1
|
Reehal P, Lyon AR, Lee G. The role of a cardio-oncology clinical nurse specialist in the United Kingdom. Asia Pac J Oncol Nurs 2025; 12:100640. [PMID: 39927090 PMCID: PMC11803865 DOI: 10.1016/j.apjon.2024.100640] [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: 07/31/2024] [Accepted: 12/10/2024] [Indexed: 02/11/2025] Open
Abstract
Cardio-oncology is a growing subspeciality of cardiology that involves the prevention and early detection of cancer therapy-related cardiovascular toxicity (CTR-CVT). Cardiovascular complications can occur before, during and after cancer treatment, due to de novo cardiotoxicity or an exacerbation of a previous cardiac condition. Therefore, cancer patients undergoing cancer treatment need to be assessed before, during and after their cancer treatment. This article describes the development and progress of a specialised nursing role, known as a Clinical Nurse Specialist (CNS), at the Royal Brompton Hospital in London, United Kingdom (UK).
Collapse
Affiliation(s)
- Priya Reehal
- Cardio-Oncology Centre of Excellence, Royal Brompton Hospital, London, United Kingdom
| | - Alexander R. Lyon
- Cardio-Oncology Centre of Excellence, Royal Brompton Hospital, London, United Kingdom
| | - Geraldine Lee
- Catherine McAuley School of Nursing & Midwifery, Brookfield Health Sciences Complex, College Road, University College Cork, Cork, Ireland
| |
Collapse
|
2
|
Siaravas KC, Papafaklis MI, Moula AI, Michalis LK, Sioka C, Katsouras CS. Multivessel Coronary Artery Disease in Cancer Patients Undergoing Percutaneous Coronary Intervention: A Systematic Review and Meta-Analysis. Life (Basel) 2025; 15:571. [PMID: 40283126 PMCID: PMC12029082 DOI: 10.3390/life15040571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/23/2025] [Accepted: 03/28/2025] [Indexed: 04/29/2025] Open
Abstract
Cancer patients have a higher propensity for adverse cardiovascular outcomes, primarily due to the toxic effects of chemotherapeutic agents and radiation therapy. The objective of this systematic review and meta-analysis was to investigate the proportion of multivessel coronary artery disease (MVD) in cancer compared to non-cancer patients undergoing percutaneous coronary intervention (PCI). We systematically screened the literature for studies providing data on MVD in patients with and without cancer who underwent PCI. Seventeen observational studies (5200 patients with active cancer/history of cancer and 55,146 control patients without cancer) were included in the analysis. Most studies did not show statistically significant differences in the incidence of MVD. Overall, there was no significant difference in MVD occurrence in the cancer group (risk ratio [RR]: 1.03; 95% confidence intervals [CI]: 0.99-1.08; p = 0.19). A high degree of heterogeneity was observed among the studies (I2 = 57.32%). Further sub-analysis using only the six studies with matched control populations did not show significant differences in MVD between those groups (RR; 0.99, 95% CI: 0.94-1.05, p = 0.79). In addition, a subgroup analysis with patients who had acute coronary syndrome, who received radiation treatment, and in studies with cancer patients with active cancer did not change the statistical results. Our report highlights that there was no significant difference in the incidence of MVD between patients with and without cancer. Further research is needed to clarify the detailed characteristics of coronary artery disease in cancer patients.
Collapse
Affiliation(s)
| | - Michail I. Papafaklis
- Division of Cardiology, Faculty of Medicine, School of Health Sciences, University of Patras, 26504 Rio, Greece
| | - Amalia I. Moula
- Achilopouleio General Hospital of Volos, 38222 Volos, Greece
| | - Lampros K. Michalis
- Second Cardiology Department, University Hospital of Ioannina, 45500 Ioannina, Greece
| | - Chrissa Sioka
- Department of Nuclear Medicine, University Hospital of Ioannina, 45500 Ioannina, Greece
| | - Christos S. Katsouras
- First Cardiology Department, University Hospital of Ioannina, 45500 Ioannina, Greece
| |
Collapse
|
3
|
Rachma B, Savitri M, Sutanto H. Cardiotoxicity in platinum-based chemotherapy: Mechanisms, manifestations, and management. CANCER PATHOGENESIS AND THERAPY 2025; 3:101-108. [PMID: 40182123 PMCID: PMC11963179 DOI: 10.1016/j.cpt.2024.04.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/17/2024] [Accepted: 04/23/2024] [Indexed: 04/05/2025]
Abstract
Platinum-based chemotherapy, a cornerstone in the treatment of various malignancies, is often limited by its potential cardiotoxic effects. Understanding these effects is crucial for optimizing patient outcomes and guiding treatment decisions. This review explores the mechanisms, clinical manifestations, detection, management, and future directions in the research of cardiotoxicity associated with platinum-based chemotherapy. The mechanisms discussed here include oxidative stress, reactive oxygen species production, DNA damage, and alterations in signaling pathways. Clinical manifestations range from mild symptoms to severe complications, including Takotsubo cardiomyopathy, as highlighted by recent case studies. The role of diagnostic tools such as echocardiography, cardiac magnetic resonance imaging, and cardiac biomarkers in early detection is emphasized, underscoring the importance of regular cardiac monitoring. Management strategies focus on cardioprotective agents, alternative chemotherapy regimens, and emerging therapeutic approaches, including the potential of nano liposomal and cubosomal formulations. The review also delves into the future of personalized medicine in predicting and managing cardiotoxicity, advocating for ongoing research to mitigate these adverse effects. This comprehensive overview aims to enhance the understanding of cardiotoxicity in platinum-based chemotherapy, informing clinical practices and promoting patient-centric care.
Collapse
Affiliation(s)
- Betty Rachma
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, 60132, Indonesia
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, 60286, Indonesia
| | - Merlyna Savitri
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, 60132, Indonesia
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, 60286, Indonesia
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, 60132, Indonesia
| | - Henry Sutanto
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, 60132, Indonesia
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, 60286, Indonesia
| |
Collapse
|
4
|
El-Taji O, Taktak S, Jones C, Brown M, Clarke N, Sachdeva A. Cardiovascular Events and Androgen Receptor Signaling Inhibitors in Advanced Prostate Cancer: A Systematic Review and Meta-Analysis. JAMA Oncol 2024; 10:874-884. [PMID: 38842801 PMCID: PMC11157448 DOI: 10.1001/jamaoncol.2024.1549] [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: 09/19/2023] [Accepted: 12/29/2023] [Indexed: 06/07/2024]
Abstract
Importance Cardiovascular (CV) events remain a substantial cause of mortality among men with advanced and metastatic prostate cancer (PCa). The introduction of novel androgen receptor signaling inhibitors (ARSI) has transformed the treatment landscape of PCa in recent years; however, their associated CV toxic effects remains unclear. Objective To assess the incidence of CV events with addition of ARSI to standard of care (SOC) in locally advanced (M0) and metastatic (M1) PCa. Data Sources Systematic searches of PubMed, Scopus, Web of Science, EMBASE, and ClinicalTrials.gov were performed from inception up to May 2023. Study Selection Randomized clinical trials of ARSI agents (abiraterone, apalutamide, darolutamide, enzalutamide) that reported CV events among individuals with M0 and M1, hormone-sensitive prostate cancer (HSPC) and castration-resistant prostate cancer (CRPC). Data Extraction and Synthesis A systematic review was performed in accordance with PRISMA guidance. Two authors screened and independently evaluated studies eligible for inclusion. Data extraction and bias assessment was subsequently performed. Main Outcomes and Measures A random-effects meta-analysis was performed to estimate risk ratios for the incidence of all grade and grade 3 or higher CV events (primary outcomes), in addition to hypertension, acute coronary syndrome (ACS), cardiac dysrhythmia, CV death, cerebrovascular event, and venous thromboembolism (secondary outcomes). Sources of heterogeneity were explored using meta-regression. Results There were 24 studies (n = 22 166 patients; median age range, 63-77 years; median follow-up time range, 3.9-96 months) eligible for inclusion. ARSI therapy was associated with increased risk of all grade CV event (risk ratio [RR], 1.75; 95% CI, 1.50-2.04; P < .001) and grade 3 or higher CV events (RR, 2.10; 95%, 1.72-2.55; P < .001). ARSI therapy also was associated with increased risk for grade 3 or higher events for hypertension (RR, 2.25; 95% CI, 1.74-2.90; P < .001), ACS (RR, 1.93; 95% CI, 1.43-1.60; P < .01), cardiac dysrhythmia (RR, 1.64; 95% CI, 1.23-2.17; P < .001), cerebrovascular events (RR, 1.86; 95% CI, 1.34-2.59; P < .001) and for CV-related death (RR, 2.02; 95% CI, 1.32-3.10; P = .001). Subgroup analysis demonstrated increased risk of all CV events across the disease spectrum (M0 HSPC: RR, 2.26; 95% CI, 1.36-3.75; P = .002; M1 HSPC: RR, 1.85; 95% CI, 1.47-2.31; P < .001; M0 CRPC: RR, 1.79; 95% CI, 1.13-2.81; P = .01; M1 CRPC: RR, 1.46; 95% CI, 1.16-1.83; P = .001). Conclusions and Relevance This systematic review and meta-analysis found that the addition of ARSIs to traditional ADT was associated with increased risk of CV events across the prostate cancer disease spectrum. These results suggest that patients with prostate cancer should be advised about and monitored for the potential of increased risk of CV events with initiation of ARSI therapy alongside conventional hormonal therapy.
Collapse
Affiliation(s)
- Omar El-Taji
- Genito Urinary Cancer Research Group, Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
- Department of Surgery, The Christie NHS Foundation Trust, Manchester, United Kingdom
- Department of Urology, Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Manchester, United Kingdom
| | - Samih Taktak
- Wirral University Teaching Hospitals NHS Foundation Trust, Wirral, United Kingdom
| | - Craig Jones
- Genito Urinary Cancer Research Group, Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
- Department of Surgery, The Christie NHS Foundation Trust, Manchester, United Kingdom
- Department of Urology, Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Manchester, United Kingdom
| | - Mick Brown
- Genito Urinary Cancer Research Group, Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Noel Clarke
- Genito Urinary Cancer Research Group, Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
- Department of Surgery, The Christie NHS Foundation Trust, Manchester, United Kingdom
- Department of Urology, Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Manchester, United Kingdom
| | - Ashwin Sachdeva
- Genito Urinary Cancer Research Group, Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
- Department of Surgery, The Christie NHS Foundation Trust, Manchester, United Kingdom
| |
Collapse
|
5
|
Gong Y, Hu X, Chen M, Wang J. Recent progress of iron-based nanomaterials in gene delivery and tumor gene therapy. J Nanobiotechnology 2024; 22:309. [PMID: 38825720 PMCID: PMC11145874 DOI: 10.1186/s12951-024-02550-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 05/14/2024] [Indexed: 06/04/2024] Open
Abstract
Gene therapy aims to modify or manipulate gene expression and change the biological characteristics of living cells to achieve the purpose of treating diseases. The safe, efficient, and stable expression of exogenous genes in cells is crucial for the success of gene therapy, which is closely related to the vectors used in gene therapy. Currently, gene therapy vectors are mainly divided into two categories: viral vectors and non-viral vectors. Viral vectors are widely used due to the advantages of persistent and stable expression, high transfection efficiency, but they also have certain issues such as infectivity, high immunological rejection, randomness of insertion mutation, carcinogenicity, and limited vector capacity. Non-viral vectors have the advantages of non-infectivity, controllable chemical structure, and unlimited vector capacity, but the transfection efficiency is low. With the rapid development of nanotechnology, the unique physicochemical properties of nanomaterials have attracted increasing attention in the field of drug and gene delivery. Among many nanomaterials, iron-based nanomaterials have attracted much attention due to their superior physicochemical properties, such as Fenton reaction, magnetic resonance imaging, magnetothermal therapy, photothermal therapy, gene delivery, magnetically-assisted drug delivery, cell and tissue targeting, and so on. In this paper, the research progress of iron-based nanomaterials in gene delivery and tumor gene therapy is reviewed, and the future application direction of iron-based nanomaterials is further prospected.
Collapse
Affiliation(s)
- Ya Gong
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, Chongqing, 400037, China
| | - Xiaoyan Hu
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100864, China
| | - Ming Chen
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
| | - Jun Wang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
6
|
Al-Jazairi AS, Bahammam N, Aljuaid D, Almutairi L, Alshahrani S, Albuhairan N, Cahusac PMB, Korayem GB. Cardiovascular adverse events of antineoplastic monoclonal antibodies among cancer patients: real-world evidence from a tertiary healthcare system. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2023; 9:35. [PMID: 37749652 PMCID: PMC10519122 DOI: 10.1186/s40959-023-00184-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 08/04/2023] [Indexed: 09/27/2023]
Abstract
BACKGROUND Antineoplastic monoclonal antibodies (mAbs), such as trastuzumab, bevacizumab, and pertuzumab have been the mainstay of therapy in cancer patients. Despite proven efficacy of the monoclonal antibodies, cardiovascular-induced adverse events such as heart failure, hypertension, ischemic heart disease, arrhythmias, thromboembolic events, and hemorrhage remain a major complication. The European society of cardiology address that concern with antineoplastic monoclonal antibodies issuing a guideline to manage and monitor chemotherapy-induced cardiotoxicity. There is limited evidence of the real-world prevalence of cardiovascular (CV) events induced by monoclonal antibodies among patients with cancer in Saudi Arabia. OBJECTIVE To evaluate the prevalence of cardiovascular adverse events among patients with cancer treated with monoclonal antibodies in Saudi Arabia. METHODS This is a retrospective study conducted in a tertiary care hospital, Riyadh, Saudi Arabia. Data were obtained from an electronic medical record of patients with cancer treated with one of the selected monoclonal antibodies, who met the inclusion criteria between January 2005 until June 2015 and have been followed up for at least one year. Patients were stratified into groups according to monoclonal antibodies treatment: trastuzumab, bevacizumab, pertuzumab, and combined mAbs. RESULTS A total of 1067 patient were included in the study, within the pre-determined study period. The prevalence of cardiovascular disease among patients with cancer treated with monoclonal antibodies was 16.3%. The prevalence of heart failure was relatively higher in the trastuzumab group (46/626 patients, 7.3%). Among 418 patients treated with bevacizumab, hypertension was the most frequent adverse event, reported in 38 patients (9.1%), followed by thromboembolism reported in 27 patients (6.5%). Treatment discontinuation owing to cardiovascular adverse events was reported in 42/1,067 patients (3.9%). CONCLUSION AND RELEVANCE Prevalence of antineoplastic monoclonal antibody induced cardiovascular adverse events among patients with cancer is substantially high in Saudi Arabia. There is an urgent need to streamline the practice for identifying high risk patients and flexible referral system for cardio-oncology care.
Collapse
Affiliation(s)
- Abdulrazaq S Al-Jazairi
- Division of Clinical Trials Transformation Initiative, King Faisal Specialist Hospital & Research Centre, PO Box 3354, Riyadh, 11211, Kingdom of Saudi Arabia.
- College of Pharmacy and Medicine, Alfaisal University, P.O. Box 50927, Riyadh, 11533, Kingdom of Saudi Arabia.
| | - Nahlah Bahammam
- College of Pharmacy, Princess Nourah Bint Abdulrahman University, P.O. Box 101283, 11655, Riyadh, Saudi Arabia
| | - Dhai Aljuaid
- College of Pharmacy, Princess Nourah Bint Abdulrahman University, P.O. Box 101283, 11655, Riyadh, Saudi Arabia
| | - Lama Almutairi
- College of Pharmacy, Princess Nourah Bint Abdulrahman University, P.O. Box 101283, 11655, Riyadh, Saudi Arabia
| | - Shroog Alshahrani
- College of Pharmacy, Princess Nourah Bint Abdulrahman University, P.O. Box 101283, 11655, Riyadh, Saudi Arabia
| | - Norah Albuhairan
- King Faisal Specialist Hospital & Research Centre, PO Box 3354, Riyadh, 11211, Kingdom of Saudi Arabia
| | - Peter M B Cahusac
- College of Pharmacy and Medicine, Alfaisal University, P.O. Box 50927, Riyadh, 11533, Kingdom of Saudi Arabia
| | - Ghazwa B Korayem
- Department of Pharmacy Practice, College of Pharmacy, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, 11671, Riyadh, Saudi Arabia
| |
Collapse
|
7
|
Battisha A, Sawalha K, Obeidat Y, Patel B. Role of Cardiac Biomarkers in Monitoring Cardiotoxicity in Chemotherapy Patients. Crit Pathw Cardiol 2023; 22:83-87. [PMID: 37607037 DOI: 10.1097/hpc.0000000000000314] [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: 08/24/2023]
Abstract
PURPOSE This review aims to highlight the different types of chemotherapy-induced cardiotoxicity and will discuss the evidence base behind the use of different cardiac biomarkers to predict cardiovascular complications. Additionally, we will review the use of cardiac biomarkers to monitor cardiac outcomes and the role of cardioprotective medications in reducing cardiovascular side effects. RECENT FINDINGS Chemotherapy has been linked to an increased risk of cardiotoxicity and heart failure. Currently, patients receiving chemotherapy undergo echocardiogram before starting chemotherapy and every 6 months to monitor for any decline in cardiac function. We reviewed the current evidence and practice guidelines of monitoring chemotherapy cardiotoxicity. SUMMARY Cardio-oncology is a rapidly evolving subspecialty in cardiology, especially with the advent of new chemotherapeutic agents, which have cardiovascular side effects. Early detection of these effects is crucial to prevent life-threatening and irreversible cardiovascular outcomes. Monitoring troponin, pro-brain natriuretic peptide, and other cardiac biomarkers during chemotherapy will help to early detect cardiotoxicity.
Collapse
Affiliation(s)
- Ayman Battisha
- From the Department of Medicine, University of Massachusetts Chan Medical School-Baystate, Springfield, MA
| | - Khalid Sawalha
- From the Department of Medicine, University of Massachusetts Chan Medical School-Baystate, Springfield, MA
| | - Yasin Obeidat
- From the Department of Medicine, University of Massachusetts Chan Medical School-Baystate, Springfield, MA
| | - Brijesh Patel
- Department of Cardiology, Heart and Vascular Institute, West Virginia University, Morgantown, WV
| |
Collapse
|
8
|
Ritter A, Quartermaine C, Pierre-Charles J, Balasubramanian S, Raeisi-Giglou P, Addison D, Miller E. Cardiotoxicity of Anti-Cancer Radiation Therapy: a Focus on Heart Failure. Curr Heart Fail Rep 2023; 20:44-55. [PMID: 36692820 DOI: 10.1007/s11897-023-00587-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/06/2022] [Indexed: 01/25/2023]
Abstract
PURPOSE OF REVIEW As the percentage of patients achieving long-term survival following treatment of their cancer grows, it is increasingly important to understand the long-term toxicities of cancer-directed treatment. In this review, we highlight the recent findings regarding radiation-induced cardiotoxicity across multiple disease sites, with a particular focus on heart failure. RECENT FINDINGS Despite its relative lack of study historically, radiation-induced heart failure has now recently been implicated in several studies of breast cancer, lung cancer, esophageal cancer, and lymphoma as a non-trivial potential consequence of thoracic radiotherapy. Data regarding specific cardiac dosimetric endpoints relevant to cardiotoxicity continue to accumulate. Radiation-induced heart failure is a rare but significant toxicity of thoracic radiotherapy, that is likely underreported. Important areas for future focus include understanding the interplay between thoracic radiotherapy and concurrent cardiotoxic systemic therapy as well as development of potential mitigation strategies and novel therapeutics.
Collapse
Affiliation(s)
- Alex Ritter
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, OH, Columbus, USA
| | - Cooper Quartermaine
- Cardio-Oncology Program, Division of Cardiology, The Ohio State University Medical Center, Columbus, OH, USA
| | - Jovan Pierre-Charles
- Cardio-Oncology Program, Division of Cardiology, The Ohio State University Medical Center, Columbus, OH, USA
| | - Suryakumar Balasubramanian
- Cardio-Oncology Program, Division of Cardiology, The Ohio State University Medical Center, Columbus, OH, USA
- Velammal Medical College Hospital & Research Institute, Madurai, India
| | - Pejman Raeisi-Giglou
- Cardio-Oncology Program, Division of Cardiology, The Ohio State University Medical Center, Columbus, OH, USA
| | - Daniel Addison
- Cardio-Oncology Program, Division of Cardiology, The Ohio State University Medical Center, Columbus, OH, USA
- Division of Cancer Control, James Cancer Hospital and Solove Research Institute at The Ohio State University, Columbus, OH, USA
| | - Eric Miller
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, OH, Columbus, USA.
| |
Collapse
|
9
|
Kumar S, Basu M, Ghosh MK. Chaperone-assisted E3 ligase CHIP: A double agent in cancer. Genes Dis 2022; 9:1521-1555. [PMID: 36157498 PMCID: PMC9485218 DOI: 10.1016/j.gendis.2021.08.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/06/2021] [Indexed: 12/11/2022] Open
Abstract
The carboxy-terminus of Hsp70-interacting protein (CHIP) is a ubiquitin ligase and co-chaperone belonging to Ubox family that plays a crucial role in the maintenance of cellular homeostasis by switching the equilibrium of the folding-refolding mechanism towards the proteasomal or lysosomal degradation pathway. It links molecular chaperones viz. HSC70, HSP70 and HSP90 with ubiquitin proteasome system (UPS), acting as a quality control system. CHIP contains charged domain in between N-terminal tetratricopeptide repeat (TPR) and C-terminal Ubox domain. TPR domain interacts with the aberrant client proteins via chaperones while Ubox domain facilitates the ubiquitin transfer to the client proteins for ubiquitination. Thus, CHIP is a classic molecule that executes ubiquitination for degradation of client proteins. Further, CHIP has been found to be indulged in cellular differentiation, proliferation, metastasis and tumorigenesis. Additionally, CHIP can play its dual role as a tumor suppressor as well as an oncogene in numerous malignancies, thus acting as a double agent. Here, in this review, we have reported almost all substrates of CHIP established till date and classified them according to the hallmarks of cancer. In addition, we discussed about its architectural alignment, tissue specific expression, sub-cellular localization, folding-refolding mechanisms of client proteins, E4 ligase activity, normal physiological roles, as well as involvement in various diseases and tumor biology. Further, we aim to discuss its importance in HSP90 inhibitors mediated cancer therapy. Thus, this report concludes that CHIP may be a promising and worthy drug target towards pharmaceutical industry for drug development.
Collapse
Affiliation(s)
- Sunny Kumar
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector–V, Salt Lake, Kolkata- 700091 & 4, Raja S.C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Malini Basu
- Department of Microbiology, Dhruba Chand Halder College, Dakshin Barasat, South 24 Paraganas, West Bengal 743372, India
| | - Mrinal K. Ghosh
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector–V, Salt Lake, Kolkata- 700091 & 4, Raja S.C. Mullick Road, Jadavpur, Kolkata 700032, India
| |
Collapse
|
10
|
Georgiadis N, Tsarouhas K, Dorne JLCM, Kass GEN, Laspa P, Toutouzas K, Koulaouzidou EA, Kouretas D, Tsitsimpikou C. Cardiotoxicity of Chemical Substances: An Emerging Hazard Class. J Cardiovasc Dev Dis 2022; 9:226. [PMID: 35877588 PMCID: PMC9316944 DOI: 10.3390/jcdd9070226] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/16/2022] [Accepted: 07/12/2022] [Indexed: 11/16/2022] Open
Abstract
(1) Background: Human health risks and hazards from chemical substances are well regulated internationally. However, cardiotoxicity, is not defined as a stand-alone hazard and therefore there are no defined criteria for the classification of substances as cardiotoxic. Identifying and regulating substances that cause cardiovascular adverse effects would undoubtedly strengthen the national health systems. (2) Methods: To overcome the aforementioned gap, a roadmap is proposed for identifying regulatory criteria from animal studies and endorse legislation in order to classify substances as cardiotoxic. The roadmap consists of: (i) the identification of the appropriate animal species and strains; (ii) the identification of the lines of scientific evidence (e.g., histopathological, biochemical and echocardiographic indices etc.) from animal studies with relevance to humans; (iii) the statistical analysis and meta-analysis for each line of scientific evidence after exposure to well-established cardiotoxicants to humans (e.g., anthracyclines) in order to identify threshold values or range of normal and/ or altered values due to exposure; (iv) validation of the above described lines of evidence in animals exposed to other alleged cardiotoxic substances (e.g., anabolic androgen steroids (AAS) and pesticides); (v) establishment of mechanisms of action based on information of either known or alleged cardiotoxicants; and (vi) introduction of novel indices and in silico methods. (3) Results: Preliminary results in rats indicate a clear distinction from normal values to values measured in rats exposed to anthracyclines regarding left ventricle (LV) fractional shortening (FS) and LV ejection fraction (EF). A distinctive pattern is similarly observed for Creatine Kinase-Myocardial Band isoenzyme (CK-MB) and cardiac tissue glutathione (GSH). These findings are encouraging and indicate that there is room for targeted research to this end, and that these specific indices and biochemical markers should be further investigated in order to be developed to regulatory criteria. (4) Conclusions: Further research should be conducted by both the scientific and regulatory community that aims to clearly define the cardiotoxicity hazard caused by chemicals and develop a full set of scientific criteria.
Collapse
Affiliation(s)
- Nikolaos Georgiadis
- European Chemicals Agency, 00150 Helsinki, Finland;
- Department of Biochemistry & Biotechnology, University of Thessaly, 38221 Larissa, Greece; (P.L.); (C.T.)
| | | | | | - George E. N. Kass
- European Food Safety Authority, 43126 Parma, Italy; (J.-L.C.M.D.); (G.E.N.K.)
| | - Petroula Laspa
- Department of Biochemistry & Biotechnology, University of Thessaly, 38221 Larissa, Greece; (P.L.); (C.T.)
| | - Konstantinos Toutouzas
- First Department of Cardiology, Hippokration Hospital, Medical School, University of Athens, 11527 Athens, Greece;
| | - Elisabeth A. Koulaouzidou
- Division of Dental Tissues’ Pathology and Therapeutics (Basic Dental Sciences, Endodontology and Operative Dentistry), School of Dentistry, Aristotle University Thessaloniki, 54124 Thessaloniki, Greece;
| | - Dimitrios Kouretas
- Department of Biochemistry & Biotechnology, University of Thessaly, 38221 Larissa, Greece; (P.L.); (C.T.)
| | - Christina Tsitsimpikou
- Department of Biochemistry & Biotechnology, University of Thessaly, 38221 Larissa, Greece; (P.L.); (C.T.)
- Directorate of Energy, Industrial & Chemical Products, General Chemical State Laboratory of Greece, 11521 Athens, Greece
| |
Collapse
|
11
|
Chianca M, Panichella G, Fabiani I, Giannoni A, L'Abbate S, Aimo A, Del Franco A, Vergaro G, Grigoratos C, Castiglione V, Cipolla CM, Fedele A, Passino C, Emdin M, Cardinale DM. Bidirectional Relationship Between Cancer and Heart Failure: Insights on Circulating Biomarkers. Front Cardiovasc Med 2022; 9:936654. [PMID: 35872912 PMCID: PMC9299444 DOI: 10.3389/fcvm.2022.936654] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Cancer and heart failure are the two leading causes of death in developed countries. These two apparently distinct clinical entities share similar risk factors, symptoms, and pathophysiological mechanisms (inflammation, metabolic disturbances, neuro-hormonal and immune system activation, and endothelial dysfunction). Beyond the well-known cardiotoxic effects of oncological therapies, cancer and heart failure are thought to be tied by a bidirectional relationship, where one disease favors the other and vice versa. In this context, biomarkers represent a simple, reproducible, sensitive and cost-effective method to explore such relationship. In this review, we recapitulate the evidence on cardiovascular and oncological biomarkers in the field of cardioncology, focusing on their role in treatment-naïve cancer patients. Cardioncological biomarkers are useful tools in risk stratification, early detection of cardiotoxicity, follow-up, and prognostic assessment. Intriguingly, these biomarkers might contribute to better understand the common pathophysiology of cancer and heart failure, thus allowing the implementation of preventive and treatment strategies in cardioncological patients
Collapse
Affiliation(s)
- Michela Chianca
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | | | - Iacopo Fabiani
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
- *Correspondence: Iacopo Fabiani
| | - Alberto Giannoni
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Serena L'Abbate
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Alberto Aimo
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | | | - Giuseppe Vergaro
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | | | | | - Carlo Maria Cipolla
- Cardioncology Unit, Cardioncology and Second Opinion Division, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.), Milan, Italy
| | - Antonella Fedele
- Cardioncology Unit, Cardioncology and Second Opinion Division, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.), Milan, Italy
| | - Claudio Passino
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Michele Emdin
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Daniela Maria Cardinale
- Cardioncology Unit, Cardioncology and Second Opinion Division, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.), Milan, Italy
| |
Collapse
|
12
|
Ho CC, Wen PC, Yu WC, Hu YW, Yang CC. Pre-existing chronic kidney disease and hypertension increased the risk of cardiotoxicity among colorectal cancer patients treated with anticancer drugs. J Chin Med Assoc 2021; 84:877-884. [PMID: 34320515 DOI: 10.1097/jcma.0000000000000590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND This population-based study was to investigate the potential risk factors of cardiotoxicity among colorectal cancer (CRC) patients treated with anticancer drugs. METHODS This was a retrospective cohort study using the National Health Insurance Research Database to identify the CRC patients receiving chemotherapy (CT) alone or CT combined with targeted therapies between 2000 and 2013. The patients enrolled were those who had the first diagnosis of CRC established ≥20 years and had no cancer history three years before the incident diagnosis of CRC. The outcomes of cardiotoxicity were defined by the diagnosis of acute myocarditis, cardiomyopathy, heart failure, hypertensive heart disease, and so on. RESULTS A total of 11 819 CRC patients were identified and 3781 were eligible; 556 (14.7%) patients developed cardiotoxicity after receiving anticancer treatment. Patients showed a similar risk of having primary outcome (hazard ratio [HR], 0.7; p = 0.3662) between CT and CT combined with targeted therapy groups, whereas the risk of developing secondary outcome was significantly different between the two groups (HR, 0.7; p = 0.0339). The hazard was found to be increased with age (60-69, HR 2.1, p = 0.0236; 70-79, HR 3.3, p = 0.0003; and ≥80, HR 3.7, p < 0.0001). CRC patients who had a prior history of hypertension exhibited a higher risk than those without hypertension (HR 1.6, p < 0.0001). The hazard of having cardiotoxicity among patients with a prior history of severe chronic kidney disease was 2.4 times than that in those without renal dysfunction, regardless of the stage of cancer (HR 2.4, p < 0.0001). CONCLUSION CRC patients over 60 years of age run a higher risk of developing cardiotoxicity when treated with anticancer drugs. For CRC patients who have a previous history of hypertension or chronic kidney disease, physicians must be careful in evaluating the risk of anticancer drugs-related cardiotoxicity. Prescribe drugs may prevent cardiotoxicity if necessary.
Collapse
Affiliation(s)
- Chin-Chin Ho
- Department of Pharmacy, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Pharmacy, Taipei Medical University, Taipei, Taiwan, ROC
- Institute of Environmental and Occupational Health Sciences, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Pin-Chun Wen
- Institute of Environmental and Occupational Health Sciences, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Wen-Chung Yu
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Yu-Wen Hu
- Division of Radiation Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Institute of Public Health, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Chen-Chang Yang
- Institute of Environmental and Occupational Health Sciences, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Division of Clinical Toxicology and Occupational Medicine, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| |
Collapse
|
13
|
Abstract
PURPOSE OF REVIEW To present a method enabling in vivo quantification of tissue membrane potential (ΔΨT), a proxy of mitochondrial membrane potential (ΔΨm), to review the origin and role of ΔΨm, and to highlight potential applications of myocardial ΔΨT imaging. RECENT FINDINGS Radiolabelled lipophilic cations have been used for decades to measure ΔΨm in vitro. Using similar compounds labeled with positron emitters and appropriate compartment modeling, this technique now allows in vivo quantification of ΔΨT with positron emission tomography. Studies have confirmed the feasibility of measuring myocardial ΔΨT in both animals and humans. In addition, ΔΨT showed very low variability among healthy subjects, suggesting that this method could allow detection of relatively small pathological changes. In vivo assessment of myocardial ΔΨT provides a new tool to study the pathophysiology of cardiovascular diseases and has the potential to serve as a new biomarker to assess disease stage, prognosis, and response to therapy.
Collapse
|
14
|
Lam CK, Wu JC. Clinical Trial in a Dish: Using Patient-Derived Induced Pluripotent Stem Cells to Identify Risks of Drug-Induced Cardiotoxicity. Arterioscler Thromb Vasc Biol 2021; 41:1019-1031. [PMID: 33472401 PMCID: PMC11006431 DOI: 10.1161/atvbaha.120.314695] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Drug-induced cardiotoxicity is a significant clinical issue, with many drugs in the market being labeled with warnings on cardiovascular adverse effects. Treatments are often prematurely halted when cardiotoxicity is observed, which limits their therapeutic potential. Moreover, cardiotoxicity is a major reason for abandonment during drug development, reducing available treatment options for diseases and creating a significant financial burden and disincentive for drug developers. Thus, it is important to minimize the cardiotoxic effects of medications that are in use or in development. To this end, identifying patients at a higher risk of developing cardiovascular adverse effects for the drug of interest may be an effective strategy. The discovery of human induced pluripotent stem cells has enabled researchers to generate relevant cell types that retain a patient's own genome and examine patient-specific disease mechanisms, paving the way for precision medicine. Combined with the rapid development of pharmacogenomic analysis, the ability of induced pluripotent stem cell-derivatives to recapitulate patient-specific drug responses provides a powerful platform to identify subsets of patients who are particularly vulnerable to drug-induced cardiotoxicity. In this review, we will discuss the current use of patient-specific induced pluripotent stem cells in identifying populations who are at risk to drug-induced cardiotoxicity and their potential applications in future precision medicine practice. Graphic Abstract: A graphic abstract is available for this article.
Collapse
Affiliation(s)
- Chi Keung Lam
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA
- Department of Biological Sciences, University of Delaware, Newark, DE
| | - Joseph C. Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA
- Department of Radiology, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
15
|
Shin W, Lee S, Lim MC, Jung J, Kim HJ, Cho H. Incidence of venous thromboembolism after standard treatment in patients with epithelial ovarian cancer in Korea. Cancer Med 2021; 10:2045-2053. [PMID: 33638309 PMCID: PMC7957187 DOI: 10.1002/cam4.3797] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 12/09/2020] [Accepted: 01/29/2021] [Indexed: 11/27/2022] Open
Abstract
Background Venous thromboembolism (VTE) is a hospital‐associated severe complication that may adversely affect patient prognosis. In this study, we evaluated the incidence of VTE and its risk factors in patients with epithelial ovarian cancer (EOC). Methods We retrospectively analyzed the electronic health record data of 1268 patients with EOC who received primary treatment at the National Cancer Center, Korea between January 2007 and December 2017 to identify patients who developed VTE. Demographic, clinical, and surgical characteristics of these patients were ascertained. Competing risks analyses were performed to estimate the cumulative incidence of VTE according to the treatment type. The associations between putative risk factors and the incidence of VTE were evaluated using the Fine–Gray regression models accounting for competing risks of death. Results VTE was the most prevalent cardiovascular event, found in 9.6% (n = 122) of all patients. Of these VTE events, 115 (94.3%) occurred within 2 years of EOC diagnosis. Advanced cancer stage at diagnosis (distant vs. localized, hazards ratio [HR])= 14.49, p = 0.015) and extended hospital stay (≥15 days, HR =3.87, p = 0.004) were associated with the incidence of VTE. There was no significant difference in the cumulative incidence of VTE between primary cytoreductive surgery followed by adjuvant chemotherapy and neoadjuvant chemotherapy followed by interval cytoreductive surgery (HR =0.81, p = 0.390). Conclusions Approximately 10% of patients with EOC were diagnosed with VTE, which was the most common cardiovascular disease found in this study. The assessment of VTE risks in patients with advanced‐stage EOC with an extended hospital stay is needed to facilitate adequate prophylactic treatment.
Collapse
Affiliation(s)
- Wonkyo Shin
- Department of Cancer Control and Population Health, National Cancer Center Graduate School of Cancer Science and Policy, Goyang, Republic of Korea.,Center for Gynecologic Cancer, Research Institute and Hospital, National Cancer Center, Goyang, Republic of Korea
| | - Sanghee Lee
- Department of Cancer Control and Population Health, National Cancer Center Graduate School of Cancer Science and Policy, Goyang, Republic of Korea
| | - Myong Cheol Lim
- Department of Cancer Control and Population Health, National Cancer Center Graduate School of Cancer Science and Policy, Goyang, Republic of Korea.,Center for Gynecologic Cancer, Research Institute and Hospital, National Cancer Center, Goyang, Republic of Korea.,Division of Tumor Immunology, Research Institute and Hospital, National Cancer Center, Goyang, Republic of Korea.,Center for Clinical Trials, Research Institute and Hospital, National Cancer Center, Goyang, Republic of Korea
| | - Jipmin Jung
- Cancer Big Data Center, National Cancer Control Institute, National Cancer Center, Goyang, Republic of Korea
| | - Hak Jin Kim
- Branch of Cardiology, Department of Internal Medicine, National Cancer Center, Goyang, Republic of Korea
| | - Hyunsoon Cho
- Department of Cancer Control and Population Health, National Cancer Center Graduate School of Cancer Science and Policy, Goyang, Republic of Korea.,Division of Cancer Registration and Surveillance, National Cancer Center, Goyang, Republic of Korea
| |
Collapse
|
16
|
Trybus W, Król T, Trybus E, Stachurska A, Król G. The potential antitumor effect of chrysophanol in relation to cervical cancer cells. J Cell Biochem 2021; 122:639-652. [PMID: 33417255 DOI: 10.1002/jcb.29891] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 12/18/2020] [Accepted: 12/28/2020] [Indexed: 12/22/2022]
Abstract
Chrysophanol is an anthraquinone with proven antitumor activity against several tumor cell lines. However, its effect on cervical cancer cells is still unknown. Therefore, HeLa cells were exposed to various concentrations of chrysophanol and then subjected to biochemical, ultrastructural, and morphological analysis. It has been shown using flow cytometry and MTT reduction assay that chrysophanol has been shown to inhibit cell viability and arrest cells in the G2/M phase of the cell cycle. Using Annexin V/propidium iodide staining, a significant increase in apoptosis was found after chrysophanol treatment on HeLa cells, and this process was mediated by caspases 3/7 with a clear inactivation of the antiapoptotic Bcl-2 family protein. However, the demonstrated increased number of cells with double-stranded DNA breaks suggests that chrysophanol also causes DNA damage. By means of electron and fluorescence microscopy, a clear effect of chrysophanol on the intensification of degradation processes, on changes in the structure of the nucleus, endoplasmic reticulum and mitochondria was demonstrated. The changes visible in the mitochondria may be related to the increase in the level of free radicals induced by chrysophanol, which induces apoptosis, inter alia, by increasing the permeability of mitochondrial membranes. The range of observed changes depended on the concentration of anthraquinone was tested.
Collapse
Affiliation(s)
- Wojciech Trybus
- Laboratory of Medical Biology, Institute of Biology, Jan Kochanowski University, Kielce, Poland
| | - Teodora Król
- Laboratory of Medical Biology, Institute of Biology, Jan Kochanowski University, Kielce, Poland
| | - Ewa Trybus
- Laboratory of Medical Biology, Institute of Biology, Jan Kochanowski University, Kielce, Poland
| | - Anna Stachurska
- Department of Immunohematology, Medical Centre of Postgraduate Education, Warsaw, Poland
| | - Grzegorz Król
- Faculty of Management, University of Warsaw, Warsaw, Poland
| |
Collapse
|
17
|
Monitoring of anthracycline-induced myocardial injury using serial cardiac magnetic resonance: An animal study. Int J Cardiol 2020; 328:111-116. [PMID: 33359332 DOI: 10.1016/j.ijcard.2020.12.040] [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: 07/21/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 11/23/2022]
Abstract
PURPOSE To assess the feasibility of using comprehensive serial cardiovascular MR (CMR) to evaluate bilateral ventricle mechanical changes and myocardial tissue characteristics, as well as correlations between the serial CMR and histology in a beagle model of anthracycline-induced cardiotoxicity. METHODS This animal study was approved by the institutional review board. Serial CMR imaging was performed in a total of fifteen beagles at baseline (n = 15), at week 16 (n = 10) and week 24 (n = 7) post-anthracycline. Feature-tracking CMR (FT-CMR) was applied to measure bilateral ventricular (left ventricle (LV) and right ventricle (RV)) global peak strain including radial (GRS), circumferential (GCS) and longitudinal (GLS) strain. The changes in strain, LV/RV functional parameters, native T1, extracellular volume fraction (ECV) and collagen volume fraction (CVF) were calculated. RESULTS Compared to baseline at weeks 16 and 24, significantly decreases to LV-GLS and native T1 were observed, while ECV significantly increased (all P < 0.05). LVEF significantly decreased and LV-EDV/ESV significantly increased at week 16 compared to baseline (all P < 0.05), but no further progression was seen at week 24. RV-GLS significantly decreased at week 16, but no further progression was seen at week 24, while RVEF was different until week 24. CVF increased significantly during modeling. Native T1 and ECV showed positive correlation with CVF (r = 0.645, P < 0.001), while LV-GLS showed negative correlation with CVF (r = -0.736, P < 0.05). CONCLUSION Cardiotoxicity affects the RV slightly and less progressively than the LV. FT-CMR-based GLS, native T1 and ECV may potentially be used as imaging biomarkers for early monitoring of anthracycline-induced myocardial Injury.
Collapse
|
18
|
Saltalamacchia G, Frascaroli M, Bernardo A, Quaquarini E. Renal and Cardiovascular Toxicities by New Systemic Treatments for Prostate Cancer. Cancers (Basel) 2020; 12:cancers12071750. [PMID: 32630204 PMCID: PMC7407336 DOI: 10.3390/cancers12071750] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/19/2020] [Accepted: 06/25/2020] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer (PC) is the most common male cancer in Western Countries. In recent years, the treatment of relapsed or metastatic disease had benefited by the introduction of a variety of new different drugs. In consideration of the relative long survival of PC patients, side effects of these drugs must be considered and monitored. In this review, we analyzed the newly developed therapies for PC treatment, describing the mechanism of action, the metabolism and latest clinical trials that led to the approval of these drugs in clinical practice. We then evaluated the cardiovascular and renal side effects from pivotal phase III and II studies and meta-analyses. Cardiovascular side effects are the most frequent, in particular hypertension, while renal toxicity is rarer and not well described in literature. Therefore, there is a need to better define the effects of these therapies, in order to personalize patient treatment on the basis of their comorbidities and preferences, in addition to their symptoms and disease load.
Collapse
Affiliation(s)
- Giuseppe Saltalamacchia
- Operative Unit of Medical Oncology, IRCCS Istituti Clinici Scientifici Maugeri, 27100 Pavia, Italy; (G.S.); (A.B.)
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy
| | - Mara Frascaroli
- Operative Unit of Translational Oncology, IRCCS Istituti Clinici Scientifici Maugeri, 27100 Pavia, Italy;
| | - Antonio Bernardo
- Operative Unit of Medical Oncology, IRCCS Istituti Clinici Scientifici Maugeri, 27100 Pavia, Italy; (G.S.); (A.B.)
| | - Erica Quaquarini
- Operative Unit of Medical Oncology, IRCCS Istituti Clinici Scientifici Maugeri, 27100 Pavia, Italy; (G.S.); (A.B.)
- Experimental Medicine School, University of Pavia, 27100 Pavia, Italy
- Correspondence: ; +39-0382-592671
| |
Collapse
|
19
|
Chiba K, Ishizaka T, Yoshimatsu Y, Mikamoto K, Maeda Y, Iguchi T, Shirai M, Yamaguchi T, Goto K, Sakurai K, Tamai S, Kataoka H, Hasegawa M, Mori K. Comprehensive analysis of cardiac function, blood biomarkers and histopathology for milrinone-induced cardiotoxicity in cynomolgus monkeys. J Pharmacol Toxicol Methods 2020; 103:106870. [PMID: 32353509 DOI: 10.1016/j.vascn.2020.106870] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 04/13/2020] [Accepted: 04/22/2020] [Indexed: 01/08/2023]
Abstract
The objective of this study was to elucidate the underlying cardiotoxic mechanism of milrinone, a cAMP phosphodiesterase 3 inhibitor, by evaluating cardiac functions, blood biomarkers including cardiac troponin I (cTnI), microRNAs (miR-1, miR-133a and miR-499a) and various endogenous metabolites, and histopathology in conscious cynomolgus monkeys. Milrinone at doses of 0, 3 and 30 mg/kg were orally administered to monkeys (n = 3-4/group), and the endpoints were evaluated 1 to 24 h post-dosing. Milrinone caused myocardial injuries characterized by myocardial degeneration/necrosis, cell infiltration and hemorrhage 24 h after drug administration. Cardiac functional analysis revealed that milrinone dose-dependently increased the maximum upstroke velocity of the left ventricular pressure and heart rate, and decreased the QA interval and systemic blood pressure 1-4 h post-dosing, being associated with pharmacological action of the drug. In the blood biomarker analysis, only plasma cTnI was dose-dependently increased 4-7 h after drug administration, suggesting that cTnI is the most sensitive biomarker for early detection of milrinone-induced myocardial injuries. In the metabolomics analysis, high dose of milrinone induced transient changes in lipid metabolism, amino acid utilization and oxidative stress, together with the pharmacological action of increased cAMP and lipolysis 1 h post-dosing before the myocardial injuries were manifested by increased cTnI levels. Taken together, milrinone showed acute positive inotropic and multiple metabolic changes including excessive pharmacological actions, resulting in myocardial injuries. Furthermore, a comprehensive analysis of cardiac functions, blood biomarkers and histopathology can provide more appropriate information for overall assessment of preclinical cardiovascular safety.
Collapse
Affiliation(s)
- Katsuyoshi Chiba
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan.
| | - Tomomichi Ishizaka
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Yu Yoshimatsu
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Kei Mikamoto
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Yu Maeda
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Takuma Iguchi
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Makoto Shirai
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Takashi Yamaguchi
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Koichi Goto
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Ken Sakurai
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Satoshi Tamai
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Hiroko Kataoka
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Miki Hasegawa
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Kazuhiko Mori
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| |
Collapse
|
20
|
Choudhury S, Chatterjee S, Pulakhandam SBK. Acute cardiogenic shock with paclitaxel use in a lung carcinoma patient. Lung India 2019; 36:568-569. [PMID: 31670314 PMCID: PMC6852215 DOI: 10.4103/lungindia.lungindia_298_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Shouvik Choudhury
- Department of Pharmacology, Burdwan Medical College, Burdwan, West Bengal, India
| | - Suparna Chatterjee
- Departments of Pharmacology, Institute of Postgraduate Medical Education and Research, Kolkata, West Bengal, India
| | - Sampath B K Pulakhandam
- Department of Pharmacology, Institute of Postgraduate Medical Education and Research, Kolkata, West Bengal, India
| |
Collapse
|
21
|
Lage R, Cebro-Márquez M, Rodríguez-Mañero M, González-Juanatey JR, Moscoso I. Omentin protects H9c2 cells against docetaxel cardiotoxicity. PLoS One 2019; 14:e0212782. [PMID: 30794687 PMCID: PMC6386316 DOI: 10.1371/journal.pone.0212782] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 02/08/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Association between obesity and cardiovascular diseases is well known, however increased susceptibility of obese patients to develop several cancer types is not so commonly known. Current data suggest that poorer overall survival in cancer patients might be associated to non-cancer-related causes such as higher risk of cardiotoxicity in obese patients treated with chemotherapeutic agents. Omentin, a novel adipokine decreased in obesity, is actually in the spotlight due to its favourable effects on inflammation, glucose homeostasis and cardiovascular diseases. Also, recent data showed that in vitro anthracycline-induced cardiomyocyte apoptosis is counteracted by omentin suggesting its cardioprotective role. OBJECTIVE Our aim was to evaluate omentin effects against docetaxel toxicity. RESULTS Our data indicate that omentin inhibits docetaxel-induced viability loss and that increased viability is associated to decreased caspase-3 expression and cell death. Although omentin reduces NOX4 expression, it failed to reduce docetaxel-induced reactive oxygen species production. Our results indicate that omentin decreases docetaxel-induced endoplasmic reticulum stress, suggesting that cardioprotective role might be associated to ERS inhibition. CONCLUSION These data suggest that omentin treatment may contribute to decrease susceptibility to DTX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Ricardo Lage
- Cardiology Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela and Health Research Institute, University Clinical Hospital of Santiago de Compostela—Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - María Cebro-Márquez
- Cardiology Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela and Health Research Institute, University Clinical Hospital of Santiago de Compostela—Santiago de Compostela, Spain
| | - Moisés Rodríguez-Mañero
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Department of Cardiology and Coronary Unit, University Clinical Hospital of Santiago de Compostela, Santiago de Compostela, Spain
| | - José Ramón González-Juanatey
- Cardiology Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela and Health Research Institute, University Clinical Hospital of Santiago de Compostela—Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Department of Cardiology and Coronary Unit, University Clinical Hospital of Santiago de Compostela, Santiago de Compostela, Spain
| | - Isabel Moscoso
- Cardiology Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela and Health Research Institute, University Clinical Hospital of Santiago de Compostela—Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| |
Collapse
|
22
|
Corley AM, Sullivan MJ, Friedman SE, O'Rourke DJ, Palac RT, Gemignani AS. Relation of Venous Thromboembolism Risk to Ischemic Stroke Risk in Hospitalized Patients with Cancer. Am J Cardiol 2019; 123:679-683. [PMID: 30528279 DOI: 10.1016/j.amjcard.2018.11.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 10/31/2018] [Accepted: 11/05/2018] [Indexed: 12/21/2022]
Abstract
Patients with cancer are at increased risk for venous thromboembolism (VTE). However, the relationship of cancer type to the risk of arterial thrombosis in patients with high VTE risk has not been described. The goal of this study is to determine the rate of arterial thrombosis in patients with different types of solid tumors stratified by VTE risk. Using the 2012 National Inpatient Sample, we identified 373,789 hospitalizations involving patients ≥18 years associated with solid tumors, stratified by type. Data were collected on clinical characteristics, VTE (deep vein thrombosis [DVT] and pulmonary embolism [PE]), and arterial thrombosis (primary diagnosis of myocardial infarction [MI] and ischemic stroke). Subjects with solid tumors (stages I to IV) were stratified by VTE risk - high versus low. Certain solid tumor types (esophageal, lung, melanoma, ovarian, pancreatic, stomach, and uterine) were found to be associated with a higher rate of VTE compared with other cancer types (6.8% vs 3.9%, p < 0.001). Multivariate analysis applied to the high VTE risk group showed no increased risk for MI (odds ratio [OR] 0.93, p = 0.74), however, the rate of ischemic stroke was increased (OR 1.22, p < 0.001). Those in the high VTE risk group who had metastatic disease were at higher risk for arterial thrombosis (MI OR 1.35, p < 0.001, ischemic stroke OR 2.43, p < 0.001). In conclusion, different cancer types are associated with increased risk of both venous and arterial thrombosis and the risk is further increased by the presence of metastatic disease.
Collapse
Affiliation(s)
- Alyssa M Corley
- Department of Internal Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Malachy J Sullivan
- Department of Internal Medicine, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire
| | - Scott E Friedman
- Division of Cardiovascular Medicine, White River Junction VA Medical Center, White River Junction, Vermont; Department of Medicine, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Daniel J O'Rourke
- Division of Cardiovascular Medicine, White River Junction VA Medical Center, White River Junction, Vermont; Department of Medicine, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Robert T Palac
- Division of Cardiovascular Medicine, White River Junction VA Medical Center, White River Junction, Vermont
| | - Anthony S Gemignani
- Division of Cardiovascular Medicine, White River Junction VA Medical Center, White River Junction, Vermont; Department of Medicine, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire.
| |
Collapse
|
23
|
Georgiadis N, Tsarouhas K, Tsitsimpikou C, Vardavas A, Rezaee R, Germanakis I, Tsatsakis A, Stagos D, Kouretas D. Pesticides and cardiotoxicity. Where do we stand? Toxicol Appl Pharmacol 2018; 353:1-14. [PMID: 29885332 DOI: 10.1016/j.taap.2018.06.004] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 05/30/2018] [Accepted: 06/03/2018] [Indexed: 01/11/2023]
Abstract
Cardiovascular diseases are among the most significant causes of mortality in humans. Pesticides toxicity and risk for human health are controlled at a European level through a well-developed regulatory network, but cardiotoxicity is not described as a separate hazard class. Specific classification criteria should be developed within the frame of Regulation (EC) No 1272/2008 in order to classify chemicals as cardiotoxic, if applicable to avoid long-term cardiovascular complications. The aim of this study was to review the cardiac pathology and function impairment due to exposure to pesticides (i.e. organophosphates, organothiophisphates, organochlorines, carbamates, pyrethroids, dipyridyl herbicides, triazoles, triazines) based on both animal and human data. The majority of human data on cardiotoxicity of pesticides come from poisoning cases and epidemiological data. Several cardiovascular complications have been reported in animal models including electrocardiogram abnormalities, myocardial infarction, impaired systolic and diastolic performance, functional remodeling and histopathological findings, such as haemorrhage, vacuolisation, signs of apoptosis and degeneration.
Collapse
Affiliation(s)
- Nikolaos Georgiadis
- European Food Safety Authority, Via Carlo Magno 1A, 43126 Parma, Italy; Department of Biochemistry- Biotechnology, School of Health Sciences, University of Thessaly, Viopolis, Larissa 41500, Greece
| | - Konstantinos Tsarouhas
- Department of Cardiology, University Hospital of Larissa, Mezourlo, Larissa 41110, Greece
| | | | - Alexandros Vardavas
- Laboratory of Toxicology, Medical School, University of Crete, Voutes, Heraklion, 71003 Crete, Greece
| | - Ramin Rezaee
- Clinical Research Unit, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ioannis Germanakis
- Paediatric Cardiology Unit, Department of Paediatrics, University Hospital Voutes, Heraklion, 71409 Crete, Greece
| | - Aristides Tsatsakis
- Laboratory of Toxicology, Medical School, University of Crete, Voutes, Heraklion, 71003 Crete, Greece
| | - Dimitrios Stagos
- Department of Biochemistry- Biotechnology, School of Health Sciences, University of Thessaly, Viopolis, Larissa 41500, Greece
| | - Demetrios Kouretas
- Department of Biochemistry- Biotechnology, School of Health Sciences, University of Thessaly, Viopolis, Larissa 41500, Greece.
| |
Collapse
|
24
|
Tuzovic M, Herrmann J, Iliescu C, Marmagkiolis K, Ziaeian B, Yang EH. Arterial Thrombosis in Patients with Cancer. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2018; 20:40. [PMID: 29627870 PMCID: PMC7658957 DOI: 10.1007/s11936-018-0635-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Cancer is a common cause of morbidity and mortality in the USA. While the association between venous thrombosis and malignancy is well established, arterial thrombosis has more recently been recognized as a serious complication of cancer and certain chemotherapeutic agents. This review aims to summarize the most recent literature regarding the incidence and risk factors for cancer-related arterial thrombosis, understand the pathophysiologic mechanisms of thrombosis, and highlight the specific diagnostic and treatment considerations relevant to cancer patients. RECENT FINDINGS Based on a recent study looking at the Surveillance, Epidemiology, and End Results (SEER) database, the incidence of arterial thromboembolic events (ATEs) in patients with cancer at 6 months is 4.7%; the presence of an ATE is predictive of worse outcomes. Certain drugs such as platinum-based agents, vascular endothelial growth factor inhibitors, tyrosine kinase inhibitors, and taxanes have been associated with high rates of ATEs. Increased platelet reactivity appears crucial to development of arterial thrombosis in cancer patients. Cancer patients have an increased risk of arterial thrombosis that is likely due to both a cancer-associated procoagulant state as well as the adverse effects of certain chemotherapeutic agents. Treatment of arterial thromboembolism in cancer patients typically requires a multidisciplinary approach in part due to high rates of thrombocytopenia and stent thrombosis in the setting of percutaneous interventions. More studies are needed to investigate optimal prophylaxis, surveillance strategies, and treatments of cancer-related arterial thromboembolic disease.
Collapse
Affiliation(s)
- Mirela Tuzovic
- UCLA Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California at Los Angeles, 100 Medical Plaza, Suite 630, Los Angeles, CA, 90095, USA
| | - Joerg Herrmann
- Division of Cardiovascular Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Cezar Iliescu
- Division of Cardiology, Department of Medicine, MD Anderson Cancer Center, University of Texas at Houston, Houston, TX, USA
| | | | - Boback Ziaeian
- UCLA Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California at Los Angeles, 100 Medical Plaza, Suite 630, Los Angeles, CA, 90095, USA
| | - Eric H Yang
- UCLA Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California at Los Angeles, 100 Medical Plaza, Suite 630, Los Angeles, CA, 90095, USA.
| |
Collapse
|
25
|
Dexrazoxane added to doxorubicin-based adjuvant chemotherapy of breast cancer: a retrospective cohort study with a comparative analysis of toxicity and survival. Anticancer Drugs 2017; 28:787-794. [PMID: 28562379 DOI: 10.1097/cad.0000000000000514] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Dexrazoxane is indicated as a cardioprotective agent for patients receiving doxorubicin who are at increased risk for cardiotoxicity. Concerns have been raised on the use of dexrazoxane, particularly in adjuvant therapy, because of the risk of interference with the antitumor effect of doxorubicin. Two meta-analyses in metastatic breast cancer have rejected this hypothesis, but have shown an apparent increase in the severity of myelosuppression when dexrazoxane is used. Here, we analyzed retrospectively a cohort of our institute database to assess whether the addition of dexrazoxane causes more bone marrow suppression in breast cancer patients receiving doxorubicin-based adjuvant therapy. The secondary objectives were assessment of the incidence of febrile neutropenia, dose-schedule modifications, recorded cardiac events or cardiac test abnormalities, and overall survival. Eight hundred and twenty-two female patients who received adjuvant (or neoadjuvant) doxorubicin and cyclophosphamide for breast cancer between 2001 and 2013 were included. One hundred and four of these patients also received dexrazoxane concurrently with the adjuvant treatment. Hospital records and, when accessible, community clinic records were reviewed. The median follow-up duration was 7 years for patients receiving dexrazoxane and 7.5 years for patients not receiving dexrazoxane. 85.6% of patients were alive at data lock. Compared with the nondexrazoxane group, patients who received dexrazoxane were older (median age at diagnosis 59 vs. 52 years) and more likely to receive dose-dense AC therapy (73 vs. 59%) and adjuvant trastuzumab treatment (29 vs. 15%). Compared with the nondexrazoxane group, dexrazoxane treatment was associated with a higher rate of hematological side effects: leukopenia (48 vs. 39%), neutropenia (45 vs. 31%, P=0.003), anemia (86 vs. 73%, P=0.005), and thrombocytopenia (37 vs. 22%, P=0.001). There were more febrile neutropenia hospitalizations (20 vs. 10%, P=0.001) and dose reductions (22 vs. 8%, P<0.001) in the dexrazoxane group, but no significant difference in the incidence of treatment delays or cancellations. The incidence of cardiac events was the same in both treatment groups with and without dexrazoxane. There was a nonsignificantly lower mortality rate in the dexrazoxane group (9.6%) compared with the nondexrazoxane group (15.0%) at data lock. Adding dexrazoxane to doxorubicin in adjuvant therapy patients leads to higher rates of bone marrow suppression in all blood components, as well as more febrile neutropenia events, and dose reductions. No differences in events defined as cardiac toxicities were detected. Dexrazoxane had no detrimental effect on survival, despite the higher hematological toxicity, the older median age, and the higher prevalence of HER2-positive disease in the dexrazoxane group.
Collapse
|
26
|
Klee NS, McCarthy CG, Martinez-Quinones P, Webb RC. Out of the frying pan and into the fire: damage-associated molecular patterns and cardiovascular toxicity following cancer therapy. Ther Adv Cardiovasc Dis 2017; 11:297-317. [PMID: 28911261 PMCID: PMC5933669 DOI: 10.1177/1753944717729141] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 08/09/2017] [Indexed: 12/18/2022] Open
Abstract
Cardio-oncology is a new and rapidly expanding field that merges cancer and cardiovascular disease. Cardiovascular disease is an omnipresent side effect of cancer therapy; in fact, it is the second leading cause of death in cancer survivors after recurrent cancer. It has been well documented that many cancer chemotherapeutic agents cause cardiovascular toxicity. Nonetheless, the underlying cause of cancer therapy-induced cardiovascular toxicity is largely unknown. In this review, we discuss the potential role of damage-associated molecular patterns (DAMPs) as an underlying contributor to cancer therapy-induced cardiovascular toxicity. With an increasing number of cancer patients, as well as extended life expectancy, understanding the mechanisms underlying cancer therapy-induced cardiovascular disease is of the utmost importance to ensure that cancer is the only disease burden that cancer survivors have to endure.
Collapse
Affiliation(s)
- Nicole S. Klee
- Department of Physiology, Medical College of Georgia at Augusta University, 1120 15 Street, Augusta, GA 30912, USA
| | - Cameron G. McCarthy
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Patricia Martinez-Quinones
- Departments of Physiology and Surgery, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - R. Clinton Webb
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| |
Collapse
|
27
|
Cardiac Nonmyocyte Cell Functions and Crosstalks in Response to Cardiotoxic Drugs. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:1089359. [PMID: 29201269 PMCID: PMC5671742 DOI: 10.1155/2017/1089359] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/24/2017] [Indexed: 01/06/2023]
Abstract
The discovery of the molecular mechanisms involved in the cardiac responses to anticancer drugs represents the current goal of cardio-oncology research. The oxidative stress has a pivotal role in cardiotoxic responses, affecting the function of all types of cardiac cells, and their functional crosstalks. Generally, cardiomyocytes are the main target of research studies on cardiotoxicity, but recently the contribution of the other nonmyocyte cardiac cells is becoming of growing interest. This review deals with the role of oxidative stress, induced by anticancer drugs, in cardiac nonmyocyte cells (fibroblasts, vascular cells, and immune cells). The alterations of functional interplays among these cardiac cells are discussed, as well. These interesting recent findings increase the knowledge about cardiotoxicity and suggest new molecular targets for both diagnosis and therapy.
Collapse
|
28
|
Santoni M, Guerra F, Conti A, Lucarelli A, Rinaldi S, Belvederesi L, Capucci A, Berardi R. Incidence and risk of cardiotoxicity in cancer patients treated with targeted therapies. Cancer Treat Rev 2017; 59:123-131. [PMID: 28818671 DOI: 10.1016/j.ctrv.2017.07.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 07/18/2017] [Accepted: 07/19/2017] [Indexed: 01/29/2023]
Abstract
BACKGROUND Cardiotoxicityis a serious side effect of molecularly targeted agents. The purpose of this study was to evaluate the incidence and Relative Risk (RR) of developing all-grade and high-grade cardiotoxicity in patients with solid tumors receiving targeted agents through a revised meta-analysis of available clinical trials. METHODS The scientific literature regarding cardiotoxicity was extensively analyzed using MEDLINE, PubMed, Embase and Cochrane Central Register of Controlled Trials (CENTRAL). Eligible studies were selected according to PRISMA statement. Summary incidence, RR, and 95% CIs were calculated using random-effects or fixed-effects models based on the heterogeneity of selected studies. RESULTS Our search yielded a total of 4998 clinical studies; of them, 31 trials were finally considered for this meta-analysis. A total of 28,538 patients were included; 7995 of these patients had breast cancer (28%), 6151 (22%) prostate cancer and 14,392 (50%) were treated for other malignancies. The highest RR of high-grade events was observed with Vandetanib (RR=7.71, 95% CI 1.04-56.99), followed by Ramucirumab (RR=5.0) and Aflibercept (RR=4.1). Grouping by drug category, the highest incidence of high-grade cardiotoxicity was shown by anti-VEGFR-TKIs (RR 5.62, 95% CI 1.49-21.24) and anti-VEGF mAbs/VEGF-trap (RR 1.82, 95% CI 1.24-2.69). Grouping by tumor type, the highest incidence of cardiotoxicity was observed in thyroid cancer (8%), followed by gastric cancer (4%). CONCLUSIONS Treatment with targeted agents in cancer patients is correlated with a significant increase in the risk of cardiotoxicity. Frequent clinical monitoring should be emphasized when using these and newer biological agents.
Collapse
Affiliation(s)
- Matteo Santoni
- Medical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I, GM Lancisi, G Salesi, Ancona, Italy.
| | - Federico Guerra
- Cardiology and Arrhythmology Clinic, Marche Polytechnic University, University Hospital "Umberto I - Lancisi - Salesi", Via Conca 71, Ancona 60126, Italy
| | - Alessandro Conti
- Urologia - Ospedale di Bressanone/Brixen Krankenhaus, Azienda Sanitaria dell'Alto Adige/Sanitätsbetrieb Südtirol, Via Dante 51, Bressanone (BZ), Italy
| | - Alessandra Lucarelli
- Medical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I, GM Lancisi, G Salesi, Ancona, Italy
| | - Silvia Rinaldi
- Medical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I, GM Lancisi, G Salesi, Ancona, Italy
| | - Laura Belvederesi
- Medical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I, GM Lancisi, G Salesi, Ancona, Italy
| | - Alessandro Capucci
- Cardiology and Arrhythmology Clinic, Marche Polytechnic University, University Hospital "Umberto I - Lancisi - Salesi", Via Conca 71, Ancona 60126, Italy
| | - Rossana Berardi
- Medical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I, GM Lancisi, G Salesi, Ancona, Italy
| |
Collapse
|
29
|
Lung cancer as a cardiotoxic state: a review. Med Oncol 2017; 34:159. [DOI: 10.1007/s12032-017-1012-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 07/22/2017] [Indexed: 12/30/2022]
|
30
|
Early Detection and Serial Monitoring of Anthracycline-Induced Cardiotoxicity Using T1-mapping Cardiac Magnetic Resonance Imaging: An Animal Study. Sci Rep 2017; 7:2663. [PMID: 28572614 PMCID: PMC5453985 DOI: 10.1038/s41598-017-02627-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 04/13/2017] [Indexed: 11/26/2022] Open
Abstract
A reliable, non-invasive diagnostic method is needed for early detection and serial monitoring of cardiotoxicity, a well-known side effect of chemotherapy. This study aimed to assess the feasibility of T1-mapping cardiac magnetic resonance imaging (CMR) for evaluating subclinical myocardial changes in a doxorubicin-induced cardiotoxicity rabbit model. Adult male New Zealand White rabbits were injected twice-weekly with doxorubicin and subjected to CMR on a clinical 3T MR system before and every 2–4 weeks post-drug administration. Native T1 and extracellular volume (ECV) values were measured at six mid-left ventricle (LV) and specific LV lesions. Histological assessments evaluated myocardial injury and fibrosis. Three pre-model and 11 post-model animals were included. Myocardial injury was observed from 3 weeks. Mean LV myocardium ECV values increased significantly from week 3 before LV ejection fraction decreases (week 6), and ECVs of the RV upper/lower insertion sites and papillary muscle exceeded those of the LV. The mean native T1 value in the mid-LV increased significantly increased from week 6, and LV myocardium ECV correlated strongly with the degree of fibrosis (r = 0.979, p < 0.001). Myocardial T1 mapping, particularly ECV values, reliably and non-invasively detected early cardiotoxicity, allowing serial monitoring of chemotherapy-induced cardiotoxicity.
Collapse
|
31
|
Lycorine inhibits breast cancer growth and metastasis via inducing apoptosis and blocking Src/FAK-involved pathway. SCIENCE CHINA-LIFE SCIENCES 2017; 60:417-428. [PMID: 28251459 DOI: 10.1007/s11427-016-0368-y] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 12/27/2016] [Indexed: 01/07/2023]
Abstract
Breast cancer is the most commonly diagnosed cancer type worldwide among women and more than 90% of patients die from tumor metastasis. Lycorine, a natural alkaloid, has been widely reported possessing potential efficacy against cancer proliferation and metastasis. In our study, the anti-tumor potency on breast cancer was evaluated in vitro and in vivo for the first time. Our results indicated that lycorine inhibited breast cancer cells growth, migration and invasion as well as induced their apoptosis. In in vivo study, lycorine not only suppressed breast tumor growth in xenograft models and inhibited breast tumor metastasis in MDA-MB-231 tail vein model. More importantly, we found lycorine had less toxicity than first-line chemotherapy drug paclitaxel at the same effective dose in vivo. Furthermore, on mechanism, lycorine inhibited tumor cell migration and invasion via blocking the Src/FAK (focal adhesion kinase)-involved pathway. In conclusion, our study implied lycorine was a potential candidate for the treatment of breast cancer by inhibition of tumor growth and metastasis.
Collapse
|
32
|
Tromp J, Steggink LC, Van Veldhuisen DJ, Gietema JA, van der Meer P. Cardio-Oncology: Progress in Diagnosis and Treatment of Cardiac Dysfunction. Clin Pharmacol Ther 2017; 101:481-490. [PMID: 28073142 DOI: 10.1002/cpt.614] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 12/19/2016] [Accepted: 01/06/2017] [Indexed: 12/11/2022]
Affiliation(s)
- J Tromp
- Department of Cardiology, University Medical Centre Groningen, University of Groningen, The Netherlands
| | - L C Steggink
- Department of Medical Oncology University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - D J Van Veldhuisen
- Department of Cardiology, University Medical Centre Groningen, University of Groningen, The Netherlands
| | - J A Gietema
- Department of Medical Oncology University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - P van der Meer
- Department of Cardiology, University Medical Centre Groningen, University of Groningen, The Netherlands
| |
Collapse
|
33
|
O’Farrell AC, Evans R, Silvola JMU, Miller IS, Conroy E, Hector S, Cary M, Murray DW, Jarzabek MA, Maratha A, Alamanou M, Udupi GM, Shiels L, Pallaud C, Saraste A, Liljenbäck H, Jauhiainen M, Oikonen V, Ducret A, Cutler P, McAuliffe FM, Rousseau JA, Lecomte R, Gascon S, Arany Z, Ky B, Force T, Knuuti J, Gallagher WM, Roivainen A, Byrne AT. A Novel Positron Emission Tomography (PET) Approach to Monitor Cardiac Metabolic Pathway Remodeling in Response to Sunitinib Malate. PLoS One 2017; 12:e0169964. [PMID: 28129334 PMCID: PMC5271313 DOI: 10.1371/journal.pone.0169964] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 12/25/2016] [Indexed: 01/17/2023] Open
Abstract
Sunitinib is a tyrosine kinase inhibitor approved for the treatment of multiple solid tumors. However, cardiotoxicity is of increasing concern, with a need to develop rational mechanism driven approaches for the early detection of cardiac dysfunction. We sought to interrogate changes in cardiac energy substrate usage during sunitinib treatment, hypothesising that these changes could represent a strategy for the early detection of cardiotoxicity. Balb/CJ mice or Sprague-Dawley rats were treated orally for 4 weeks with 40 or 20 mg/kg/day sunitinib. Cardiac positron emission tomography (PET) was implemented to investigate alterations in myocardial glucose and oxidative metabolism. Following treatment, blood pressure increased, and left ventricular ejection fraction decreased. Cardiac [18F]-fluorodeoxyglucose (FDG)-PET revealed increased glucose uptake after 48 hours. [11C]Acetate-PET showed decreased myocardial perfusion following treatment. Electron microscopy revealed significant lipid accumulation in the myocardium. Proteomic analyses indicated that oxidative metabolism, fatty acid β-oxidation and mitochondrial dysfunction were among the top myocardial signalling pathways perturbed. Sunitinib treatment results in an increased reliance on glycolysis, increased myocardial lipid deposition and perturbed mitochondrial function, indicative of a fundamental energy crisis resulting in compromised myocardial energy metabolism and function. Our findings suggest that a cardiac PET strategy may represent a rational approach to non-invasively monitor metabolic pathway remodeling following sunitinib treatment.
Collapse
Affiliation(s)
- Alice C. O’Farrell
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Rhys Evans
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Johanna M. U. Silvola
- Turku PET Centre, Turku University Hospital and Åbo Akademi University, Turku, Finland
| | - Ian S. Miller
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Emer Conroy
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Suzanne Hector
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
- Roche Innovation Center Basel, F Hoffman La Roche, Basel, Switzerland
| | | | - David W. Murray
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
- Oncomark Ltd, Dublin, Ireland
| | - Monika A. Jarzabek
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
- Roche Innovation Center Basel, F Hoffman La Roche, Basel, Switzerland
| | | | | | | | - Liam Shiels
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Celine Pallaud
- Roche Innovation Center Basel, F Hoffman La Roche, Basel, Switzerland
| | - Antti Saraste
- Turku PET Centre, Turku University Hospital and Åbo Akademi University, Turku, Finland
- Heart Center, Turku University Hospital and Åbo Akademi University, Turku, Finland
| | - Heidi Liljenbäck
- Turku PET Centre, Turku University Hospital and Åbo Akademi University, Turku, Finland
| | - Matti Jauhiainen
- Public Health Genomics Unit, National Institute for Health and Welfare, Helsinki, Finland
| | - Vesa Oikonen
- Turku PET Centre, Turku University Hospital and Åbo Akademi University, Turku, Finland
| | - Axel Ducret
- Roche Innovation Center Basel, F Hoffman La Roche, Basel, Switzerland
| | - Paul Cutler
- Roche Innovation Center Basel, F Hoffman La Roche, Basel, Switzerland
| | - Fionnuala M. McAuliffe
- UCD Obstetrics & Gynaecology, School of Medicine, University College, Dublin, National Maternity Hospital, Dublin, Ireland
| | | | | | | | - Zoltan Arany
- Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, United States of America
| | - Bonnie Ky
- Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, United States of America
| | - Thomas Force
- Vanderbilt University School of Medicine, Nashville, United States of America
| | - Juhani Knuuti
- Turku PET Centre, Turku University Hospital and Åbo Akademi University, Turku, Finland
| | - William M. Gallagher
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland
- Oncomark Ltd, Dublin, Ireland
| | - Anne Roivainen
- Turku PET Centre, Turku University Hospital and Åbo Akademi University, Turku, Finland
- Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Annette T. Byrne
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
- * E-mail:
| |
Collapse
|
34
|
Veccia A, Maines F, Kinspergher S, Galligioni E, Caffo O. Cardiovascular toxicities of systemic treatments of prostate cancer. Nat Rev Urol 2017; 14:230-243. [PMID: 28117849 DOI: 10.1038/nrurol.2016.273] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Prostate cancer is the most common cancer in men, with an incidence that is expected to increase in the coming years. Prostate cancer is usually diagnosed in men >65 years of age, thus the concurrent presence of cardiovascular diseases might influence the treatment, owing to the increased risk of cardiovascular mortality. The introduction of new drugs, such as abiraterone and enzalutamide for the management of metastatic disease has created further interest in treatment-related cardiovascular toxicities, although limited data from trials specifically designed to identify cardiovascular toxicities of these agents are currently available. The only available data are derived from published phase II-III study reports, expanded access or compassionate use programmes and meta-analyses of the effects of systemic therapies that are already approved for use in clinical practice or are in the early phases of development. These data are conflicting, although they seem to suggest that certain drugs are associated with an increased risk of cardiovascular adverse events. Clinical trial methodology could be improved by the enrolment of greater numbers of patients >65 years of age, and the use of comprehensive cardiological evaluations. Moreover, closer collaboration between oncologists and cardiologists is essential for the identification and/or management of cardiovascular adverse events in patients with prostate cancer.
Collapse
Affiliation(s)
- Antonello Veccia
- Medical Oncology Department, Santa Chiara Hospital, Largo Medaglie d'Oro 38100 Trento, Italy
| | - Francesca Maines
- Medical Oncology Department, Santa Chiara Hospital, Largo Medaglie d'Oro 38100 Trento, Italy
| | - Stefania Kinspergher
- Medical Oncology Department, Santa Chiara Hospital, Largo Medaglie d'Oro 38100 Trento, Italy
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Piazzale L.A. Scuro 10, 37124 Verona, Italy
| | - Enzo Galligioni
- Medical Oncology Department, Santa Chiara Hospital, Largo Medaglie d'Oro 38100 Trento, Italy
| | - Orazio Caffo
- Medical Oncology Department, Santa Chiara Hospital, Largo Medaglie d'Oro 38100 Trento, Italy
| |
Collapse
|
35
|
Tang XM, Chen H, Liu Y, Huang BL, Zhang XQ, Yuan JM, He X. The cardiotoxicity of cetuximab as single therapy in Chinese chemotherapy-refractory metastatic colorectal cancer patients. Medicine (Baltimore) 2017; 96:e5946. [PMID: 28099361 PMCID: PMC5279106 DOI: 10.1097/md.0000000000005946] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The cardiac safety of cetuximab, particularly as single approach, has not been investigated extensively. This trial was designed to evaluate the cardiac safety of cetuximab as salvage monotherapy in Chinese chemotherapy-refractory metastatic colorectal cancer (mCRC) patients.Cetuximab was administrated at an initial dose of 400 mg/mon day 1 (week 1), followed by a maintenance dose of 250 mg/m on day 1 of each 7-day cycle. Electrocardiograph (ECG), routine laboratory tests, and troponin I (TNI) Ultra were performed at baseline, during, and after the cetuximab therapy. The incidence of abnormal ECGs, elevated TNI Ultra, cardiac events, and noncardiac events were recorded and analyzed.TNI Ultra+ was found in 20 patients (32.3%) during the cetuximab therapy.TNI Ultra+ occurred more frequently in patients with more than 3 organs affected and accepted fourth or above lines of chemotherapy. The most frequent abnormal ECG was ST depression in 24 (38.7%) patients. The elevated TNI Ultra and abnormal ECGs could recover after the cetuximab therapy. The most of cardiac adverse events were mild and transient and the noncardiac adverse events were also consistent with the known safety profile for cetuximab.Cetuximab showed its cardiac safety as a single agent for chemotherapy-refractory mCRC patients. And TNI Ultra and ECG could be sensitive and convenient approaches for the surveillance of adverse events.
Collapse
Affiliation(s)
- Xue-Miao Tang
- Department of Ultrasound and Electrocardiogram
- State Key Laboratory of Oncology in Southern China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Hao Chen
- Department of Clinical Laboratory
- State Key Laboratory of Oncology in Southern China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yu Liu
- Department of Nursing, Sun Yat-Sen University Cancer Center
- State Key Laboratory of Oncology in Southern China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Bin-Lian Huang
- Department of Radiation Oncology
- State Key Laboratory of Oncology in Southern China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xiu-Quan Zhang
- Department of Ultrasound and Electrocardiogram
- State Key Laboratory of Oncology in Southern China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jian-Mei Yuan
- Department of Ultrasound and Electrocardiogram
- State Key Laboratory of Oncology in Southern China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xia He
- Department of Clinical Laboratory
- State Key Laboratory of Oncology in Southern China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| |
Collapse
|
36
|
|
37
|
Guerra F, Marchesini M, Contadini D, Menditto A, Morelli M, Piccolo E, Battelli N, Pistelli M, Berardi R, Cascinu S, Capucci A. Speckle-tracking global longitudinal strain as an early predictor of cardiotoxicity in breast carcinoma. Support Care Cancer 2016; 24:3139-3145. [PMID: 26923461 DOI: 10.1007/s00520-016-3137-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 02/22/2016] [Indexed: 02/07/2023]
Abstract
PURPOSE Recent development in anticancer therapies for breast carcinoma allowed an improvement in patients' survival, notwithstanding a parallel increase of cardiovascular morbidity. Cardiotoxicity of anticancer therapies represents a relevant problem due to its insidious onset and potentially irreversible cardiac damage. The aim of the present study was to test whether 2D speckle tracking analysis can help in predicting overt systolic dysfunction. METHODS A "real world" cohort of 69 patients with breast carcinoma undergoing adjuvant and/or neo-adjuvant chemotherapy was tested 2D-speckle tracking analysis before the beginning of chemotherapy and every 3 months for 1 year. Clinical data, 12-lead ECGs, and lab tests were collected according to the same visit protocol. RESULTS Over 1-year follow-up, 19 patients (27 %) developed cardiac dysfunction according to the CREC criteria, with an average onset time from enrolment of 6.8 months. A global longitudinal strain (GLS) threshold ≥-16 % at 3 months from chemotherapy was able to predict subsequent systolic dysfunction development with high sensitivity (80 %) and specificity (90 %) and a negative predictive value of 92 %. After the introduction of cardioprotective drugs, left ventricular ejection fraction (LVEF) progressively recovered, while alterations of GLS persisted at 1-year follow-up. CONCLUSIONS Strain imaging with 2D speckle tracking allows the identification of patients at low-risk for chemotherapy-related systolic dysfunction and can help optimizing resources allocations and improving follow-up quality. GLS can also provide a more accurate prognostic index of resolved systolic dysfunction when compared to standard LVEF.
Collapse
Affiliation(s)
- Federico Guerra
- Cardiology and Arrhythmology Clinic, Marche Polytechnic University, University Hospital "Ospedali Riuniti", Via Conca 71, Ancona, Italy.
| | - Marco Marchesini
- Cardiology and Arrhythmology Clinic, Marche Polytechnic University, University Hospital "Ospedali Riuniti", Via Conca 71, Ancona, Italy
| | - Daniele Contadini
- Cardiology and Arrhythmology Clinic, Marche Polytechnic University, University Hospital "Ospedali Riuniti", Via Conca 71, Ancona, Italy
| | - Alessio Menditto
- Cardiology and Arrhythmology Clinic, Marche Polytechnic University, University Hospital "Ospedali Riuniti", Via Conca 71, Ancona, Italy
| | - Marco Morelli
- Cardiology and Arrhythmology Clinic, Marche Polytechnic University, University Hospital "Ospedali Riuniti", Via Conca 71, Ancona, Italy
| | - Elisa Piccolo
- Cardiology and Arrhythmology Clinic, Marche Polytechnic University, University Hospital "Ospedali Riuniti", Via Conca 71, Ancona, Italy
| | - Nicola Battelli
- Oncology Clinic, Marche Polytechnic University, University Hospital "Ospedali Riuniti", Via Conca 71, Ancona, Italy
| | - Mirco Pistelli
- Oncology Clinic, Marche Polytechnic University, University Hospital "Ospedali Riuniti", Via Conca 71, Ancona, Italy
| | - Rossana Berardi
- Oncology Clinic, Marche Polytechnic University, University Hospital "Ospedali Riuniti", Via Conca 71, Ancona, Italy
| | - Stefano Cascinu
- Oncology Clinic, Marche Polytechnic University, University Hospital "Ospedali Riuniti", Via Conca 71, Ancona, Italy
| | - Alessando Capucci
- Cardiology and Arrhythmology Clinic, Marche Polytechnic University, University Hospital "Ospedali Riuniti", Via Conca 71, Ancona, Italy
| |
Collapse
|
38
|
Ahmed SH, Moussa Sherif DE, Fouad Y, Kelany M, Abdel-Rahman O. Principles of a risk evaluation and mitigation strategy (REMS) for breast cancer patients receiving potentially cardiotoxic adjuvant treatments. Expert Opin Drug Saf 2016; 15:911-23. [DOI: 10.1517/14740338.2016.1170115] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
39
|
Effets cardiovasculaires graves des chimiothérapies, thérapies ciblées et des traitements immunosuppresseurs. ACTA ACUST UNITED AC 2016. [DOI: 10.1007/s13546-015-1161-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
40
|
Rosa GM, Gigli L, Tagliasacchi MI, Di Iorio C, Carbone F, Nencioni A, Montecucco F, Brunelli C. Update on cardiotoxicity of anti-cancer treatments. Eur J Clin Invest 2016; 46:264-284. [PMID: 26728634 DOI: 10.1111/eci.12589] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 12/30/2015] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIMS Anti-cancer treatments markedly improved the prognosis of patients, but unfortunately might be hampered by cardiotoxicity. Both symptomatic and asymptomatic clinical forms of heart failure have been reported, which may be reversible or irreversible. The aim of this review is to provide an overview of the antineoplastic agents associated with cardiac toxicity and of the available diagnostic techniques. METHODS AND METHODS This narrative review is based on material from MEDLINE and PUBMED up to November 2015. We looked at the terms antineoplastic drugs and cardiac toxicity in combination with echocardiography, troponins, cardiac magnetic resonance, and positron emission tomography. RESULTS Anthracyclines, monoclonal antibodies, fluoropyrimidines, taxanes, alkylating agents, vinka alkaloids were reported to induce different clinical manifestations of cardioxicity. Chest radiotherapy is also associated with various forms of cardiac damage, which are indistinguishable from those found in patients with heart disease of other aetiologies and that may even appear several years after administration. Among diagnostic techniques, echocardiography is a noninvasive, cost-effective, and widely available imaging tool. Nuclear imaging and cardiac magnetic resonance may be used but are not so widely available and are more difficult to perform. Finally, some biomarkers, such as troponins, may be used to evaluate cardiac damage, but establishing the optimal timing of troponin assessment remains unclear and defining the cut-off point for positivity is still an important goal. CONCLUSIONS Cardiotoxicity of anti-cancer treatments is associated with development of heart failure. Novel diagnostic tools might be relevant to early recognize irreversible forms cardiac diseases.
Collapse
Affiliation(s)
- Gian Marco Rosa
- Division of Cardiology, Department of Internal Medicine, University of Genoa - IRCCS Azienda Ospedaliera Universitaria San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Lorenzo Gigli
- Division of Cardiology, Department of Internal Medicine, University of Genoa - IRCCS Azienda Ospedaliera Universitaria San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Maria Isabella Tagliasacchi
- Division of Cardiology, Department of Internal Medicine, University of Genoa - IRCCS Azienda Ospedaliera Universitaria San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Cecilia Di Iorio
- Division of Cardiology, Department of Internal Medicine, University of Genoa - IRCCS Azienda Ospedaliera Universitaria San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Federico Carbone
- Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, Geneva University, Geneva, Switzerland
| | - Alessio Nencioni
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Azienda Ospedaliera Universitaria San Martino-IST, Istituto Nazionale per la Ricerca Sul Cancro, Genoa, Italy
| | - Fabrizio Montecucco
- Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, Geneva University, Geneva, Switzerland
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa - IRCCS Azienda Ospedaliera Universitaria San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Claudio Brunelli
- Division of Cardiology, Department of Internal Medicine, University of Genoa - IRCCS Azienda Ospedaliera Universitaria San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| |
Collapse
|
41
|
Chaudry M, Banchs J, Chavez-MacGregor M. Anthracycline or trastuzumab-related cardiotoxicity: do we have a predictive biomarker? Biomark Med 2016; 10:315-28. [DOI: 10.2217/bmm.15.124] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Chemotherapy-induced cardiotoxicity, is a well-known and potentially serious complication strongly impacting the quality of life and overall survival of breast cancer patients. The current diagnostic approach to detect cardiac damage is the estimation of left ventricular ejection fraction by echocardiography. However, this approach exhibits less sensitivity toward early prediction of cardiomyopathy, not allowing for preventive strategies. Measurement of serum cardiac-specific biomarkers can be a valid diagnostic tool for identifying patients prone to developing cardiotoxocity and in whom closer cardiac monitoring and preventive strategies are pivotal. In this article, we review work done on biomarkers in recent years, with an emphasis on troponin and B-type natriuretic peptide, which are currently the most studied in this field. We also briefly discuss current and emerging imaging techniques for early detection of cardiomyopathy.
Collapse
Affiliation(s)
- Misbat Chaudry
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-3721, USA
| | - Jose Banchs
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mariana Chavez-MacGregor
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-3721, USA
- Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
42
|
Wieshammer S, Dreyhaupt J, Müller D, Momm F, Jakob A. Limitations of N-Terminal Pro-B-Type Natriuretic Peptide in the Diagnosis of Heart Disease among Cancer Patients Who Present with Cardiac or Pulmonary Symptoms. Oncology 2016; 90:143-50. [PMID: 26871423 DOI: 10.1159/000443505] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 12/18/2015] [Indexed: 11/19/2022]
Abstract
OBJECTIVES Recognizing heart disease is relevant to oncologists because cancer patients are at an increased risk of cardiac mortality due to shared risk factors and the adverse effects of cancer therapy. This study assessed the extent to which the measurement of N-terminal pro-B-type natriuretic peptide (NT-proBNP) aids in the diagnosis of heart disease in addition to a history of coronary artery disease and the presence of atrial fibrillation (composite test). The NT- proBNP cutoff value was 100 pg/ml. METHODS A series of 583 consecutive cancer patients (68.4 ± 11.0 years) who were referred because of cardiac or pulmonary symptoms prospectively underwent a diagnostic work-up. Heart disease was diagnosed if at least one of the following conditions was present: (a) history of coronary artery disease, (b) atrial fibrillation, (c) impaired left ventricular systolic function, (d) significant valvular disease, (e) pulmonary hypertension, or (f) left ventricular hypertrophy. RESULTS Except for (a), all 6 conditions were associated with NT-proBNP >100 pg/ml. The sensitivity/specificity values of the composite test were 0.92/0.50 for any heart disease. Several extracardiac covariates were associated with NT-proBNP >100 pg/ml, which contributed to the low test specificity. CONCLUSIONS The low specificity of NT-proBNP limits its value for the diagnosis of heart disease in cancer patients.
Collapse
Affiliation(s)
- Siegfried Wieshammer
- Departments of Cardiology, Pulmonology and Critical Care Medicine, Radiation Oncology and Medical Oncology, Offenburg Hospital, Offenburg, and Institute of Epidemiology and Medical Biometry, University of Ulm, Ulm, Germany
| | | | | | | | | |
Collapse
|
43
|
Ibraheem A, Stankowski-Drengler TJ, Gbolahan OB, Engel JM, Onitilo AA. Chemotherapy-induced cardiotoxicity in breast cancer patients. BREAST CANCER MANAGEMENT 2016. [DOI: 10.2217/bmt-2016-0002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Chemotherapy-induced cardiotoxicity (CIC) is a well-documented side effect of breast cancer treatment. Nearly all chemotherapeutic agents can cause CIC with the highest occurrence found in anthracycline and trastuzumab use. Treatment- and patient-related risk factors contribute to the development of CIC making risk modification an important consideration during breast cancer treatment. Prevention and early detection of cardiotoxicity are key to minimizing permanent and devastating cardiac damage; therefore, early involvement of a cardiologist including periodic cardiac monitoring during and after chemotherapy exposure is recommended.
Collapse
Affiliation(s)
| | | | | | - Jessica M Engel
- Marshfield Clinic Cancer Care at St Michaels, Stevens Point, WI 54481, USA
| | - Adedayo A Onitilo
- Marshfield Clinic Weston Center, Oncology/Hematology Department, Weston, WI 54481, USA
| |
Collapse
|
44
|
Galal A, El-Bakly WM, Al Haleem ENA, El-Demerdash E. Selective A3 adenosine receptor agonist protects against doxorubicin-induced cardiotoxicity. Cancer Chemother Pharmacol 2016; 77:309-322. [PMID: 26676227 DOI: 10.1007/s00280-015-2937-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 11/30/2015] [Indexed: 01/23/2023]
Abstract
PURPOSE Doxorubicin (DOX) is an effective anticancer drug; however, its clinical use is limited by its cardiotoxic effect. Adenosine was proved to mediate anti-inflammatory effects and protected from myocardial ischemia/reperfusion injury. So the present work was designed to examine the effectiveness of a selective A3 adenosine receptor agonist (Cl-IB-MECA) in DOX-induced cardiotoxicity and to elucidate the underlying mechanisms via studying its effect on different oxidative stress, inflammatory and apoptotic markers. METHODS Firstly the potential cardioprotective dose of Cl-IB-MECA was screened in male Wistar rats at different doses (20, 40 and 80 µg/kg; i.v) against a single dose of DOX (15 mg/kg; i.p). Secondly, the dose of 40 µg/kg Cl-IB-MECA was selected for further assessment of the cardioprotective mechanisms. RESULTS Cl-IB-MECA at a dose 40 µg/kg (i.v) protects against DOX-induced bradycardia, elevated creatine kinase isoenzyme-MB and histopathological changes. Also, it significantly ameliorates oxidative stress injury evoked by DOX as evidenced by inhibition of reduced glutathione depletion and lipid peroxidation as well as elevation of antioxidant enzyme activities. Additionally, DOX provoked inflammatory responses by increasing the expressions of nuclear factor kappa B and the levels of tumor necrosis factor alpha. Cl-IB-MECA pretreatment significantly inhibited these inflammatory responses. Furthermore, DOX induced apoptotic tissue damage by increasing cytochrome c expressions which was suppressed by Cl-IB-MECA pretreatment. CONCLUSION Cl-IB-MECA protects against DOX-induced cardiotoxicity through restoration of the oxidant/antioxidant status and consequential suppression of DOX-induced inflammatory responses and abrogation of the resultant apoptotic signals.
Collapse
Affiliation(s)
- Aya Galal
- Cardiac Surgery Hospital, Ain Shams University, Cairo, Egypt
| | - Wesam M El-Bakly
- Department of Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ekram Nemr Abd Al Haleem
- Department of Pharmacology and Toxicology, Faculty of Pharmacy for Girls, Al Azhar University, Cairo, Egypt
| | - Ebtehal El-Demerdash
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Abbasia, Cairo, Egypt.
| |
Collapse
|
45
|
Tabernero A, González-Garcinuño Á, Galán MA, Martín del Valle EM. Survey of supercritical fluid techniques for producing drug delivery systems for a potential use in cancer therapy. REV CHEM ENG 2016. [DOI: 10.1515/revce-2015-0059] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
AbstractStandard drug delivery systems for cancer treatment usually comprise a device with a specific size and shape (depending on the type of cancer that has to be treated), which is composed by a biodegradable compound with a chemotherapeutic entrapped within it. This device should have a molecule (mainly a protein) bound to its surface to target only cancer cells. On the contrary, supercritical fluids (SCF) have been widely used in the pharmaceutical industry for creating drug delivery systems or for extracting drugs from natural sources. This review explains the potential of SCFs for cancer therapies by studying the current uses of the different high-pressure processes that can be useful for this medical treatment, such as the development of new drug delivery systems (with their drug release) or the extraction of chemotherapeutics from a vegetal matrix.
Collapse
|
46
|
Korzeniowska K, Jankowski J, Cieślewicz A, Jabłecka A. Current approach for detection of sub-clinical left ventricular dysfunction associated with chemotherapy. Pharmacol Rep 2015; 67:1098-102. [DOI: 10.1016/j.pharep.2015.03.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 03/12/2015] [Accepted: 03/18/2015] [Indexed: 01/27/2023]
|
47
|
Nguyen KL, Alrezk R, Mansourian PG, Naeim A, Rettig MB, Lee CC. The Crossroads of Geriatric Cardiology and Cardio-Oncology. CURRENT GERIATRICS REPORTS 2015; 4:327-337. [PMID: 26543801 PMCID: PMC4624825 DOI: 10.1007/s13670-015-0147-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Cancer and cardiovascular disease (CVD) are two major causes of mortality in older adults. With improved survival and outcomes from cancer and CVD, the role of the geriatrician is evolving. Geriatricians provide key skills to facilitate patient-centered and value-based care in the growing older population of cancer patients (and survivors). Cancer treatment in older adults is particularly injurious with respect to complications stemming from cancer therapy and as well as to CVD related to cancer therapy in the context of physiologic aging. To best meet their natural potential as caregiving leaders, geriatricians must hone skills and insights pertaining to oncologic and cardiovascular care, insights that can inform and enhance key management expertise. In this paper, we will review common chemotherapy and radiation-induced cardiovascular complications, screening recommendations, and advance the concept of a geriatric, cardiology, and oncology collaboration. We assert that geriatricians are well suited to a leadership role in the care of older cardio-oncology patients and in the education of primary care physicians and subspecialists on geriatric principles.
Collapse
Affiliation(s)
- Kim-Lien Nguyen
- VA Greater Los Angeles Healthcare System, 11301 Wilshire Blvd, MC 111E, Los Angeles, CA 90073 USA ; Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90073 USA
| | - Rami Alrezk
- VA Greater Los Angeles Healthcare System, 11301 Wilshire Blvd, MC 111E, Los Angeles, CA 90073 USA ; Division of Geriatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA USA ; GRECC, VA Greater Los Angeles Healthcare System, Los Angeles, CA USA
| | - Pejman G Mansourian
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90073 USA
| | - Arash Naeim
- Division of Geriatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA USA ; Division of Hematology-Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA USA
| | - Matthew B Rettig
- VA Greater Los Angeles Healthcare System, 11301 Wilshire Blvd, MC 111E, Los Angeles, CA 90073 USA ; Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, CA USA ; Division of Hematology-Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA USA
| | - Cathy C Lee
- VA Greater Los Angeles Healthcare System, 11301 Wilshire Blvd, MC 111E, Los Angeles, CA 90073 USA ; Division of Geriatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA USA ; GRECC, VA Greater Los Angeles Healthcare System, Los Angeles, CA USA
| |
Collapse
|
48
|
Emerging delivery systems to reduce doxorubicin cardiotoxicity and improve therapeutic index: focus on liposomes. Anticancer Drugs 2015; 26:241-58. [PMID: 25415656 DOI: 10.1097/cad.0000000000000182] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Anthracyclines are powerful anticancer agents and among the most important tools in the chemotherapy armamentarium of medical oncologists. They are approved for use in the treatment of a broad variety of solid and hematologic neoplasms. However, the usefulness of these agents, particularly doxorubicin, the most widely used anthracycline, is limited by considerable toxicity, especially damage to the cardiac muscle, which is cumulative and mostly irreversible, restricting extended use of this drug. In the last 30 years, extensive research with a variety of drug-delivery systems has attempted to overcome this limitation, with clinical success mostly confined to liposome formulations. Liposomal doxorubicin, and particularly pegylated liposomal doxorubicin, has shown significant pharmacologic advantages and an added clinical value over doxorubicin. Here, we review the mechanisms of action and toxicity of doxorubicin, and ways to reduce toxicity, with a focus on liposome-based drug-delivery systems.
Collapse
|
49
|
Storozynsky E. Multimodality assessment and treatment of chemotherapy-induced cardiotoxicity. Future Cardiol 2015; 11:421-4. [DOI: 10.2217/fca.15.49] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Eugene Storozynsky
- University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA
| |
Collapse
|
50
|
Cross MJ, Berridge BR, Clements PJM, Cove-Smith L, Force TL, Hoffmann P, Holbrook M, Lyon AR, Mellor HR, Norris AA, Pirmohamed M, Tugwood JD, Sidaway JE, Park BK. Physiological, pharmacological and toxicological considerations of drug-induced structural cardiac injury. Br J Pharmacol 2015; 172:957-74. [PMID: 25302413 PMCID: PMC4314188 DOI: 10.1111/bph.12979] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 09/30/2014] [Accepted: 10/06/2014] [Indexed: 01/01/2023] Open
Abstract
The incidence of drug-induced structural cardiotoxicity, which may lead to heart failure, has been recognized in association with the use of anthracycline anti-cancer drugs for many years, but has also been shown to occur following treatment with the new generation of targeted anti-cancer agents that inhibit one or more receptor or non-receptor tyrosine kinases, serine/threonine kinases as well as several classes of non-oncology agents. A workshop organized by the Medical Research Council Centre for Drug Safety Science (University of Liverpool) on 5 September 2013 and attended by industry, academia and regulatory representatives, was designed to gain a better understanding of the gaps in the field of structural cardiotoxicity that can be addressed through collaborative efforts. Specific recommendations from the workshop for future collaborative activities included: greater efforts to identify predictive (i) preclinical; and (ii) clinical biomarkers of early cardiovascular injury; (iii) improved understanding of comparative physiology/pathophysiology and the clinical predictivity of current preclinical in vivo models; (iv) the identification and use of a set of cardiotoxic reference compounds for comparative profiling in improved animal and human cellular models; (v) more sharing of data (through publication/consortia arrangements) on target-related toxicities; (vi) strategies to develop cardio-protective agents; and (vii) closer interactions between preclinical scientists and clinicians to help ensure best translational efforts.
Collapse
Affiliation(s)
- M J Cross
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of LiverpoolLiverpool, UK
| | - B R Berridge
- Safety Assessment, GlaxoSmithKlineResearch Triangle Park, NC, USA
| | - P J M Clements
- David Jack Centre for Research & Development, GlaxoSmithKlineWare, Herts, UK
| | - L Cove-Smith
- Clinical & Experimental Pharmacology, Cancer Research UK Manchester Institute, University of ManchesterManchester, UK
| | - T L Force
- Center for Translational Medicine and Cardiology Division, Temple University School of MedicinePhiladelphia, PA, USA
| | - P Hoffmann
- Preclinical Safety, Novartis Pharm CorpEast Hanover, NJ, USA
| | - M Holbrook
- Safety Pharmacology, Covance Laboratories, Ltd.Harrogate, North Yorkshire, UK
| | - A R Lyon
- NIHR Cardiovascular Biomedical Research Unit, Royal Brompton Hospital and Imperial CollegeLondon, UK
| | - H R Mellor
- Drug Safety Evaluation, Vertex Pharmaceuticals (Europe), Ltd.Abingdon, Oxfordshire, UK
| | - A A Norris
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of LiverpoolLiverpool, UK
| | - M Pirmohamed
- The Wolfson Centre for Personalised Medicine, Institute of Translational Medicine, University of LiverpoolLiverpool, UK
| | - J D Tugwood
- Clinical & Experimental Pharmacology, Cancer Research UK Manchester Institute, University of ManchesterManchester, UK
| | - J E Sidaway
- Innovative Medicines, AstraZeneca R&DMacclesfield, UK
| | - B K Park
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of LiverpoolLiverpool, UK
| |
Collapse
|