preclinical models. Front Immunol 2024; 15:1488853. [PMID: 39502700 PMCID: PMC11534862 DOI: 10.3389/fimmu.2024.1488853] [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: 08/30/2024] [Accepted: 09/30/2024] [Indexed: 11/08/2024] Open
Abstract
Introduction This study aimed to elucidate the differential immunological mechanisms and characteristics of hypertension induced by VEGF inhibitors (VEGFi) and VEGF receptor inhibitors (VEGFRi), with the goal of optimizing monitoring strategies and treatment protocols. Methods We investigated the risk of immune-related adverse events associated with VEGFi/VEGFRi-induced hypertension by analyzing the FDA Adverse Event Reporting System (FAERS) database. Findings were corroborated with blood pressure characteristics observed in clinical patients and preclinical models exposed to various VEGF/VEGFRi. Clinical and preclinical studies were conducted to compare immunological responses and hypertension profiles between inhibitor classes. An integrative analysis across cancer types and species was performed, focusing on key signaling pathways. Results Analysis of FAERS data, in conjunction with clinical observations, revealed that both VEGFi and VEGFRi significantly elevated the risk of immune-mediated, blood pressure-related adverse events (ROR=7.75, 95% CI: 7.76-7.95). Subsequent clinical and preclinical studies demonstrated differential immunological responses and hypertension profiles between inhibitor classes. VEGFRi exhibited a more rapid onset, greater blood pressure elevation, and higher incidence of immune-mediated adverse events compared to VEGFi (Systolic BP: ROR=0 for VEGFi vs. ROR=12.25, 95% CI: 6.54-22.96 for VEGFRi; Diastolic BP: ROR=5.09, 95% CI: 0.60-43.61 for VEGFi vs. ROR=12.90, 95% CI: 3.73-44.55 for VEGFRi). Integrative analysis across cancer types and species, focusing on key signaling pathways, revealed that VEGF/VEGFRi-induced blood pressure elevation was associated with immunomodulation of the mitogen activated protein kinase (MAPK) pathway (R=-0.379, P=0.0435), alterations in triglyceride metabolism (R=-0.664, P=0.0001), modulation of myo-inositol 1,4,5-trisphosphate-sensitive calcium release channel activity (R=0.389, P=0.0378), and dysregulation of nitric oxide eNOS activation and metabolism (R=-0.439, P=0.0179). Discussion The temporal dynamics of these effects demonstrated greater significance than dose-dependent responses. Both VEGFi and VEGFRi significantly augmented the risk of immune-mediated, blood pressure-related adverse events, with VEGFRi inducing a more rapid and pronounced onset of blood pressure elevation and a higher incidence of immune-related, blood pressure-associated adverse events compared to VEGFi.
Collapse
Affiliation(s)
- Hongyu Kuang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qingkai Yan
- Department of Cardiology, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Zhanzhi Li
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Kailai Li
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuehui Yin
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
85
|
Liu S, Horowitz JD, Koczwara B, Sverdlov AL, Packer N, Clark RA. Cardiac events among a cohort of 17,389 patients receiving cancer chemotherapy: short and long term implications. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:72. [PMID: 39415228 PMCID: PMC11481733 DOI: 10.1186/s40959-024-00269-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 09/25/2024] [Indexed: 10/18/2024]
Abstract
BACKGROUND The association between cardiovascular disease and carcinogenesis is bidirectional and well-established. Furthermore, cancer treatment improves overall patient survival, potentially at the cost of incremental and fatal cardiovascular disease (CVD). AIM To evaluate (a) In a real-world cohort, the proportion of patients offered cancer chemotherapy who have antecedent CVD (CVDA); (b) The rates of patient admission with subsequent development of CVD (CVDS) requiring hospital admission post assignment to chemotherapy; (c) The impact of CVDA and CVDS on mortality rates relative to those seen in patients without overt CVD (CVD-) and (d) The time course of mortality in CVD- versus CVDS patients. METHODS Retrospective analysis was performed in deidentified linked health data sets. Correlates of mortality were evaluated by Cox proportional hazards evaluation. Relative and absolute time-variability of CVD as a primary cause of death were determined. RESULTS Of the total 17,389 patients, there were 2,159 with CVDA. Over a median follow-up time of 4.6 years, CVDS admissions (n = 8,529) occurred more commonly in the presence of CVDA (70.0% vs. 46.1%, p < 0.001), and more than 50% of CVDS cases occurred in the first 12 months of follow-up. The 5-year mortality rates were 71.5% for CVDA, 64.7% for CVDS, and 40.8% for CVD- (p < 0.001). Development of CVDS was associated with a substantially increased risk of mortality in the next 12 months. The development of CVDs was also associated with an increased risk of cardiovascular, as against non-cardiovascular, mortality (7.1% vs. 1.6%, p < 0.001). CONCLUSIONS Approximately 50% of patients assigned to cancer chemotherapy developed CVDS, heralding a particularly high risk of mortality over the next 12 months. Both CVDA and CVDS are associated with substantial increases in mortality rates relative to those in CVD- patients. This increased risk merits close individual monitoring.
Collapse
Affiliation(s)
- Saifei Liu
- Cardiovascular Pathophysiology & Therapeutics Group, The Basil Hetzel Institute for Translational Research, University of Adelaide, Adelaide, SA, Australia.
- Caring Futures Institute, Flinders University, Southern Adelaide Local Health Network, Adelaide, SA, Australia.
| | - John D Horowitz
- Cardiovascular Pathophysiology & Therapeutics Group, The Basil Hetzel Institute for Translational Research, University of Adelaide, Adelaide, SA, Australia
| | - Bogda Koczwara
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- Southern Adelaide Local Health Network, Adelaide, SA, Australia
| | - Aaron L Sverdlov
- Newcastle Centre of Excellence in Cardio-Oncology: Calvary Mater Newcastle, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
- Cardiovascular Department, John Hunter Hospital, Newcastle, NSW, Australia
| | - Natalie Packer
- Southern Adelaide Local Health Network, Adelaide, SA, Australia
| | - Robyn A Clark
- Caring Futures Institute, Flinders University, Southern Adelaide Local Health Network, Adelaide, SA, Australia
- Southern Adelaide Local Health Network, Adelaide, SA, Australia
| |
Collapse
|
86
|
Napiórkowska-Baran K, Doligalska A, Drozd M, Czarnowska M, Łaszczych D, Dolina M, Szymczak B, Schmidt O, Bartuzi Z. Management of a Patient with Cardiovascular Disease Should Include Assessment of Primary and Secondary Immunodeficiencies: Part 2-Secondary Immunodeficiencies. Healthcare (Basel) 2024; 12:1977. [PMID: 39408157 PMCID: PMC11477378 DOI: 10.3390/healthcare12191977] [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: 08/16/2024] [Revised: 09/19/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Cardiovascular diseases are among the most common chronic diseases, generating high social and economic costs. Secondary immunodeficiencies occur more often than primary ones and may result from the co-occurrence of specific diseases, treatment, nutrient deficiencies and non-nutritive bio-active compounds that result from the industrial nutrient practices. OBJECTIVES The aim of this article is to present selected secondary immunodeficiencies and their impact on the cardiovascular system. RESULTS The treatment of a patient with cardiovascular disease should include an assess-ment for immunodeficiencies, because the immune and cardiovascular systems are closely linked. CONCLUSIONS Immune system dysfunctions can significantly affect the course of cardiovascular diseases and their treatment. For this reason, comprehensive care for a patient with cardiovascular disease requires taking into account potential immunodeficiencies, which can have a significant impact on the patient's health.
Collapse
Affiliation(s)
- Katarzyna Napiórkowska-Baran
- Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland;
| | - Agata Doligalska
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland; (A.D.); (M.D.); (M.C.); (D.Ł.); (M.D.); (B.S.); (O.S.)
| | - Magdalena Drozd
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland; (A.D.); (M.D.); (M.C.); (D.Ł.); (M.D.); (B.S.); (O.S.)
| | - Marta Czarnowska
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland; (A.D.); (M.D.); (M.C.); (D.Ł.); (M.D.); (B.S.); (O.S.)
| | - Dariusz Łaszczych
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland; (A.D.); (M.D.); (M.C.); (D.Ł.); (M.D.); (B.S.); (O.S.)
| | - Marcin Dolina
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland; (A.D.); (M.D.); (M.C.); (D.Ł.); (M.D.); (B.S.); (O.S.)
| | - Bartłomiej Szymczak
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland; (A.D.); (M.D.); (M.C.); (D.Ł.); (M.D.); (B.S.); (O.S.)
| | - Oskar Schmidt
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland; (A.D.); (M.D.); (M.C.); (D.Ł.); (M.D.); (B.S.); (O.S.)
| | - Zbigniew Bartuzi
- Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland;
| |
Collapse
|
87
|
Bender JD, Pillay-Smiley N, Hill GD, de Blank P, Hummel TR, Weiss BD, Kumar A, Zang H, Ollberding NJ, Ryan TD. Cardiac Dysfunction in Children and Young Adults Treated With MEK Inhibitors: A Retrospective, Single-Center Study. JACC CardioOncol 2024; 6:794-796. [PMID: 39479314 PMCID: PMC11520206 DOI: 10.1016/j.jaccao.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 06/10/2024] [Accepted: 07/01/2024] [Indexed: 11/02/2024] Open
Affiliation(s)
- Jonathan D. Bender
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Natasha Pillay-Smiley
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Garick D. Hill
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Peter de Blank
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Trent R. Hummel
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Brian D. Weiss
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Ashish Kumar
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Bone Marrow Transplantation and Immune Deficiency, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Huaiyu Zang
- Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Nicholas J. Ollberding
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Biostatistics and Epidemiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Thomas D. Ryan
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
88
|
Keramida K, Farmakis D, Rakisheva A, Tocchetti CG, Ameri P, Asteggiano R, Barac A, Bax J, Bayes-Genis A, Bergler Klein J, Bucciarelli-Ducci C, Celutkiene J, Coats AJS, Cohen Solal A, Dent S, Filippatos G, Ghosh A, Hermann J, Koop Y, Lenihan D, Lopez Fernandez T, Lyon AR, Mercurio V, Moura B, Piepoli M, Sener YZ, Suter T, Sverdlov AL, Tadic M, Thum T, van der Meer P, van Linthout S, Metra M, Rosano G. The right heart in patients with cancer. A scientific statement of the Heart Failure Association (HFA) of the ESC and the ESC Council of Cardio-Oncology. Eur J Heart Fail 2024; 26:2077-2093. [PMID: 39193837 DOI: 10.1002/ejhf.3412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 07/24/2024] [Accepted: 07/30/2024] [Indexed: 08/29/2024] Open
Affiliation(s)
- Kalliopi Keramida
- Cardiology Department, General Anti-Cancer Oncological Hospital Agios Savvas, Athens, Greece
| | - Dimitrios Farmakis
- Department of Cardiology, Athens University Hospital Attikon, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Amina Rakisheva
- City Cardiological Center, Almaty, Kazakhstan, Qonaev City Hospital, Almaty Region, Almaty, Kazakhstan
| | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences (DISMET), 'Federico II' University, Naples, Italy
- Interdepartmental Center of Clinical and Translational Sciences (CIRCET), 'Federico II' University, Naples, Italy
- Interdepartmental Hypertension Research Center (CIRIAPA), 'Federico II' University, Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), 'Federico II' University, Naples, Italy
| | - Pietro Ameri
- Department of Internal Medicine, University of Genova, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Riccardo Asteggiano
- Internal Medicine, Department of Medicine and Surgery, University of Insubria, Varese, Italy
- LARC, Laboratorio Analisi e Ricerca Clinica, Turin, Italy
| | - Ana Barac
- MedStar Heart and Vascular Institute, Georgetown University, Washington, DC, USA
| | - Jeroen Bax
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Antoni Bayes-Genis
- CIBER Cardiovascular, Madrid, Spain
- Institut del Cor, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | | | | | - Jelena Celutkiene
- Centre of Cardiology and Angiology, Clinic of Cardiac and Vascular Diseases, Faculty of Medicine, Institute of Clinical Medicine, Vilnius University, Vilnius, Lithuania
| | | | - Alain Cohen Solal
- Université Paris Cité, INSERM U-942 MASCOT, Cardiology Department, Lariboisière Hospital, Paris, France
| | - Susan Dent
- Duke Cancer Institute, Department of Medicine, Duke University, Durham, NC, USA
| | - Gerasimos Filippatos
- Department of Cardiology, Athens University Hospital Attikon, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Arjun Ghosh
- Barts Heart Centre, University College London Hospital and Hatter Cardiovascular Institute, London, UK
| | - Joerg Hermann
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Yvonne Koop
- Department of Cardiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Daniel Lenihan
- Cardio-Oncology Program, St Francis Healthcare, Cape Girardeau, MO, USA
| | - Teresa Lopez Fernandez
- Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, Madrid, Spain
- Cardiology Department, Hospital Universitario Quirónsalud Madrid, Madrid, Spain
| | - Alexander R Lyon
- National Heart and Lung Institute, Imperial College London and Cardio-Oncology Service, Royal Brompton Hospital, London, UK
| | - Valentina Mercurio
- Department of Translational Medical Sciences (DISMET), 'Federico II' University, Naples, Italy
- Interdepartmental Center of Clinical and Translational Sciences (CIRCET), 'Federico II' University, Naples, Italy
- Interdepartmental Hypertension Research Center (CIRIAPA), 'Federico II' University, Naples, Italy
| | - Brenda Moura
- Armed Forces Hospital, Porto and Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Massimo Piepoli
- Clinical Cardiology, IRCCS Policlinico San Donato, Milan, Italy
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | | | - Thomas Suter
- Cardiology Department, University of Ulm, Ulm, Germany
| | - Aaron L Sverdlov
- Newcastle Centre of Excellence in Cardio-Oncology, The University of Newcastle, Hunter Medical Research Institute, Calvary Mater Newcastle, Hunter New England Health, Newcastle, NSW, Australia
- Cardiovascular Department, John Hunter Hospital, New Lambton Heights, NSW, Australia
| | - Marijana Tadic
- Klinik für Innere Medizin II, Universitätsklinikum Ulm, Ulm, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Hannover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| | - Peter van der Meer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Sophie van Linthout
- Berlin Institute of Health (BIH) at Charité, BIH Center for Regenerative Therapies, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Marco Metra
- Institute of Cardiology, ASST Spedali Civili, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | | |
Collapse
|
89
|
Nobre Menezes M, Tavares da Silva M, Magalhães A, Melica B, Cristina Toste J, Calé R, Almeida M, Fiuza M, Infante de Oliveira E. Response to the letter "Cardio-oncology guidelines, structural heart disease and Kounis syndrome in the upcoming guidelines". Rev Port Cardiol 2024; 43:585-586. [PMID: 39002720 DOI: 10.1016/j.repc.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/15/2024] Open
Affiliation(s)
- Miguel Nobre Menezes
- Unidade de Cardiologia de Intervenção Joaquim Oliveira, Serviço de Cardiologia, Departamento de Coração e Vasos, Centro Hospitalar Universitário Lisboa Norte, Hospital de Santa Maria, Portugal; Centro Cardiovascular da Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Faculdade de Medicina da Universidade de Lisboa, Portugal; Grupo de Estudos de Cardio-Oncologia, Sociedade Portuguesa de Cardiologia, Portugal; Associação Portuguesa de Intervenção Cardiovascular, Sociedade Portuguesa de Cardiologia, Portugal.
| | - Marta Tavares da Silva
- Serviço de Cardiologia, Centro Hospitalar e Universitário de São João, Porto, Portugal; UnIC@RISE, Departamento de Cirurgia e Fisiologia, Faculdade de Medicina da Universidade do Porto, Porto, Portugal; Grupo de Estudos de Cardio-Oncologia, Sociedade Portuguesa de Cardiologia, Portugal; Associação Portuguesa de Intervenção Cardiovascular, Sociedade Portuguesa de Cardiologia, Portugal
| | - Andreia Magalhães
- Centro Cardiovascular da Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Faculdade de Medicina da Universidade de Lisboa, Portugal; Serviço de Cardiologia, Departamento de Coração e Vasos, Centro Hospitalar Universitário Lisboa Norte, Hospital de Santa Maria, Portugal; Grupo de Estudos de Cardio-Oncologia, Sociedade Portuguesa de Cardiologia, Portugal
| | - Bruno Melica
- Cardiology Department, Vila Nova de Gaia Hospital, Vila Nova de Gaia, Portugal; Associação Portuguesa de Intervenção Cardiovascular, Sociedade Portuguesa de Cardiologia, Portugal
| | - Júlia Cristina Toste
- Department of Cardiology, Hospital da Luz, Lisbon, Portugal; NOVA Medical School, Lisbon, Portugal; Grupo de Estudos de Cardio-Oncologia, Sociedade Portuguesa de Cardiologia, Portugal
| | - Rita Calé
- Cardiology Department, Hospital Garcia de Orta EPE, Almada, Portugal; Associação Portuguesa de Intervenção Cardiovascular, Sociedade Portuguesa de Cardiologia, Portugal
| | - Manuel Almeida
- Unidade de Intervenção Cardiovascular I Hospital de Santa Cruz, Centro Hospitalar de Lisboa Ocidental, Portugal; CHRC NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisboa, Portugal; Associação Portuguesa de Intervenção Cardiovascular, Sociedade Portuguesa de Cardiologia, Portugal
| | - Manuela Fiuza
- Centro Cardiovascular da Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Faculdade de Medicina da Universidade de Lisboa, Portugal; Serviço de Cardiologia, Departamento de Coração e Vasos, Centro Hospitalar Universitário Lisboa Norte, Hospital de Santa Maria, Portugal; Grupo de Estudos de Cardio-Oncologia, Sociedade Portuguesa de Cardiologia, Portugal
| | - Eduardo Infante de Oliveira
- Unidade de Intervenção Cardiovascular I Hospital de Santa Cruz, Centro Hospitalar de Lisboa Ocidental, Portugal; Associação Portuguesa de Intervenção Cardiovascular, Sociedade Portuguesa de Cardiologia, Portugal; Hospital Lusíadas Lisboa, Portugal
| |
Collapse
|
90
|
Osataphan N, Abdel-Qadir H, Zebrowska AM, Borowiec A. Sodium-Glucose Cotransporter 2 Inhibitors During Cancer Therapy: Benefits, Risks, and Ongoing Clinical Trials. Curr Oncol Rep 2024; 26:1188-1196. [PMID: 38990501 PMCID: PMC11480197 DOI: 10.1007/s11912-024-01577-8] [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: 07/02/2024] [Indexed: 07/12/2024]
Abstract
PURPOSE OF REVIEW The goal of this paper is to summarize the data pertaining to the use of sodium-glucose cotransporter-2 inhibitors (SGLT-2i) for the prevention of cardiotoxicity in patients receiving anthracyclines for cancer treatment. We discuss the potential efficacy of this class of medications, incorporating insights from existing literature and ongoing studies. RECENT FINDINGS SGLT2i are a class of medications which were initially developed for treatment of Type 2 diabetes and later extended to treat heart failure with reduced and preserved ejection fraction regardless of diabetes status. There remains a need for effective and safe treatments to preventing cardiotoxicity in anthracycline-treated patients. It has been proposed that SGLT2i may provide protection against the cardiotoxic effects of anthracyclines. Some of the proposed mechanisms include beneficial metabolic, neurohormonal, and hemodynamic effects, renal protection, as well as a decrease in inflammation, oxidative stress, apoptosis, mitochondrial dysfunction and ion homeostasis. There is emerging evidence from basic science and observational studies that SGLT2i may play a role in the prevention of chemotherapy-induced cardiotoxicity. Randomized controlled trials are needed to conclusively determine the role of SGLT2 inhibitors as a cardioprotective therapy in patients receiving anthracyclines for the treatment of cancer.
Collapse
Affiliation(s)
- Nichanan Osataphan
- Division of Cardiology, Ted Rogers Program in Cardiotoxicity Prevention, Peter Munk Cardiac Center, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON, Canada
- Division of Cardiology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Husam Abdel-Qadir
- Division of Cardiology, Ted Rogers Program in Cardiotoxicity Prevention, Peter Munk Cardiac Center, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON, Canada
- Women's College Hospital, Toronto, ON, Canada
| | - Agnieszka Maria Zebrowska
- Department of Cancer & Cardio-Oncology Diagnostics, The Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena Str 5, 02-781, Warsaw, Poland
- Unit for Screening Studies in Inherited Cardiovascular Diseases, The Cardinal Stefan Wyszynski National Institute of Cardiology, Warsaw, Poland
| | - Anna Borowiec
- Department of Cancer & Cardio-Oncology Diagnostics, The Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena Str 5, 02-781, Warsaw, Poland.
| |
Collapse
|
91
|
Raisi-Estabragh Z, Murphy AC, Ramalingam S, Scherrer-Crosbie M, Lopez-Fernandez T, Reynolds KL, Aznar M, Lin AE, Libby P, Cordoba R, Bredsen-Masley C, Wechalekar A, Apperley J, Cheng RK, Manisty CH. Cardiovascular Considerations Before Cancer Therapy: Gaps in Evidence and JACC: CardioOncology Expert Panel Recommendations. JACC CardioOncol 2024; 6:631-654. [PMID: 39479317 PMCID: PMC11520216 DOI: 10.1016/j.jaccao.2024.07.017] [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: 09/11/2023] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 11/02/2024] Open
Abstract
Baseline cardiovascular assessment before the initiation of potentially cardiotoxic cancer therapies is a key component of cardio-oncology, aiming to reduce cardiovascular complications and morbidity in patients and survivors. Recent clinical practice guidelines provide both general and cancer therapy-specific recommendations for baseline cardiovascular toxicity risk assessment and management, including the use of dedicated risk scores, cardiovascular imaging, and biomarker testing. However, the value of such interventions in altering disease trajectories has not been established, with many recommendations based on expert opinion or Level of Evidence: C, studies with a potential for high risk of bias. Advances in understanding underlying mechanisms of cardiotoxicity and the increased availability of genetic and immunologic profiling present new opportunities for personalized risk assessment. This paper evaluates the existing evidence on cardiovascular care of cancer patients before cardiotoxic cancer therapy and highlights gaps in evidence and priorities for future research.
Collapse
Affiliation(s)
- Zahra Raisi-Estabragh
- William Harvey Research Institute, National Institute for Health and Care Research Barts Biomedical Research Centre, Queen Mary University of London, London, United Kingdom
- Barts Heart Centre, St Bartholomew’s Hospital, Barts Health NHS Trust, West Smithfield, London, United Kindgom
| | | | - Sivatharshini Ramalingam
- Cardio-Oncology Service, Royal Brompton Hospital, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Marielle Scherrer-Crosbie
- Division of Cardiology, The Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Teresa Lopez-Fernandez
- Hospital Universitario La Paz, Instituto de Investigación La Paz-IdiPaz, Madrid, Spain
- Hospital Universitario Quiron Pozuelo, Madrid, Spain
| | - Kerry L. Reynolds
- Division of Oncology and Hematology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Marianne Aznar
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Amy E. Lin
- Department of Medicine, Division of Cardiology, Section of Cardio-Oncology and Immunology, University of California-San Francisco, San Francisco, California, USA
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Raul Cordoba
- Fundacion Jimenez Diaz University Hospital, Health Research Institute Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Madrid, Spain
| | - Christine Bredsen-Masley
- Division of Hematology/Oncology, Department of Medicine, St. Michael’s Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Ashu Wechalekar
- Department of Haematology, University College London Hospitals NHS Trust, London, United Kingdom
| | - Jane Apperley
- Department of Clinical Haematology, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
- Centre for Haematology, Imperial College London, London, United Kingdom
| | - Richard K. Cheng
- Division of Cardiology, University of Washington Medical Center, Seattle, Washington, USA
| | - Charlotte H. Manisty
- Barts Heart Centre, St Bartholomew’s Hospital, Barts Health NHS Trust, West Smithfield, London, United Kindgom
- Institute of Cardiovascular Science, University College London, London, United Kingdom
| |
Collapse
|
92
|
Camilli M, Cipolla CM, Dent S, Minotti G, Cardinale DM. Anthracycline Cardiotoxicity in Adult Cancer Patients: JACC: CardioOncology State-of-the-Art Review. JACC CardioOncol 2024; 6:655-677. [PMID: 39479333 PMCID: PMC11520218 DOI: 10.1016/j.jaccao.2024.07.016] [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: 03/26/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 11/02/2024] Open
Abstract
Since their introduction in the 1960s, anthracyclines have been a significant breakthrough in oncology, introducing dramatic changes in the treatment of solid and hematologic malignancies. Although new-generation targeted drugs and cellular therapies are revolutionizing contemporary oncology, anthracyclines remain the cornerstone of treatment for lymphomas, acute leukemias, and soft tissue sarcomas. However, their clinical application is limited by a dose-dependent cardiotoxicity that can reduce cardiac performance and eventually lead to overt heart failure. The field of cardio-oncology has emerged to safeguard the cardiovascular health of cancer patients receiving these therapies. It focuses on controlling risk factors, implementing preventive strategies, ensuring appropriate surveillance, and managing complications. This state-of-the-art review summarizes the current indications for anthracyclines in modern oncology, explores recent evidence on pathophysiology and epidemiology, and discusses advances in cardioprotection measures in the anthracycline-treated patient. Additionally, it highlights key clinical challenges and research gaps in this area.
Collapse
Affiliation(s)
- Massimiliano Camilli
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Carlo Maria Cipolla
- Cardioncology Unit, Cardioncology and Second Opinion Division, European Institute of Oncology IRCCS, Milan, Italy
| | - Susan Dent
- Duke Cancer Institute, Duke University, Durham, North Carolina, USA
- Division of Medical Oncology, Duke Cancer Institute, Duke University, Durham, North Carolina, USA
| | - Giorgio Minotti
- Università e Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Daniela Maria Cardinale
- Cardioncology Unit, Cardioncology and Second Opinion Division, European Institute of Oncology IRCCS, Milan, Italy
| |
Collapse
|
93
|
Tocchetti CG, Farmakis D, Koop Y, Andres MS, Couch LS, Formisano L, Ciardiello F, Pane F, Au L, Emmerich M, Plummer C, Gulati G, Ramalingam S, Cardinale D, Brezden-Masley C, Iakobishvili Z, Thavendiranathan P, Santoro C, Bergler-Klein J, Keramida K, de Boer RA, Maack C, Lutgens E, Rassaf T, Fradley MG, Moslehi J, Yang EH, De Keulenaer G, Ameri P, Bax J, Neilan TG, Herrmann J, Mbakwem AC, Mirabel M, Skouri H, Hirsch E, Cohen-Solal A, Sverdlov AL, van der Meer P, Asteggiano R, Barac A, Ky B, Lenihan D, Dent S, Seferovic P, Coats AJS, Metra M, Rosano G, Suter T, Lopez-Fernandez T, Lyon AR. Cardiovascular toxicities of immune therapies for cancer - a scientific statement of the Heart Failure Association (HFA) of the ESC and the ESC Council of Cardio-Oncology. Eur J Heart Fail 2024; 26:2055-2076. [PMID: 39087551 DOI: 10.1002/ejhf.3340] [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: 11/12/2023] [Revised: 05/22/2024] [Accepted: 06/03/2024] [Indexed: 08/02/2024] Open
Abstract
The advent of immunological therapies has revolutionized the treatment of solid and haematological cancers over the last decade. Licensed therapies which activate the immune system to target cancer cells can be broadly divided into two classes. The first class are antibodies that inhibit immune checkpoint signalling, known as immune checkpoint inhibitors (ICIs). The second class are cell-based immune therapies including chimeric antigen receptor T lymphocyte (CAR-T) cell therapies, natural killer (NK) cell therapies, and tumour infiltrating lymphocyte (TIL) therapies. The clinical efficacy of all these treatments generally outweighs the risks, but there is a high rate of immune-related adverse events (irAEs), which are often unpredictable in timing with clinical sequalae ranging from mild (e.g. rash) to severe or even fatal (e.g. myocarditis, cytokine release syndrome) and reversible to permanent (e.g. endocrinopathies).The mechanisms underpinning irAE pathology vary across different irAE complications and syndromes, reflecting the broad clinical phenotypes observed and the variability of different individual immune responses, and are poorly understood overall. Immune-related cardiovascular toxicities have emerged, and our understanding has evolved from focussing initially on rare but fatal ICI-related myocarditis with cardiogenic shock to more common complications including less severe ICI-related myocarditis, pericarditis, arrhythmias, including conduction system disease and heart block, non-inflammatory heart failure, takotsubo syndrome and coronary artery disease. In this scientific statement on the cardiovascular toxicities of immune therapies for cancer, we summarize the pathophysiology, epidemiology, diagnosis, and management of ICI, CAR-T, NK, and TIL therapies. We also highlight gaps in the literature and where future research should focus.
Collapse
Affiliation(s)
- Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences (DISMET), Center for Basic and Clinical Immunology Research (CISI), Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Naples, Italy
| | - Dimitrios Farmakis
- Department of Cardiology, Attikon University Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Yvonne Koop
- Department of Cardiovascular Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Dutch Heart Foundation, The Hague, The Netherlands
| | - Maria Sol Andres
- Royal Brompton Hospital, Part of Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Liam S Couch
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Luigi Formisano
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Fortunato Ciardiello
- Department of Precision Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Fabrizio Pane
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Lewis Au
- Skin and Renal Unit, Royal Marsden NHS Foundation Trust, London, UK
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Max Emmerich
- The Francis Crick Institute, London, UK
- The Royal Marsden Hospital, London, UK
- St. John's Institute of Dermatology, Guy's and St Thomas' Hospital, London, UK
| | - Chris Plummer
- Department of Cardiology, Freeman Hospital, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Geeta Gulati
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Cardiology, Division of Medicine, Oslo University Hospital, Ullevål, Oslo, Norway
- Division of Research and Innovation, Akershus University Hospital, Lørenskog, Norway
| | - Sivatharshini Ramalingam
- Royal Brompton Hospital, Part of Guy's and St Thomas' NHS Foundation Trust, London, UK
- Oxted Health Centre, Oxted, UK
| | - Daniela Cardinale
- CardioOncology Unit, European Institute of Oncology, IRCCS, Milan, Italy
| | | | - Zaza Iakobishvili
- Department of Community Cardiology, Tel Aviv Jaffa District, Clalit Health Services, Tel Aviv, Israel
- Department of Cardiology, Assuta Ashdod University Hospital, Ashdod, Israel
- Faculty of Health Sciences, Ben Gurion University of the Negev, Be'er Sheva, Israel
| | - Paaladinesh Thavendiranathan
- Ted Rogers Program in Cardiotoxicity Prevention, Peter Munk Cardiac Center, Toronto General Hospital, University of Toronto, Toronto, ON, Canada
| | - Ciro Santoro
- Department of Advanced Biomedical Science, Federico II University Hospital, Naples, Italy
| | | | - Kalliopi Keramida
- Cardiology Department, General Anti-Cancer, Oncological Hospital, Agios Savvas, Athens, Greece
| | - Rudolf A de Boer
- Department of Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany
- Medical Clinic 1, University Clinic Würzburg, Würzburg, Germany
| | - Esther Lutgens
- Department of Cardiovascular Medicine and Immunology, Mayo Clinic, Rochester, MN, USA
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Essen, Germany
| | - Michael G Fradley
- Cardio-Oncology Center of Excellence, Division of Cardiology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Javid Moslehi
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Eric H Yang
- UCLA Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Gilles De Keulenaer
- Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Pietro Ameri
- Department of Internal Medicine, University of Genova, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Jeroen Bax
- Department of Cardiology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Tomas G Neilan
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Joerg Herrmann
- Cardio-Oncology Clinic, Division of Preventive Cardiology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Amam C Mbakwem
- College of Medicine, University of Lagos/Lagos University Teaching Hospital Idi Araba, Lagos, Nigeria
| | | | - Hadi Skouri
- Cardiology Division, Sheikh Shakhbout Medical City, Khalifa University, Abu Dhabi, UAE
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Alain Cohen-Solal
- Paris Cité University INSERM U 948 MASCOT Research Unit Cardiology, Lariboisere Universitaire Hospital, AP-HP, Paris, France
| | - Aaron L Sverdlov
- Newcastle Centre of Excellence in Cardio-Oncology, University of Newcastle, Hunter Medical Research Institute, Calvary Mater Newcastle, Hunter New England Health, Newcastle, NSW, Australia
- Cardiovascular Department, John Hunter Hospital, New Lambton Heights, NSW, Australia
| | - Peter van der Meer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Riccardo Asteggiano
- Internal Medicine, Department of Medicine and Surgery, University of Insubria, Varese, Italy
- LARC, Laboratorio Analisi e Ricerca Clinica, Turin, Italy
| | - Ana Barac
- Inova Schar Heart and Vascular Institute, Falls Church, VA, USA
| | - Bonnie Ky
- Division of Cardiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel Lenihan
- Cape Cardiology, St Francis Healthcare, Cape Girardeau, MO, USA
| | - Susan Dent
- Duke Cancer Institute, Department of Medicine, Duke University, Durham, NC, USA
| | - Petar Seferovic
- University Medical Center, Medical Faculty University of Belgrade, Serbian Academy of Sciences and Arts, Belgrade, Serbia
| | | | - Marco Metra
- Cardiology, ASST Spedali Civili, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Giuseppe Rosano
- Department of Human Sciences and Promotion of Quality of Life, San Raffaele Open University of Rome, Rome, Italy
- Cardiology, San Raffaele Cassino Hospital, Cassino, FR, Italy
| | - Thomas Suter
- Bern University Hospital, University of Bern, Bern, Switzerland
| | - Teresa Lopez-Fernandez
- Cardio-Oncology Unit, Cardiology Department, IdiPAZ Research Institute, La Paz University Hospital, Madrid, Spain
- Cardiology Department, Quironsalud Madrid University Hospital, Madrid, Spain
| | - Alexander R Lyon
- Cardio-Oncology Service, Royal Brompton Hospital and National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
94
|
Nikolova AP, Ky B. Immune Checkpoint Inhibitors Myocarditis: The Nuts and Bolts. JACC. HEART FAILURE 2024; 12:1794-1796. [PMID: 39207325 DOI: 10.1016/j.jchf.2024.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 09/04/2024]
Affiliation(s)
| | - Bonnie Ky
- University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
95
|
Barac A, Wadlow RC, Deeken JF, deFilippi C. Cardiac Troponin I and T in ICI Myocarditis Screening, Diagnosis, and Prognosis. JACC CardioOncol 2024; 6:804-807. [PMID: 39479330 PMCID: PMC11520212 DOI: 10.1016/j.jaccao.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2024] Open
Affiliation(s)
- Ana Barac
- Inova Schar Heart and Vascular, Fairfax, Virginia, USA
- Inova Schar Cancer, Fairfax, Virginia, USA
| | | | | | | |
Collapse
|
96
|
Bloom M, Alvarez-Cardona JA, Ganatra S, Barac A, Pusic I, Lenihan D, Dent S. How to utilize current guidelines to manage patients with cancer at high risk for heart failure. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:63. [PMID: 39342407 PMCID: PMC11438295 DOI: 10.1186/s40959-024-00259-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/19/2024] [Indexed: 10/01/2024]
Abstract
Heart failure (HF) in patients with cancer is associated with high morbidity and mortality. The success of cancer therapy has resulted in an exponential rise in the population of cancer survivors, however cardiovascular disease (CVD) is now a major life limiting condition more than 5 years after cancer diagnosis [Sturgeon, Deng, Bluethmann, et al 40(48):3889-3897, 2019]. Prevention and early detection of CVD, including cardiomyopathy (CM) and HF is of paramount importance. The European Society of Cardiology (ESC) published guidelines on Cardio-Oncology (CO) [Lyon, López-Fernández, Couch, et al 43(41):4229-4361, 2022] detailing cardiovascular (CV) risk stratification, prevention, monitoring, diagnosis, and treatment throughout the course and following completion of cancer therapy. Here we utilize a case to summarize aspects of the ESC guideline relevant to HF clinicians, with a focus on risk stratification, early detection, prevention of CM and HF, and the role for guideline directed medical therapy in patients with cancer.
Collapse
Affiliation(s)
- Michelle Bloom
- Leon H. Charney Division of Cardiology, New York University School of Medicine, NYU Langone Health, New York, NY, USA
| | - Jose A Alvarez-Cardona
- Leon H. Charney Division of Cardiology, New York University School of Medicine, NYU Langone Health, New York, NY, USA
| | - Sarju Ganatra
- Division of Cardiovascular Medicine, Lahey Hospital and Medical Center, Department of Medicine, Beth Israel Lahey Health, Burlington, MA, USA
| | - Ana Barac
- Inova Schar Heart and Vascular and Inova Inova Schar Cancer, Falls Church, VA, USA
| | - Iskra Pusic
- Division of Oncology, Department of Medicine, Washington University, St. Louis, MO, USA
| | - Daniel Lenihan
- International Cardio-Oncology Society, FL and Cape Cardiology Group, Cape Girardeau, Tampa, MO, USA
| | - Susan Dent
- Duke Cancer Institute, Department of Medicine, Duke University, DUMC 3446, Durham, NC, 27710, USA.
| |
Collapse
|
97
|
Borowiec A, Ozdowska P, Rosinska M, Zebrowska AM, Jagiello-Gruszfeld A, Jasek S, Waniewska J, Kotowicz B, Kosela-Paterczyk H, Lampka E, Pogoda K, Cieszanowski A, Nowecki Z, Walewski J. Coronary artery calcium score and other risk factors in patients at moderate and high risk of cancer therapy-related cardiovascular toxicity. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:64. [PMID: 39342402 PMCID: PMC11437645 DOI: 10.1186/s40959-024-00266-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 09/16/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND The presence and burden of coronary artery calcium (CAC) is a strong predictor of cardiovascular events. Current guidelines of the European Society of Cardiology (ESC) for cardio-oncology do not recommend the use of the CAC score to determine the status of risk in cancer patients. The aim of this study is to evaluate the presence and burden of CAC on cardiac tomography and the distribution of the cardiovascular toxicity risk factors in patients with moderate and high baseline risk of cancer therapy-related cardiovascular toxicity. METHODS The study prospectively included cancer patients, diagnosed and qualified for systemic treatment with anthracycline chemotherapy. Clinical data and blood samples were collected from all patients. Additionally, the echocardiography and coronary computed tomography (CCTA) with the calculation of the coronary artery calcium (CAC) score were performed. RESULTS A total of 80 patients (mean age 60.5 years, 75 female) were included in the study. The majority of patients (62, 77.5%) had breast cancer, 11 (13.8%) were diagnosed with sarcoma, and 7 (8.8%) with lymphoma. There were 42 (52.5%) patients classified as having moderate (MR) and 38 (47.5%) as having high risk (HR) of cancer therapy-related cardiovascular toxicity according to current ESC guidelines. In comparison with moderate risk, high risk patients were older and more likely to have hypertension, hyperlipidaemia and chronic kidney disease. The mean coronary artery calcium score was significantly higher in the HR group (150.4 vs. 24.8; p = 0.000). Furthermore, cardiac biomarkers were also higher in high-risk patients (p = 0.000). In echocardiographic parameters global longitudinal strain (GLS) was lower (p = 0.012), and diastolic dysfunction was more common in the HR group. However, the left ventricle ejection fraction (LVEF) was similar in the MR and HR groups. CONCLUSIONS In patients at high and moderate risk for cancer therapy-related cardiovascular toxicity, cardiovascular toxicity risk factors were common and more prevalent in the high-risk group. The coronary artery calcium score was also significantly higher in the high-risk group. Assessing the presence and burden of coronary artery calcium is an attractive option to assess additional cardiovascular risk in cancer patients.
Collapse
Affiliation(s)
- Anna Borowiec
- Department of Cancer & Cardio-Oncology Diagnostics, The Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland.
| | - Patrycja Ozdowska
- Department of Cancer & Cardio-Oncology Diagnostics, The Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Magdalena Rosinska
- Department of Computational Oncology, The Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Agnieszka Maria Zebrowska
- Department of Cancer & Cardio-Oncology Diagnostics, The Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
- Unit for Screening Studies in Inherited Cardiovascular Diseases, The Cardinal Stefan, Wyszynski National Institute of Cardiology, Warsaw, Poland
| | | | - Sławomir Jasek
- Department of Cancer & Cardio-Oncology Diagnostics, The Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Joanna Waniewska
- Department of Radiology I, The Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Beata Kotowicz
- Cancer Biomarker and Cytokines Laboratory Unit, The Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Hanna Kosela-Paterczyk
- Department of Soft Tissue/Bone Sarcoma and Melanoma, The Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Elzbieta Lampka
- Department of Lymphoid Malignancies, The Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Katarzyna Pogoda
- Department of Brest Cancer & Reconstructive Surgery, The Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Andrzej Cieszanowski
- Second Department of Clinical Radiology, Medical University of Warsaw, Warsaw, Poland
| | - Zbigniew Nowecki
- Department of Brest Cancer & Reconstructive Surgery, The Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Jan Walewski
- Department of Lymphoid Malignancies, The Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| |
Collapse
|
98
|
Kępski J, Szmit S, Lech-Marańda E. Time Relationship between the Occurrence of a Thromboembolic Event and the Diagnosis of Hematological Malignancies. Cancers (Basel) 2024; 16:3196. [PMID: 39335167 PMCID: PMC11430228 DOI: 10.3390/cancers16183196] [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: 08/14/2024] [Revised: 09/07/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
OBJECTIVES Venous and arterial thromboembolism (VTE/ATE) often coexist with onco-hematologic diagnosis. This study aimed to assess the time relationship between the diagnosis of VTE/ATE and blood cancers. The second aim was to identify VTE/ATE risk factors related to the type of hematology disease and cardiac history. METHODS A total of 1283 patients underwent cardio-oncology evaluation at the Institute of Hematology and Transfusion Medicine in Warsaw from March 2021 through March 2023 (2 years), and 101 (7.8%) cases were identified with VTE/ATE. RESULTS ATE compared with VTE significantly occurred more often before the diagnosis and treatment of hematologic malignancy: 33/47 (70.2%) vs. 15/54 (27.8%), p < 0.0001. The risk of a VTE episode is exceptionally high in the first months after the diagnosis of an onco-hematological disease and the initiation of anticancer treatment. The higher frequency of VTE was associated with acute myeloid leukemia (17 cases/270 patients/6.30%/p = 0.055), acute lymphocytic leukemia (7 cases/76 patients/9.21%/p = 0.025), and chronic myeloproliferative disease (7 cases/48 patients/14.58%/p = 0.0003). Only the risk of VTE was significantly increased before (OR = 6.79; 95% CI: 1.85-24.95; p = 0.004) and after diagnosis of myeloproliferative disease (OR = 3.12; 95% CI: 1.06-9.16; p = 0.04). CONCLUSIONS ATEs occur more often than VTE before a diagnosis of blood cancer. The risk of VTE is exceptionally high before and after diagnosis of chronic myeloproliferative disease.
Collapse
Affiliation(s)
- Jarosław Kępski
- Department of Cardio-Oncology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland
| | - Sebastian Szmit
- Department of Cardio-Oncology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland
| | - Ewa Lech-Marańda
- Department of Hematology, Institute of Hematology and Transfusion Medicine, 02-776 Warsaw, Poland
| |
Collapse
|
99
|
He Y, Liu X, Wang M, Ke H, Ge C. Neutrophil-to-lymphocyte ratio as a predictor of cardiovascular mortality in cancer survivors. Sci Rep 2024; 14:20980. [PMID: 39251691 PMCID: PMC11385526 DOI: 10.1038/s41598-024-72027-5] [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: 07/20/2024] [Accepted: 09/03/2024] [Indexed: 09/11/2024] Open
Abstract
This study aims to evaluate the neutrophil-to-lymphocyte ratio (NLR) as a predictive biomarker for cardiovascular mortality among cancer patients, utilizing data from the National Health and Nutrition Examination Survey (NHANES). From the NHANES dataset (2007-2018), we analyzed 4974 cancer survivors, investigating the prognostic significance of NLR for all-cause, cardiovascular, and cancer-specific mortality. Survival outcomes were analyzed using Cox regression and Kaplan-Meier methods. Optimal NLR cutoffs were identified as 2.61 for differentiating the higher NLR group from lower NLR group. Elevated NLR levels significantly correlated with increased all-cause mortality (HR 1.11, 95% CI 1.07-1.14, P < 0.001) and cardiovascular mortality (HR 1.14, 95% CI 1.08-1.21, P < 0.001) in adjusted models. Subgroup analyses revealed that age, sex, smoking status, and hypertension significantly influence NLR's association with cardiovascular mortality. Specific cancers including breast, prostate, non-melanoma skin, colon and melanoma experience increased all-cause and cardiovascular mortality in the higher NLR group compared to lower NLR group. Elevated NLR is a significant predictor of increased mortality in cancer patients, particularly for cardiovascular outcomes. These findings support that NLR acts as a pivotal prognostic tool with significant implications for clinical practice in the realm of cardio-oncology.
Collapse
Affiliation(s)
- Yan He
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Qingxiu District, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Xing Liu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Qingxiu District, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Min Wang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Qingxiu District, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Honghong Ke
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Qingxiu District, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Chenliang Ge
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Qingxiu District, Nanning, 530021, Guangxi Zhuang Autonomous Region, China.
| |
Collapse
|
100
|
Nain P, Stabellini N, Makram OM, Rast J, Yerraguntla S, Gopu G, Bhave A, Seth L, Patel V, Jiang S, Malik S, Shetewi A, Montero AJ, Cullen J, Agarwal N, Wang X, Ky B, Baldassarre LA, Weintraub NL, Harris RA, Guha A. Adverse social determinants of health elevate uncontrolled hypertension risk: a cardio-oncology prospective cohort study. JNCI Cancer Spectr 2024; 8:pkae064. [PMID: 39115393 PMCID: PMC11368120 DOI: 10.1093/jncics/pkae064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/14/2024] [Accepted: 06/25/2024] [Indexed: 09/03/2024] Open
Abstract
The role of social determinants of health (SDOH) in controlling hypertension (HTN) in cancer patients is unknown. We hypothesize that high SDOH scores correlate with uncontrolled HTN in hypertensive cancer patients. In our prospective study, patients completed the Protocol for Responding to & Assessing Patients' Assets, Risks & Experiences questionnaire. After integrating home and clinic blood pressure readings, uncontrolled HTN was defined as systolic blood pressure greater than or equal to 140 mm Hg and/or diastolic blood pressure greater than or equal to 90 mm Hg. Using Cox regression, we analyzed the impact of SDOH on HTN control, adjusting for relevant factors. The study involved 318 participants (median age 66.4, median follow-up 166 days, SDOH score 6.5 ± 3.2), with stress, educational insecurity, and social isolation as prevalent adverse SDOH. High SDOH scores led to 77% increased risk of uncontrolled HTN (adjusted hazards ratio = 1.77; 95% confidence interval = 1.10 to 2.83, P = .018). Urban residents with high SDOH scores were at an even greater risk. Identifying SDOH and mitigating underlying factors may help control HTN, the most typical disease process treated in all cardio-oncology clinics.
Collapse
Affiliation(s)
- Priyanshu Nain
- Department of Medicine, Division of Cardiology, and Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
- Cardio-Oncology Program, Department of Medicine, Cardiology Division, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Nickolas Stabellini
- Cardio-Oncology Program, Department of Medicine, Cardiology Division, Medical College of Georgia at Augusta University, Augusta, GA, USA
- Division of Hematology and Oncology, Department of Medicine, University Hospitals/Seidman Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Omar M Makram
- Department of Medicine, Division of Cardiology, and Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
- Cardio-Oncology Program, Department of Medicine, Cardiology Division, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Johnathan Rast
- Department of Internal Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | | | - Gaurav Gopu
- Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Aditya Bhave
- Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Lakshya Seth
- Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Vraj Patel
- Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Stephanie Jiang
- Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Sarah Malik
- Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Ahmed Shetewi
- Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Alberto J Montero
- Division of Hematology and Oncology, Department of Medicine, University Hospitals/Seidman Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Jennifer Cullen
- Cancer Population Sciences, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Neeraj Agarwal
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Xiaoling Wang
- Georgia Prevention Institute, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Bonnie Ky
- Department of Biostatistics, Epidemiology, & Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lauren A Baldassarre
- Section of Cardiovascular Medicine, Department of Medicine and Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Neal L Weintraub
- Department of Medicine, Division of Cardiology, and Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
- Cardio-Oncology Program, Department of Medicine, Cardiology Division, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Ryan A Harris
- Georgia Prevention Institute, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Avirup Guha
- Department of Medicine, Division of Cardiology, and Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
- Cardio-Oncology Program, Department of Medicine, Cardiology Division, Medical College of Georgia at Augusta University, Augusta, GA, USA
| |
Collapse
|