1
|
Marwick TH, Dewar E, Nolan M, Shirazi M, Dias P, Wright L, Fitzgerald B, Kearney L, Srivastava P, Atherton J, Negishi K, Sverdlov AL, Wahi S, Otton J, Selvanayagam J, Thomas L, Thavendiranathan P. Strain surveillance during chemotherapy to improve cardiovascular outcomes: the SUCCOUR-MRI trial. Eur Heart J 2024; 45:4414-4424. [PMID: 39217601 PMCID: PMC11542702 DOI: 10.1093/eurheartj/ehae574] [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: 06/18/2024] [Revised: 07/17/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND AND AIMS The detection of cancer therapy-related cardiac dysfunction (CTRCD) by reduction of left ventricular ejection fraction (LVEF) during chemotherapy usually triggers the initiation of cardioprotective therapy. This study addressed whether the same approach should be applied to patients with worsening of global longitudinal strain (GLS) without attaining thresholds of LVEF. METHODS Strain surveillance during chemotherapy for improving cardiovascular outcomes (SUCCOUR-MRI) was a prospective multicentre randomized controlled trial involving 14 sites. Of 355 patients receiving anthracyclines with normal baseline LVEF, 333 patients (age 59 ± 13 years, 79% women) with at least one other CTRCD risk factor, able to undergo magnetic resonance imaging (MRI), GLS, and three-dimensional echocardiography were tracked over 12 months. A total of 105 patients (age 59 ± 13 years, 75% women, 69% breast cancer) developing GLS-CTRCD (>12% relative reduction of GLS without a change in LVEF) were randomized to cardioprotection with neurohormonal antagonists vs. usual care. The primary endpoint was 12-month change in MRI-LVEF; the secondary endpoint was MRI-LVEF-defined CTRCD. RESULTS During follow-up, two patients died, and two developed heart failure. Most patients were randomized at 3 months (62%). Median doses of angiotensin inhibition/blockade and beta-blockade were 75% and 50% of respective targets; 21 (43%) had side-effects attributed to cardioprotection. Due to a smaller LVEF change from baseline with cardioprotection than usual care (-2.5 ± 5.4% vs. -5.6 ± 5.9%, P = .009), follow-up LVEF was higher after cardioprotection (59 ± 5% vs. 55 ± 6%, P < .0001). After adjustment for baseline LVEF, the mean (95% confidence interval) difference in the change in LVEF between the two groups was -3.6% (-1.8% to -5.5%, P < .001). After cardioprotection, 1/49 patients developed 12-month LVEF-CTRCD, compared to 6/56 in usual care (P = .075). Global longitudinal strain improved at 3 months post-randomization in the cardioprotection group, with little change with usual care. CONCLUSIONS In patients with isolated GLS reduction after anthracyclines, cardioprotection is associated with better preservation of 12-month MRI-LVEF compared with usual care.
Collapse
Affiliation(s)
- Thomas H Marwick
- Imaging Research, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Cardiovascular Imaging, Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Elizabeth Dewar
- Imaging Research, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Mark Nolan
- Imaging Research, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Cardio-Oncology Section, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Mitra Shirazi
- Department of Cardiology, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Peter Dias
- Advara Heart Care, Murdoch, WA, Australia
| | - Leah Wright
- Imaging Research, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | | | | | | | - John Atherton
- University of Queensland Faculty of Medicine, Royal Brisbane and Women’s Hospital, Brisbane, QLD, Australia
| | - Kazuaki Negishi
- Cardiology Department, Nepean Hospital, Kingswood, NSW, Australia
- Nepean Clinical School, University of Sydney, Kingswood, NSW, Australia
| | - 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
| | - Sudhir Wahi
- Princess Alexandra Hospital, Brisbane, Australia
| | - James Otton
- Liverpool Hospital, Liverpool, NSW, Australia
| | | | - Liza Thomas
- Westmead Clinical School, University of Sydney and University of New South Wales, Sydney, NSW, Australia
| | | |
Collapse
|
2
|
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
|
3
|
Farooq J, Sultana R, James JP, Fathima C Z, Almutairy AF, Hussain ASM. Efficacy of Thymoquinone and Hesperidin in Attenuating Cardiotoxicity from 5-Fluorouracil: Insights from In Vivo and In Silico Studies. TOXICS 2024; 12:688. [PMID: 39330616 PMCID: PMC11435605 DOI: 10.3390/toxics12090688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 09/28/2024]
Abstract
5-Fluorouracil (5-FU) is widely used in chemotherapy but poses serious risks of cardiotoxicity, which can significantly affect treatment outcomes. Identifying interventions that can prevent these adverse effects without undermining anticancer efficacy is crucial. This study investigates the efficacy of Thymoquinone (TQ) and Hesperidin (HESP) in preventing cardiotoxicity induced by 5-FU in Wistar rats and elucidates the molecular interactions through docking studies. We employed an experimental design involving multiple groups of Wistar rats exposed to 5-FU, with and without the concurrent administration of TQ and HESP. Cardiac function markers, oxidative stress indicators, and inflammatory markers were assessed. Additionally, molecular docking was used to analyze the interaction of TQ and HESP with key inflammatory proteins. Treatment with TQ and HESP not only lowered levels of cardiac enzymes but also improved antioxidant capacity and reduced inflammation in cardiac tissues. Notably, the combination of TQ and HESP provided more significant protective effects than either agent alone. Molecular docking supported these findings, showing effective binding of TQ and HESP to inflammatory targets. TQ and HESP demonstrate potential as protective agents against cardiotoxicity in 5-FU-treated rats, with their combined use offering enhanced protection. These findings suggest a viable strategy for reducing cardiac risks associated with 5-FU chemotherapy.
Collapse
Affiliation(s)
- Juveriya Farooq
- Department of Pharmacology, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, India
| | - Rokeya Sultana
- Department of Pharmacognosy, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, India
| | - Jainey P James
- Department of Pharmaceutical Chemistry, NGSM Institute of Pharmaceutical Sciences (NGSMIPS), Nitte (Deemed to be University), Mangalore 575018, India
| | - Zakiya Fathima C
- Department of Pharmaceutical Chemistry, NGSM Institute of Pharmaceutical Sciences (NGSMIPS), Nitte (Deemed to be University), Mangalore 575018, India
| | - Ali F Almutairy
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia
| | | |
Collapse
|
4
|
Daniele AJ, Gregorietti V, Costa D, López-Fernández T. Use of EMPAgliflozin in the prevention of CARDiotoxicity: the EMPACARD - PILOT trial. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:58. [PMID: 39237985 PMCID: PMC11375926 DOI: 10.1186/s40959-024-00260-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 08/22/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND Anthracycline-based chemotherapy represents a cornerstone treatment for a number of common cancers, including breast cancer, lymphoma, and sarcoma. However, anthracycline-induced cardiotoxicity remains a significant concern, often presenting as a decline in cardiac function which can ultimately lead to heart failure (HF) or asymptomatic left ventricular dysfunction, in up to 10-15% of patients.Sodium-glucose transport protein 2 inhibitor (SGLT2i) therapies have been demonstrated to reduce the incidence of HF in high-risk non-cancer patients. Preliminary retrospective data suggest their role in mitigating the incidence of HF during or after anthracycline treatment METHODS: The EMPACARD-PILOT trial was a prospective case‒control study involving breast cancer patients scheduled to undergo anthracycline-based chemotherapy in a 4-cycle protocol of 60 mg/m2 doxorubicin. We used the HFA/ICOS risk score to identify patients at high or very high risk of cardiotoxicity. Patients with diabetes mellitus or stable heart failure with preserved ejection fraction (HFpEF) were prescribed empagliflozin (10 mg per day), starting seven days before the administration of anthracyclines and continuing for a period of six months. Those not meeting these criteria served as controls. The primary endpoint was cancer therapy-related cardiac dysfunction (CTRCD) incidence. CTRCD was defined as either a decrease in left ventricular ejection fraction (LVEF) of at least 10% to a final value below 50% or a reduction in global longitudinal strain (GLS) of at least 15% from baseline at any point during the study. The secondary endpoints included mortality and hospitalization due to cardiovascular causes or clinical heart failure. Exploratory endpoints included increases in serum troponin and NT-proBNP levels and a decrease in the glomerular filtration rate (GFR). The safety endpoints tracked includedketoacidosis, hypoglycemia, sepsis, neutropenic fever, and urinary tract infections. RESULTS During the enrollment period, 785 breast cancer patients were analysed. Of these, 107 met the inclusion criteria, and 76 subsequently provided informed consent. The study was conducted with comparable adherence rates of 81.5% in both the empagliflozin group (n = 38) and the control group (n = 38). The follow-up data from 62 patients revealed a significant reduction in the primary outcome within 6 months for the empagliflozin group compared with the control group (6.5% vs. 35.5%, p = 0.005), with a relative risk of 0.18 (95% CI: 0.04-0.75). Compared with the control treatment, treatment with empagliflozin also significantly preserved the ejection fraction at 6 months follow-up (56.8% ± 5.8% vs. 53.7% ± 6.7, p = 0.029). However, there were no significant differences between the groups in terms of NT-proBNP, cTnI, clinical heart failure, GFR, or mortality/hospitalization due to heart failure. CONCLUSION Empagliflozin is associated with reduced incidence of CTRCD in high-risk patients treated with anthracyclines. These data should serve as the foundation for a clinical trial to test whether SGLT2 inhibitors can reduce the incidence of heart failure in this patient group.
Collapse
Affiliation(s)
| | | | - Diego Costa
- Sanatorio Sagrado Corazón, Buenos Aires, Argentina
| | | |
Collapse
|
5
|
Repp ML, Edwards MD, Burch CS, Rao A, Chinyere IR. PCSK9 Inhibitors and Anthracyclines: The Future of Cardioprotection in Cardio-Oncology. HEARTS 2024; 5:375-388. [PMID: 39268545 PMCID: PMC11391951 DOI: 10.3390/hearts5030027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024] Open
Abstract
The field of cardio-oncology is an expanding frontier within cardiovascular medicine, and the need for evidence-based guidelines is apparent. One of the emerging focuses within cardio-oncology is the concomitant use of medications for cardioprotection in the setting of chemotherapy regimens that have known cardiovascular toxicity. While clinical trials focusing on cardioprotection during chemotherapy are sparse, an inaugural trial exploring the prophylactic potential of Sodium-Glucose Cotransporter-2 inhibitors (SGLT2is) for anthracycline (ANT)-induced cardiotoxicity has recently commenced. Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, though less studied in this oncology demographic, have exhibited promise in preclinical studies for conferring cardiac protection during non-ischemic toxic insults. While primarily used to reduce low-density lipoprotein, PCSK9 inhibitors exhibit pleiotropic effects, including the attenuation of inflammation, reactive oxygen species, and endothelial dysfunction. In ANT-induced cardiotoxicity, these same processes are accelerated, resulting in premature termination of treatment, chronic cardiovascular sequelae, heart failure, and/or death. This review serves a dual purpose: firstly, to provide a concise overview of the mechanisms implicated in ANT-induced cardiotoxicity, and, finally, to summarize the existing preclinical data supporting the theoretical possibility of the cardioprotective effects of PCSK9 inhibition in ANT-induced cardiotoxicity.
Collapse
Affiliation(s)
- Matthew L Repp
- Department of Medicine, University of Colorado, Aurora, CO 80045, USA
| | - Mark D Edwards
- Department of Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Christopher S Burch
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Amith Rao
- Department of Medicine, Banner University Medicine, Tucson, AZ 85724, USA
| | - Ikeotunye Royal Chinyere
- Department of Medicine, Banner University Medicine, Tucson, AZ 85724, USA
- Sarver Heart Center, University of Arizona, 1501 North Campbell Avenue, Room 6154, Tucson, AZ 85724, USA
| |
Collapse
|
6
|
Martinez-Dominguez P, Ana-Bayona MJS, Guerra EC, Espinola-Zavaleta N. Cancer Therapy-Related Cardiac Dysfunction: Strategies for Enhancing Cardiac Recovery. Methodist Debakey Cardiovasc J 2024; 20:109-112. [PMID: 39184162 PMCID: PMC11342827 DOI: 10.14797/mdcvj.1430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 06/19/2024] [Indexed: 08/27/2024] Open
Abstract
Chemotherapy has markedly improved cancer outcomes, yet cancer therapy-related cardiac dysfunction (CTRCD) poses a significant challenge, affecting around 10% of patients. CTRCD can be asymptomatic or present with heart failure symptoms. Multimodality imaging, particularly echocardiography, remains pivotal for monitoring cardiac function. Potential biomarkers for CTRCD assessment include troponin and B-type natriuretic peptide. Pharmacological interventions, such as dexrazoxane, angiotensin-converting enzyme inhibitors, and statins, play a crucial role in primary prevention and mitigating cardiotoxicity alongside cardiac rehabilitation programs. Thus, a comprehensive approach is essential for optimal cardiac recovery and improved patient outcomes.
Collapse
Affiliation(s)
- Pavel Martinez-Dominguez
- National Institute of Cardiology Ignacio Chavez, Mexico City, Mexico
- Autonomous University of Chihuahua, Chihuahua, Mexico
| | | | - Enrique C. Guerra
- National Institute of Cardiology Ignacio Chavez, Mexico City, Mexico
- National Autonomous University of Mexico, Mexico City, Mexico
| | - Nilda Espinola-Zavaleta
- National Institute of Cardiology Ignacio Chavez, Mexico City, Mexico
- ABC Medical Center, Mexico City, Mexico
| |
Collapse
|
7
|
Obeidat O, Obeidat A, Ismail MF, Obeidat A, Alqudah Q, Al-Ani H, Abughazaleh S, Tarawneh M, Daise M, Alzghoul B. Impact of breast cancer on in-hospital mortality and health care utilization in female heart failure patients: a retrospective cohort study. Proc AMIA Symp 2024; 37:794-801. [PMID: 39165821 PMCID: PMC11332614 DOI: 10.1080/08998280.2024.2364166] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 05/13/2024] [Accepted: 05/27/2024] [Indexed: 08/22/2024] Open
Abstract
Background Heart failure (HF) and breast cancer are major health concerns with overlapping risk factors. This study investigated the impact of breast cancer on in-hospital mortality, length of stay, and health care charges in patients with HF. Methods A retrospective cohort study was conducted using data from the National Inpatient Sample, focusing on female patients diagnosed with both breast cancer and HF. A control group of patients with HF without breast cancer was also analyzed. Main outcomes included in-hospital mortality, length of stay, and health care charges. Statistical analysis involved logistic and linear regression models. Results The study included 17,551 unweighted cases of breast cancer, corresponding to 87,755 weighted cases. Breast cancer, particularly metastatic, was associated with increased in-hospital mortality across various types of HF. Patients with breast cancer and HF had longer hospital stays, which was more pronounced in metastatic cases. However, the impact on hospital charges was not consistent across the different HF types. Racial disparities were evident, with Native Americans showing the highest mortality risk in acute HF. Conclusion Breast cancer significantly increases the in-hospital mortality risk and length of hospital stay in patients with HF. These findings highlight the need for integrated cardiovascular and oncological care, especially in the presence of metastatic breast cancer. The study underscores the importance of tailored management strategies for patients with HF with concurrent breast cancer and points toward the necessity for addressing racial disparities in health care.
Collapse
Affiliation(s)
- Omar Obeidat
- Graduate Medical Education, University of Central Florida College of Medicine, Gainesville, Florida, USA
- Department of Internal Medicine, HCA Florida North Florida Hospital, Gainesville, Florida, USA
| | - Ali Obeidat
- Division of Cardiology, Department of Internal Medicine, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Mohamed F. Ismail
- Graduate Medical Education, University of Central Florida College of Medicine, Gainesville, Florida, USA
- Department of Internal Medicine, HCA Florida North Florida Hospital, Gainesville, Florida, USA
| | - Abedallah Obeidat
- Division of Cardiology, Department of Internal Medicine, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Qusai Alqudah
- Graduate Medical Education, University of Central Florida College of Medicine, Gainesville, Florida, USA
- Department of Internal Medicine, HCA Florida North Florida Hospital, Gainesville, Florida, USA
| | - Hashim Al-Ani
- Graduate Medical Education, University of Central Florida College of Medicine, Gainesville, Florida, USA
- Department of Internal Medicine, HCA Florida North Florida Hospital, Gainesville, Florida, USA
| | - Saeed Abughazaleh
- St. Elizabeth’s Medical Center, Tufts University School of Medicine, Brighton, Massachusetts
| | - Mohammad Tarawneh
- St. Elizabeth’s Medical Center, Tufts University School of Medicine, Brighton, Massachusetts
| | - Moh’d Daise
- Graduate Medical Education, University of Central Florida College of Medicine, Gainesville, Florida, USA
- Department of Internal Medicine, HCA Florida North Florida Hospital, Gainesville, Florida, USA
| | - Bashar Alzghoul
- Department of Medicine, University of Florida Health, Gainesville, Florida, USA
| |
Collapse
|
8
|
Mohsenizadeh SA, Rajaeinejad M, Khoshfetrat M, Arefizadeh R, Mousavi SH, Mosaed R, Kazemi-Galougahi MH, Jalaeikhoo H, Faridfar A, Nikandish M, Alavi-Moghadam S, Arjmand B. Anthracycline-Induced Cardiomyopathy in Cancer Survivors: Management and Long-Term Implications. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024. [PMID: 38842787 DOI: 10.1007/5584_2024_804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
Recent advancements in personalized treatments, such as anthracycline chemotherapy, coupled with timely diagnoses, have contributed to a decrease in cancer-specific mortality rates and an improvement in cancer prognosis. Anthracyclines, a potent class of antibiotics, are extensively used as anticancer medications to treat a broad spectrum of tumors. Despite these advancements, a considerable number of cancer survivors face increased risks of treatment complications, particularly the cardiotoxic effects of chemotherapeutic drugs like anthracyclines. These effects can range from subclinical manifestations to severe consequences such as irreversible heart failure and death, highlighting the need for effective management of chemotherapy side effects for improved cancer care outcomes. Given the lack of specific treatments, early detection of subclinical cardiac events post-anthracycline therapy and the implementation of preventive strategies are vital. An interdisciplinary approach involving cardiovascular teams is crucial for the prevention and efficient management of anthracycline-induced cardiotoxicity. Various factors, such as age, gender, duration of treatment, and comorbidities, should be considered significant risk factors for developing chemotherapy-related cardiotoxicity. Tools such as electrocardiography, echocardiography, nuclear imaging, magnetic resonance imaging, histopathologic evaluations, and serum biomarkers should be appropriately used for the early detection of anthracycline-related cardiotoxicity. Furthermore, understanding the underlying biological mechanisms is key to developing preventive measures and personalized treatment strategies to mitigate anthracycline-induced cardiotoxicity. Exploring specific cardiotoxic mechanisms and identifying genetic variations can offer fresh perspectives on innovative, personalized treatments. This chapter aims to discuss cardiomyopathy following anthracycline therapy, with a focus on molecular mechanisms, preventive strategies, and emerging treatments.
Collapse
Affiliation(s)
| | - Mohsen Rajaeinejad
- AJA Cancer Epidemiology Research and Treatment Center (AJA-CERTC), AJA University of Medical Sciences, Tehran, Iran
| | - Mehran Khoshfetrat
- Department of Cardiology, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Reza Arefizadeh
- Department of Cardiology, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Seyed Hossein Mousavi
- Department of Cardiology, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Reza Mosaed
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran
- Student Research Committee, AJA University of Medical Sciences, Tehran, Iran
| | | | - Hasan Jalaeikhoo
- AJA Cancer Epidemiology Research and Treatment Center (AJA-CERTC), AJA University of Medical Sciences, Tehran, Iran
| | - Ali Faridfar
- AJA Cancer Epidemiology Research and Treatment Center (AJA-CERTC), AJA University of Medical Sciences, Tehran, Iran
| | - Mohsen Nikandish
- AJA Cancer Epidemiology Research and Treatment Center (AJA-CERTC), AJA University of Medical Sciences, Tehran, Iran
| | - Sepideh Alavi-Moghadam
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Lai J, Chen Z, Liu J, Zhu C, Huang H, Yi Y, Cai G, Liao N. A radiogenomic multimodal and whole-transcriptome sequencing for preoperative prediction of axillary lymph node metastasis and drug therapeutic response in breast cancer: a retrospective, machine learning and international multicohort study. Int J Surg 2024; 110:2162-2177. [PMID: 38215256 PMCID: PMC11019980 DOI: 10.1097/js9.0000000000001082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 12/27/2023] [Indexed: 01/14/2024]
Abstract
BACKGROUND Axillary lymph nodes (ALN) status serves as a crucial prognostic indicator in breast cancer (BC). The aim of this study was to construct a radiogenomic multimodal model, based on machine learning and whole-transcriptome sequencing (WTS), to accurately evaluate the risk of ALN metastasis (ALNM), drug therapeutic response and avoid unnecessary axillary surgery in BC patients. METHODS In this study, conducted a retrospective analysis of 1078 BC patients from The Cancer Genome Atlas (TCGA), The Cancer Imaging Archive (TCIA), and Foshan cohort. These patients were divided into the TCIA cohort ( N =103), TCIA validation cohort ( N =51), Duke cohort ( N =138), Foshan cohort ( N =106), and TCGA cohort ( N =680). Radiological features were extracted from BC radiological images and differentially expressed gene expression was calibrated using technology. A support vector machine model was employed to screen radiological and genetic features, and a multimodal model was established based on radiogenomic and clinical pathological features to predict ALNM. The accuracy of the model predictions was assessed using the area under the curve (AUC) and the clinical benefit was measured using decision curve analysis. Risk stratification analysis of BC patients was performed by gene set enrichment analysis, differential comparison of immune checkpoint gene expression, and drug sensitivity testing. RESULTS For the prediction of ALNM, rad-score was able to significantly differentiate between ALN- and ALN+ patients in both the Duke and Foshan cohorts ( P <0.05). Similarly, the gene-score was able to significantly differentiate between ALN- and ALN+ patients in the TCGA cohort ( P <0.05). The radiogenomic multimodal nomogram demonstrated satisfactory performance in the TCIA cohort (AUC 0.82, 95% CI: 0.74-0.91) and the TCIA validation cohort (AUC 0.77, 95% CI: 0.63-0.91). In the risk sub-stratification analysis, there were significant differences in gene pathway enrichment between high and low-risk groups ( P <0.05). Additionally, different risk groups may exhibit varying treatment responses ( P <0.05). CONCLUSION Overall, the radiogenomic multimodal model employs multimodal data, including radiological images, genetic, and clinicopathological typing. The radiogenomic multimodal nomogram can precisely predict ALNM and drug therapeutic response in BC patients.
Collapse
Affiliation(s)
- Jianguo Lai
- Department of Breast Cancer, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Yuexiu District, Guangzhou, Guangdong
| | - Zijun Chen
- The Second Clinical School of Southern Medical University, Guangzhou
| | - Jie Liu
- Department of Breast Cancer, Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University
| | - Chao Zhu
- Department of Blood Transfusion, The First Affiliated Hospital of Nanchang University
| | - Haoxuan Huang
- Department of Urology, Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Ying Yi
- Department of Radiology, The First People's Hospital of Foshan, Foshan, Guangdong
| | - Gengxi Cai
- Department of Breast Surgery, The First People’s Hospital of Foshan, Foshan, Guangdong
| | - Ning Liao
- Department of Breast Cancer, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Yuexiu District, Guangzhou, Guangdong
| |
Collapse
|
10
|
Nagy A, Börzsei D, Hoffmann A, Török S, Veszelka M, Almási N, Varga C, Szabó R. A Comprehensive Overview on Chemotherapy-Induced Cardiotoxicity: Insights into the Underlying Inflammatory and Oxidative Mechanisms. Cardiovasc Drugs Ther 2024:10.1007/s10557-024-07574-0. [PMID: 38492161 DOI: 10.1007/s10557-024-07574-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/05/2024] [Indexed: 03/18/2024]
Abstract
While oncotherapy has made rapid progress in recent years, side effects of anti-cancer drugs and treatments have also come to the fore. These side effects include cardiotoxicity, which can cause irreversible cardiac damages with long-term morbidity and mortality. Despite the continuous in-depth research on anti-cancer drugs, an improved knowledge of the underlying mechanisms of cardiotoxicity are necessary for early detection and management of cardiac risk. Although most reviews focus on the cardiotoxic effect of a specific individual chemotherapeutic agent, the aim of our review is to provide comprehensive insight into various agents that induced cardiotoxicity and their underlying mechanisms. Characterization of these mechanisms are underpinned by research on animal models and clinical studies. In order to gain insight into these complex mechanisms, we emphasize the role of inflammatory processes and oxidative stress on chemotherapy-induced cardiac changes. A better understanding and identification of the interplay between chemotherapy and inflammatory/oxidative processes hold some promise to prevent or at least mitigate cardiotoxicity-associated morbidity and mortality among cancer survivors.
Collapse
Affiliation(s)
- András Nagy
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Denise Börzsei
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Alexandra Hoffmann
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Szilvia Török
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Médea Veszelka
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Nikoletta Almási
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Csaba Varga
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Renáta Szabó
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary.
| |
Collapse
|
11
|
Amin AM, Khlidj Y, Abuelazm M, Ibrahim AA, Tanashat M, Imran M, Nazir A, Shaikhkhalil H, Abdelazeem B. The efficacy and safety of exercise regimens to mitigate chemotherapy cardiotoxicity: a systematic review and meta-analysis of randomized controlled trials. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:10. [PMID: 38395955 PMCID: PMC10885653 DOI: 10.1186/s40959-024-00208-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 02/05/2024] [Indexed: 02/25/2024]
Abstract
BACKGROUND Cardiotoxicity is one of the most common adverse events of the chemotherapy. Physical exercise was shown to be cardioprotective. We aim to estimate the efficacy and safety of exercise in cancer patients receiving cardiotoxic chemotherapy. METHODS We conducted a systematic review and meta-analysis of randomized controlled trials (RCTs), which were retrieved by systematically searching PubMed, Web of Science, SCOPUS, Cochrane, Clinical Trials.gov, and MedRxiv through July 17th, 2023. We used RevMan V. 5.4 to pool dichotomous data using risk ratio (RR) and continuous data using mean difference (MD), with a 95% confidence interval (CI). PROSPERO ID CRD42023460902. RESULTS We included thirteen RCTs with a total of 952 patients. Exercise significantly increased VO2 peak (MD: 1.95 with 95% CI [0.59, 3.32], P = 0.005). However, there was no significant effect regarding left ventricular ejection fraction, global longitudinal strain, cardiac output, stroke volume, left ventricular end-diastolic volume, left ventricular end-systolic volume, E/A ratio, resting heart rate, peak heart rate, resting systolic blood pressure, and resting diastolic blood pressure. Also, there was no significant difference regarding any adverse events (AEs) (RR: 4.44 with 95% CI [0.47, 41.56], P = 0.19), AEs leading to withdrawal (RR: 2.87 with 95% CI [0.79, 10.43], P = 0.11), serious AEs (RR: 3.00 with 95% CI [0.14, 65.90], P = 0.49), or all-cause mortality (RR: 0.25 with 95% CI [0.03, 2.22], P = 0.21). CONCLUSION Exercise is associated with increased VO2 peak in cancer patients receiving cardiotoxic chemotherapy. However, there was no significant difference between exercise and usual care regarding the echocardiographic and safety outcomes.
Collapse
Affiliation(s)
| | - Yehya Khlidj
- Faculty of Medicine, Algiers University, Algiers, Algeria
| | | | | | | | - Muhammad Imran
- University College of Medicine and Dentistry, The University of Lahore, Lahore, Pakistan
| | - Abubakar Nazir
- Faculty of Medicine, King Edward Medical University, Lahore, Pakistan
| | | | - Basel Abdelazeem
- Department of Cardiology, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
12
|
Hernández-Durán J, López-Gutiérrez LV, Palacio-Mejía MI, Aguilera L, Burgos L, Giraldo CIS. What Do We Know About Heart Failure in Latin American Women? Curr Probl Cardiol 2024; 49:102085. [PMID: 37716539 DOI: 10.1016/j.cpcardiol.2023.102085] [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: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 09/18/2023]
Abstract
Data about heart failure in Latin America is scarce. Women living in this region of the world are exposed to a mix of traditional risk factors for heart failure, neglected diseases, and social determinants of health. The aim of this review is to present what we know about heart failure in Latin American women and to establish the needs for future research.
Collapse
Affiliation(s)
| | | | | | - Luisa Aguilera
- Puerta de Hierro Cardiovascular Institute, Zapopan, Mexico
| | | | | |
Collapse
|
13
|
Brown SA, Hamid A, Pederson E, Bs AH, Maddula R, Goodman R, Lamberg M, Caraballo P, Noseworthy P, Lukan O, Echefu G, Berman G, Choudhuri I. Simplified rules-based tool to facilitate the application of up-to-date management recommendations in cardio-oncology. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2023; 9:37. [PMID: 37891699 PMCID: PMC10605976 DOI: 10.1186/s40959-023-00179-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 05/24/2023] [Indexed: 10/29/2023]
Abstract
BACKGROUND Millions of cancer survivors are at risk of cardiovascular diseases, a leading cause of morbidity and mortality. Tools to potentially facilitate implementation of cardiology guidelines, consensus recommendations, and scientific statements to prevent atherosclerotic cardiovascular disease (ASCVD) and other cardiovascular diseases are limited. Thus, inadequate utilization of cardiovascular medications and imaging is widespread, including significantly lower rates of statin use among cancer survivors for whom statin therapy is indicated. METHODS In this methodological study, we leveraged published guidelines documents to create a rules-based tool to include guidelines, expert consensus, and medical society scientific statements relevant to point of care cardiovascular disease prevention in the cardiovascular care of cancer survivors. Any overlap, redundancy, or ambiguous recommendations were identified and eliminated across all converted sources of knowledge. The integrity of the tool was assessed with use case examples and review of subsequent care suggestions. RESULTS An initial selection of 10 guidelines, expert consensus, and medical society scientific statements was made for this study. Then 7 were kept owing to overlap and revisions in society recommendations over recent years. Extensive formulae were employed to translate the recommendations of 7 selected guidelines into rules and proposed action measures. Patient suitability and care suggestions were assessed for several use case examples. CONCLUSION A simple rules-based application was designed to provide a potential format to deliver critical cardiovascular disease best-practice prevention recommendations at the point of care for cancer survivors. A version of this tool may potentially facilitate implementing these guidelines across clinics, payers, and health systems for preventing cardiovascular diseases in cancer survivors. TRIAL REGISTRATION ClinicalTrials.Gov Identifier: NCT05377320.
Collapse
Affiliation(s)
- Sherry-Ann Brown
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA.
| | | | | | | | | | | | | | | | - Peter Noseworthy
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Opeoluwa Lukan
- Department of Internal Medicine, Baton Rouge General Medical Center, Baton Rouge, LA, USA
| | - Gift Echefu
- Department of Internal Medicine, Baton Rouge General Medical Center, Baton Rouge, LA, USA
| | | | | |
Collapse
|
14
|
Boluda B, Solana-Altabella A, Cano I, Martínez-Cuadrón D, Acuña-Cruz E, Torres-Miñana L, Rodríguez-Veiga R, Navarro-Vicente I, Martínez-Campuzano D, García-Ruiz R, Lloret P, Asensi P, Osa-Sáez A, Aguero J, Rodríguez-Serrano M, Buendía-Fuentes F, Megías-Vericat JE, Martín-Herreros B, Barragán E, Sargas C, Salas M, Wooddell M, Dharmani C, Sanz MA, De la Rubia J, Montesinos P. Incidence and Risk Factors for Development of Cardiac Toxicity in Adult Patients with Newly Diagnosed Acute Myeloid Leukemia. Cancers (Basel) 2023; 15:2267. [PMID: 37190195 PMCID: PMC10136564 DOI: 10.3390/cancers15082267] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/03/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
The incidence of cardiac morbimortality in acute myeloid leukemia (AML) is not well known. We aim to estimate the cumulative incidence (CI) of cardiac events in AML patients and to identify risk factors for their occurrence. Among 571 newly diagnosed AML patients, 26 (4.6%) developed fatal cardiac events, and among 525 treated patients, 19 (3.6%) experienced fatal cardiac events (CI: 2% at 6 months; 6.7% at 9 years). Prior heart disease was associated with the development of fatal cardiac events (hazard ratio (HR) = 6.9). The CI of non-fatal cardiac events was 43.7% at 6 months and 56.9% at 9 years. Age ≥ 65 (HR = 2.2), relevant cardiac antecedents (HR = 1.4), and non-intensive chemotherapy (HR = 1.8) were associated with non-fatal cardiac events. The 9-year CI of grade 1-2 QTcF prolongation was 11.2%, grade 3 was 2.7%, and no patient had grade 4-5 events. The 9-year CI of grade 1-2 cardiac failure was 1.3%, grade 3-4 was 15%, and grade 5 was 2.1%; of grade 1-2, arrhythmia was 1.9%, grade 3-4 was 9.1%, and grade 5 was 1%. Among 285 intensive therapy patients, median overall survival decreased in those experiencing grade 3-4 cardiac events (p < 0.001). We observed a high incidence of cardiac toxicity associated with significant mortality in AML.
Collapse
Affiliation(s)
- Blanca Boluda
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain (I.N.-V.)
- Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Antonio Solana-Altabella
- Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- Pharmacy Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
| | - Isabel Cano
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain (I.N.-V.)
- Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - David Martínez-Cuadrón
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain (I.N.-V.)
- Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Evelyn Acuña-Cruz
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain (I.N.-V.)
- Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Laura Torres-Miñana
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain (I.N.-V.)
- Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Rebeca Rodríguez-Veiga
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain (I.N.-V.)
- Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Irene Navarro-Vicente
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain (I.N.-V.)
- Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - David Martínez-Campuzano
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain (I.N.-V.)
| | - Raquel García-Ruiz
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain (I.N.-V.)
| | - Pilar Lloret
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain (I.N.-V.)
| | - Pedro Asensi
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain (I.N.-V.)
| | - Ana Osa-Sáez
- Cardiology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
| | - Jaume Aguero
- Cardiology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
| | | | | | | | - Beatriz Martín-Herreros
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain (I.N.-V.)
- Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Eva Barragán
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain (I.N.-V.)
- Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Claudia Sargas
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain (I.N.-V.)
- Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Maribel Salas
- Daiichi Sankyo, Inc., Basking Ridge, NJ 07920, USA
- Center for Real-World Effectiveness and Safety of Therapeutics (CREST), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | | | | | - Miguel A. Sanz
- Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Javier De la Rubia
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain (I.N.-V.)
- Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Internal Medicine, School of Medicine and Dentistry, Catholic University of Valencia, 46001 Valencia, Spain
| | - Pau Montesinos
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain (I.N.-V.)
- Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|