51
|
Cavalcanti IDL, Soares JCS, Medeiros SMDFRDS, Cavalcanti IMF, Lira Nogueira MCDB. Can antioxidant vitamins avoid the cardiotoxicity of doxorubicin in treating breast cancer? PHARMANUTRITION 2021. [DOI: 10.1016/j.phanu.2021.100259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
52
|
|
53
|
Dobson R, Ghosh AK, Ky B, Marwick T, Stout M, Harkness A, Steeds R, Robinson S, Oxborough D, Adlam D, Stanway S, Rana B, Ingram T, Ring L, Rosen S, Plummer C, Manisty C, Harbinson M, Sharma V, Pearce K, Lyon AR, Augustine DX. British Society for Echocardiography and British Cardio-Oncology Society guideline for transthoracic echocardiographic assessment of adult cancer patients receiving anthracyclines and/or trastuzumab. Echo Res Pract 2021; 8:G1-G18. [PMID: 34106116 PMCID: PMC8052569 DOI: 10.1530/erp-21-0001] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 02/26/2021] [Indexed: 12/17/2022] Open
Abstract
The subspecialty of cardio-oncology aims to reduce cardiovascular morbidity and mortality in patients with cancer or following cancer treatment. Cancer therapy can lead to a variety of cardiovascular complications, including left ventricular systolic dysfunction, pericardial disease, and valvular heart disease. Echocardiography is a key diagnostic imaging tool in the diagnosis and surveillance for many of these complications. The baseline assessment and subsequent surveillance of patients undergoing treatment with anthracyclines and/or human epidermal growth factor (EGF) receptor (HER) 2-positive targeted treatment (e.g. trastuzumab and pertuzumab) form a significant proportion of cardio-oncology patients undergoing echocardiography. This guideline from the British Society of Echocardiography and British Cardio-Oncology Society outlines a protocol for baseline and surveillance echocardiography of patients undergoing treatment with anthracyclines and/or trastuzumab. The methodology for acquisition of images and the advantages and disadvantages of techniques are discussed. Echocardiographic definitions for considering cancer therapeutics-related cardiac dysfunction are also presented.
Collapse
Affiliation(s)
- Rebecca Dobson
- Cardio-Oncology Service, Liverpool Heart and Chest NHS Foundation Trust, Liverpool, UK
| | - Arjun K Ghosh
- Cardio-Oncology Service, Barts Heart Centre, Barts Health NHS Trust, London, UK
- Cardio-Oncology Service, Hatter Cardiovascular Research Institute, University College London and University College London Hospitals NHS Foundation Trust, London, UK
| | - Bonnie Ky
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Tom Marwick
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Martin Stout
- University Hospital South Manchester NHS Foundation Trust, Manchester, UK
| | - Allan Harkness
- East Suffolk and North Essex NHS Foundation Trust, Colchester, UK
| | - Rick Steeds
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | | | | | - David Adlam
- University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Susannah Stanway
- Cardio-Oncology Service, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | - Bushra Rana
- Imperial College Healthcare NHS Trust, London, UK
| | - Thomas Ingram
- The Shrewsbury and Telford Hospital NHS Trust, Shrewsbury, UK
| | - Liam Ring
- West Suffolk NHS Foundation Trust, Bury St Edmunds, UK
| | - Stuart Rosen
- Royal Brompton and Harefield NHS Foundation Trust and Imperial College London, London, UK
| | - Chris Plummer
- The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle, UK
| | - Charlotte Manisty
- Cardio-Oncology Service, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | | | - Vishal Sharma
- Royal Liverpool and Broadgreen University Hospitals NHS Trust, Liverpool, UK
| | - Keith Pearce
- University Hospital South Manchester NHS Foundation Trust, Manchester, UK
| | - Alexander R Lyon
- Royal Brompton and Harefield NHS Foundation Trust and Imperial College London, London, UK
| | - Daniel X Augustine
- Department of Cardiology, Royal United Hospitals Bath NHS Foundation Trust, Bath, UK
- Department for Health, University of Bath, Bath, UK
| | - the British Society of Echocardiography (BSE) and the British Society of Cardio-Oncology (BCOS)
- Cardio-Oncology Service, Liverpool Heart and Chest NHS Foundation Trust, Liverpool, UK
- Cardio-Oncology Service, Barts Heart Centre, Barts Health NHS Trust, London, UK
- Cardio-Oncology Service, Hatter Cardiovascular Research Institute, University College London and University College London Hospitals NHS Foundation Trust, London, UK
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Baker Heart and Diabetes Institute, Melbourne, Australia
- University Hospital South Manchester NHS Foundation Trust, Manchester, UK
- East Suffolk and North Essex NHS Foundation Trust, Colchester, UK
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- North West Anglia Foundation Trust, UK
- Liverpool John Moores University, Liverpool, UK
- University Hospitals of Leicester NHS Trust, Leicester, UK
- Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London, UK
- Imperial College Healthcare NHS Trust, London, UK
- The Shrewsbury and Telford Hospital NHS Trust, Shrewsbury, UK
- West Suffolk NHS Foundation Trust, Bury St Edmunds, UK
- Royal Brompton and Harefield NHS Foundation Trust and Imperial College London, London, UK
- The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle, UK
- Belfast Health and Social Care Trust, Belfast, UK
- Royal Liverpool and Broadgreen University Hospitals NHS Trust, Liverpool, UK
- Department of Cardiology, Royal United Hospitals Bath NHS Foundation Trust, Bath, UK
- Department for Health, University of Bath, Bath, UK
| |
Collapse
|
54
|
Ansheles AA, Sergienko IV, Prus YA, Sergienko VB. Nuclear imaging of chemotherapy-induced cardiotoxicity. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2021. [DOI: 10.15829/1728-8800-2021-2537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The high efficiency of modern chemotherapy has made it possible to achieve great success in the treatment of cancer. Cardiovascular adverse effects are a major disadvantage of anticancer therapy, often requiring low and less effective doses or even drug withdrawal. Nuclear imaging techniques are the most sensitive in early detection of left ventricular damage and dysfunction during chemotherapy. This review presents modern data on the potential of nuclear imaging of cardiotoxicity.
Collapse
Affiliation(s)
| | | | - Yu. A. Prus
- National Medical Research Center of Cardiology
| | | |
Collapse
|
55
|
Dobson R, Ghosh AK, Ky B, Marwick T, Stout M, Harkness A, Steeds R, Robinson S, Oxborough D, Adlam D, Stanway S, Rana B, Ingram T, Ring L, Rosen S, Plummer C, Manisty C, Harbinson M, Sharma V, Pearce K, Lyon AR, Augustine DX. BSE and BCOS Guideline for Transthoracic Echocardiographic Assessment of Adult Cancer Patients Receiving Anthracyclines and/or Trastuzumab. JACC CardioOncol 2021; 3:1-16. [PMID: 34396303 PMCID: PMC8352267 DOI: 10.1016/j.jaccao.2021.01.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 01/06/2023] Open
Abstract
The subspecialty of cardio-oncology aims to reduce cardiovascular morbidity and mortality in patients with cancer or following cancer treatment. Cancer therapy can lead to a variety of cardiovascular complications, including left ventricular systolic dysfunction, pericardial disease, and valvular heart disease. Echocardiography is a key diagnostic imaging tool in the diagnosis and surveillance for many of these complications. The baseline assessment and subsequent surveillance of patients undergoing treatment with anthracyclines and/or human epidermal growth factor receptor (HER) 2-positive targeted treatment (e.g., trastuzumab and pertuzumab) form a significant proportion of cardio-oncology patients undergoing echocardiography. This guideline from the British Society of Echocardiography and British Cardio-Oncology Society outlines a protocol for baseline and surveillance echocardiography of patients undergoing treatment with anthracyclines and/or trastuzumab. The methodology for acquisition of images and the advantages and disadvantages of techniques are discussed. Echocardiographic definitions for considering cancer therapeutics-related cardiac dysfunction are also presented.
Collapse
Key Words
- 2D, 2-dimensional
- 3D, 3-dimensional
- A2C, apical 2-chamber
- A3C, apical 3-chamber
- A4C, apical 4-chamber
- BSE, British Society of Echocardiography
- CMR, cardiac magnetic resonance
- CTRCD, cancer therapy–related cardiac dysfunction
- ECG, electrocardiogram
- GLS, global longitudinal strain
- HER2 therapy
- HER2, human epidermal growth factor receptor 2
- LV, left ventricular
- LVEF, left ventricular ejection fraction
- MV, mitral valve
- RH, right heart
- ROI, region of interest
- RV, right ventricular
- TDI, tissue Doppler imaging
- TRV, tricuspid regurgitant velocity
- anthracycline
- echocardiography
- guidelines
- imaging
Collapse
Affiliation(s)
- Rebecca Dobson
- Cardio-Oncology Service, Liverpool Heart and Chest NHS Foundation Trust, Liverpool, United Kingdom
| | - Arjun K. Ghosh
- Cardio-Oncology Service, Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom
- Cardio-Oncology Service, Hatter Cardiovascular Research Institute, University College London and University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Bonnie Ky
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, USA
| | - Tom Marwick
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Martin Stout
- University Hospital South Manchester NHS Foundation Trust, Manchester, United Kingdom
| | - Allan Harkness
- East Suffolk and North Essex NHS Foundation Trust, Colchester, United Kingdom
| | - Rick Steeds
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | | | | | - David Adlam
- University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
| | - Susannah Stanway
- Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London, United Kingdom
| | - Bushra Rana
- Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Thomas Ingram
- The Shrewsbury and Telford Hospital NHS Trust, Shrewsbury, United Kingdom
| | - Liam Ring
- West Suffolk NHS Foundation Trust, Bury St. Edmunds, United Kingdom
| | - Stuart Rosen
- Royal Brompton and Harefield NHS Foundation Trust and Imperial College London, London, United Kingdom
| | - Chris Plummer
- The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle, United Kingdom
| | - Charlotte Manisty
- Cardio-Oncology Service, Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom
| | - Mark Harbinson
- Belfast Health and Social Care Trust, Belfast, United Kingdom
| | - Vishal Sharma
- Royal Liverpool and Broadgreen University Hospitals NHS Trust, Liverpool, United Kingdom
| | - Keith Pearce
- University Hospital South Manchester NHS Foundation Trust, Manchester, United Kingdom
| | - Alexander R. Lyon
- Royal Brompton and Harefield NHS Foundation Trust and Imperial College London, London, United Kingdom
| | - Daniel X. Augustine
- Department of Cardiology, Royal United Hospitals Bath NHS Foundation Trust, Bath, United Kingdom
| | - British Society of Echocardiography (BSE) and theBritish Society of Cardio-Oncology (BCOS)
- Cardio-Oncology Service, Liverpool Heart and Chest NHS Foundation Trust, Liverpool, United Kingdom
- Cardio-Oncology Service, Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom
- Cardio-Oncology Service, Hatter Cardiovascular Research Institute, University College London and University College London Hospitals NHS Foundation Trust, London, United Kingdom
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, USA
- Baker Heart and Diabetes Institute, Melbourne, Australia
- University Hospital South Manchester NHS Foundation Trust, Manchester, United Kingdom
- East Suffolk and North Essex NHS Foundation Trust, Colchester, United Kingdom
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
- North West Anglia Foundation Trust, United Kingdom
- Liverpool John Moores University, Liverpool, United Kingdom
- University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
- Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London, United Kingdom
- Imperial College Healthcare NHS Trust, London, United Kingdom
- The Shrewsbury and Telford Hospital NHS Trust, Shrewsbury, United Kingdom
- West Suffolk NHS Foundation Trust, Bury St. Edmunds, United Kingdom
- Royal Brompton and Harefield NHS Foundation Trust and Imperial College London, London, United Kingdom
- The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle, United Kingdom
- Belfast Health and Social Care Trust, Belfast, United Kingdom
- Royal Liverpool and Broadgreen University Hospitals NHS Trust, Liverpool, United Kingdom
- Department of Cardiology, Royal United Hospitals Bath NHS Foundation Trust, Bath, United Kingdom
| |
Collapse
|
56
|
Kihara M, Kaiya H, Hirai Y, Katayama H, Terao A, Nishikawa M. Salmon acyl-ghrelin increases food intake and reduces doxorubicin-induced myocardial apoptosis in rats, likely by anti-oxidative activity. Peptides 2021; 137:170471. [PMID: 33340558 DOI: 10.1016/j.peptides.2020.170471] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 10/26/2020] [Accepted: 11/28/2020] [Indexed: 12/16/2022]
Abstract
We had reported that orally administered ghrelin-containing salmon stomach extract prevents doxorubicin (DOX)-induced cardiotoxicity. In this study, we investigated the binding affinity of salmon ghrelin to rat ghrelin receptor and the cardioprotective effects of subcutaneous (sc) injected synthetic salmon ghrelin in rats with DOX-induced acute heart failure in order to clarify the potential efficacy of salmon ghrelin. Intracellular calcium mobilization assay was performed on rat GHS-R1a-expressing CHO cells to reveal ghrelin activity. Rats were divided into five groups; the normal control (I), and toxic control (II) groups were given saline (sc, twice daily), and the salmon acyl-ghrelin (sAG) (III), salmon unacylated-ghrelin (sUAG) (IV), and rat acyl-ghrelin (rAG) (V) groups were given corresponding synthetic ghrelins (sc, twice daily), respectively. After seven days of treatment, DOX (20 mg/kg BW) or saline was administered to the corresponding groups by intraperitoneal injection. The toxic control group was the negative control group for the DOX-induced cardiotoxicity groups. While sAG displayed similar affinity to rAG upon application to GHS-R1a-expressing cells, and also decreased DOX-induced apoptosis and increased food intake, sUAG did not. Both sAG and rAG improved DOX-induced deterioration, showing anti-oxidative activity. The anti-oxidative activity of sAG might contribute to the protective effects on cardiomyocytes. The results also suggest that, similar to rAG, sAG is a potent protectant against DOX-induced cardiotoxicity and a potential functional component in orally administered ghrelin-containing salmon stomach extract, which prevented DOX-induced cardiotoxicity in our previous study.
Collapse
Affiliation(s)
- Minoru Kihara
- Department of Marine Biology and Sciences, School of Biological Sciences, Tokai University, 5-1-1-1 Minamisawa, Minami-ku, Sapporo, 005-8601, Japan.
| | - Hiroyuki Kaiya
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-shinmachi, Suita, 564-8565, Japan
| | - Yumi Hirai
- Department of Marine Biology and Sciences, School of Biological Sciences, Tokai University, 5-1-1-1 Minamisawa, Minami-ku, Sapporo, 005-8601, Japan
| | - Hidekazu Katayama
- Department of Applied Biochemistry, School of Engineering, Tokai University, 4-1-1 Kitakaname, Hiratsuka, Kanagawa 259-1292, Japan
| | - Akira Terao
- Liberal Arts Education Center, Sapporo Campus, Tokai University, 5-1-1-1 Minamisawa, Minami-ku, Sapporo, 005-8601, Japan
| | - Masazumi Nishikawa
- Department of Food Management, School of Food, Agricultural and Environmental Sciences, Miyagi University, 2-2-1 Hatadate, Taihaku-ku, Sendai, 982-0215, Japan
| |
Collapse
|
57
|
Antonucci S, Di Sante M, Tonolo F, Pontarollo L, Scalcon V, Alanova P, Menabò R, Carpi A, Bindoli A, Rigobello MP, Giorgio M, Kaludercic N, Di Lisa F. The Determining Role of Mitochondrial Reactive Oxygen Species Generation and Monoamine Oxidase Activity in Doxorubicin-Induced Cardiotoxicity. Antioxid Redox Signal 2021; 34:531-550. [PMID: 32524823 PMCID: PMC7885901 DOI: 10.1089/ars.2019.7929] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Aims: Doxorubicin cardiomyopathy is a lethal pathology characterized by oxidative stress, mitochondrial dysfunction, and contractile impairment, leading to cell death. Although extensive research has been done to understand the pathophysiology of doxorubicin cardiomyopathy, no effective treatments are available. We investigated whether monoamine oxidases (MAOs) could be involved in doxorubicin-derived oxidative stress, and in the consequent mitochondrial, cardiomyocyte, and cardiac dysfunction. Results: We used neonatal rat ventricular myocytes (NRVMs) and adult mouse ventricular myocytes (AMVMs). Doxorubicin alone (i.e., 0.5 μM doxorubicin) or in combination with H2O2 induced an increase in mitochondrial formation of reactive oxygen species (ROS), which was prevented by the pharmacological inhibition of MAOs in both NRVMs and AMVMs. The pharmacological approach was supported by the genetic ablation of MAO-A in NRVMs. In addition, doxorubicin-derived ROS caused lipid peroxidation and alterations in mitochondrial function (i.e., mitochondrial membrane potential, permeability transition, redox potential), mitochondrial morphology (i.e., mitochondrial distribution and perimeter), sarcomere organization, intracellular [Ca2+] homeostasis, and eventually cell death. All these dysfunctions were abolished by MAO inhibition. Of note, in vivo MAO inhibition prevented chamber dilation and cardiac dysfunction in doxorubicin-treated mice. Innovation and Conclusion: This study demonstrates that the severe oxidative stress induced by doxorubicin requires the involvement of MAOs, which modulate mitochondrial ROS generation. MAO inhibition provides evidence that mitochondrial ROS formation is causally linked to all disorders caused by doxorubicin in vitro and in vivo. Based upon these results, MAO inhibition represents a novel therapeutic approach for doxorubicin cardiomyopathy.
Collapse
Affiliation(s)
| | - Moises Di Sante
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Federica Tonolo
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Laura Pontarollo
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Valeria Scalcon
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Petra Alanova
- Department of Biomedical Sciences, University of Padova, Padova, Italy.,Institute for Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Roberta Menabò
- Neuroscience Institute, National Research Council of Italy (CNR), Padova, Italy
| | - Andrea Carpi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Alberto Bindoli
- Neuroscience Institute, National Research Council of Italy (CNR), Padova, Italy
| | | | - Marco Giorgio
- Department of Biomedical Sciences, University of Padova, Padova, Italy.,European Institute of Oncology (IEO), Milan, Italy
| | - Nina Kaludercic
- Neuroscience Institute, National Research Council of Italy (CNR), Padova, Italy
| | - Fabio Di Lisa
- Department of Biomedical Sciences, University of Padova, Padova, Italy.,Neuroscience Institute, National Research Council of Italy (CNR), Padova, Italy
| |
Collapse
|
58
|
Engwall MJ, Everds N, Turk JR, Vargas HM. The Effects of Repeat-Dose Doxorubicin on Cardiovascular Functional Endpoints and Biomarkers in the Telemetry-Equipped Cynomolgus Monkey. Front Cardiovasc Med 2021; 8:587149. [PMID: 33708802 PMCID: PMC7941602 DOI: 10.3389/fcvm.2021.587149] [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: 07/24/2020] [Accepted: 01/18/2021] [Indexed: 12/02/2022] Open
Abstract
Purpose: Doxorubicin-related heart failure has been recognized as a serious complication of cancer chemotherapy. This paper describes a cardiovascular safety pharmacology study with chronic dosing of doxorubicin in a non-human primate model designed to characterize the onset and magnitude of left ventricular dysfunction (LVD) using invasive and non-invasive methods. Methods: Cynomolgus monkeys (N = 12) were given repeated intravenous injections of doxorubicin over 135 days (19 weeks) with dosing holidays when there was evidence of significantly decreased hematopoiesis; a separate group (N = 12) received vehicle. Arterial and left ventricular pressure telemetry and cardiac imaging by echocardiography allowed regular hemodynamic assessments and determination of LVD. Blood samples were collected for hematology, clinical chemistry, and assessment of cardiac troponin (cTnI) and N-terminal pro b-type natriuretic peptide (NT-proBNP). Myocardial histopathology was a terminal endpoint. Results: There was variable sensitivity to the onset of treatment effects, for example 25% of doxorubicin-treated animals exhibited LVD (e.g., decreases in ejection fraction) following 50–63 days (cumulative dose: 8–9 mg/kg) on study. All animals deteriorated into heart failure with additional dosing 135 days (total cumulative dose: 11–17 mg/kg). Reductions in arterial pressure and cardiac contractility, as well as QTc interval prolongation, was evident following doxorubicin-treatment. Both cTnI and NT-proBNP were inconsistently higher at the end of the study in animals with LVD. Measurements collected from control animals were consistent and stable over the same time frame. Minimal to mild, multifocal, vacuolar degeneration of cardiomyocytes was observed in 7 of 12 animals receiving doxorubicin and 0 of 12 animals receiving vehicle. Conclusions: This repeat-dose study of doxorubicin treatment in the cynomolgus monkey demonstrated a clinically relevant pattern of progressive heart failure. Importantly, the study revealed how both telemetry and non-invasive echocardiography measurements could track the gradual onset of LVD.
Collapse
Affiliation(s)
- Michael J Engwall
- Translational Safety and Bioanalytical Sciences, Amgen Inc., Thousand Oaks, CA, United States
| | - Nancy Everds
- Non-clinical Sciences, Seattle Genetics, Inc., Seattle, WA, United States
| | - James R Turk
- Translational Safety and Bioanalytical Sciences, Amgen Inc., Thousand Oaks, CA, United States
| | - Hugo M Vargas
- Translational Safety and Bioanalytical Sciences, Amgen Inc., Thousand Oaks, CA, United States
| |
Collapse
|
59
|
Liu YQ, Wang XL, He DH, Cheng YX. Protection against chemotherapy- and radiotherapy-induced side effects: A review based on the mechanisms and therapeutic opportunities of phytochemicals. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 80:153402. [PMID: 33203590 DOI: 10.1016/j.phymed.2020.153402] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 09/29/2020] [Accepted: 10/26/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Although great achievements have been made in the field of cancer therapy, chemotherapy and radiotherapy remain the mainstay cancer therapeutic modalities. However, they are associated with various side effects, including cardiocytotoxicity, nephrotoxicity, myelosuppression, neurotoxicity, hepatotoxicity, gastrointestinal toxicity, mucositis, and alopecia, which severely affect the quality of life of cancer patients. Plants harbor a great chemical diversity and flexible biological properties that are well-compatible with their use as adjuvant therapy in reducing the side effects of cancer therapy. PURPOSE This review aimed to comprehensively summarize the molecular mechanisms by which phytochemicals ameliorate the side effects of cancer therapies and their potential clinical applications. METHODS We obtained information from PubMed, Science Direct, Web of Science, and Google scholar, and introduced the molecular mechanisms by which chemotherapeutic drugs and irradiation induce toxic side effects. Accordingly, we summarized the underlying mechanisms of representative phytochemicals in reducing these side effects. RESULTS Representative phytochemicals exhibit a great potential in reducing the side effects of chemotherapy and radiotherapy due to their broad range of biological activities, including antioxidation, antimutagenesis, anti-inflammation, myeloprotection, and immunomodulation. However, since a majority of the phytochemicals have only been subjected to preclinical studies, clinical trials are imperative to comprehensively evaluate their therapeutic values. CONCLUSION This review highlights that phytochemicals have interesting properties in relieving the side effects of chemotherapy and radiotherapy. Future studies are required to explore the clinical benefits of these phytochemicals for exploitation in chemotherapy and radiotherapy.
Collapse
Affiliation(s)
- Yong-Qiang Liu
- Institute of Clinical Pharmacology, Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Research Center of Chinese Herbal Resources Science and Engineering, Key Laboratory of Chinese Medicinal Resource from Lingnan, Ministry of Education, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Xiao-Lu Wang
- Institute of Traditional Chinese Medicine and Natural Products, Jinan University, Guangzhou 510632, China
| | - Dan-Hua He
- Institute of Clinical Pharmacology, Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Research Center of Chinese Herbal Resources Science and Engineering, Key Laboratory of Chinese Medicinal Resource from Lingnan, Ministry of Education, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yong-Xian Cheng
- School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Shenzhen 518060, China.
| |
Collapse
|
60
|
Broberg AM, Geisler J, Tuohinen S, Skytta T, Hrafnkelsdóttir ÞJ, Nielsen KM, Hedayati E, Omland T, Offersen BV, Lyon AR, Gulati G. Prevention, Detection, and Management of Heart Failure in Patients Treated for Breast Cancer. Curr Heart Fail Rep 2020; 17:397-408. [PMID: 32979150 PMCID: PMC7683437 DOI: 10.1007/s11897-020-00486-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/08/2020] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW Long-term survival has increased significantly in breast cancer patients, and cardiovascular side effects are surpassing cancer-related mortality. We summarize risk factors, prevention strategies, detection, and management of cardiotoxicity, with focus on left ventricular dysfunction and heart failure, during breast cancer treatment. RECENT FINDINGS Baseline treatment of cardiovascular risk factors is recommended. Anthracycline and trastuzumab treatment constitute a substantial risk of developing cardiotoxicity. There is growing evidence that this can be treated with beta blockers and angiotensin antagonists. Early detection of cardiotoxicity with cardiac imaging and circulating cardiovascular biomarkers is currently evaluated in clinical trials. Chest wall irradiation accelerates atherosclerotic processes and induces fibrosis. Immune checkpoint inhibitors require consideration for surveillance due to a small risk of severe myocarditis. Cyclin-dependent kinases4/6 inhibitors, cyclophosphamide, taxanes, tyrosine kinase inhibitors, and endocrine therapy have a lower-risk profile for cardiotoxicity. Preventive and management strategies to counteract cancer treatment-related left ventricular dysfunction or heart failure in breast cancer patients should include a comprehensive cardiovascular risk assessment and individual clinical evaluation. This should include both patient and treatment-related factors. Further clinical trials especially on early detection, cardioprevention, and management are urgently needed.
Collapse
Affiliation(s)
- Agneta Månsson Broberg
- Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Jürgen Geisler
- Department of Oncology, Akershus University Hospital, Lørenskog & Institute of Clinical Medicine, University of Oslo, Campus AHUS, Lørenskog, Norway
| | - Suvi Tuohinen
- Heart and Lung Center, Helsinki University Hospital, Helsinki, Finland
| | - Tanja Skytta
- Department of Oncology, Tampere University Hospital, Tampere, Finland
| | - Þórdís Jóna Hrafnkelsdóttir
- Department of Cardiology, Landspitali University Hospital, Reykjavík, Iceland and Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | | | - Elham Hedayati
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
- Department of Breast Cancer, Sarcoma and Endocrine Tumors, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Torbjørn Omland
- Department of Cardiology, Akershus University Hospital, Lørenskog and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Birgitte V. Offersen
- Department of Experimental Clinical Oncology & Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Alexander R. Lyon
- Cardio-Oncology Service, Royal Brompton Hospital and Imperial College London, London, UK
| | - Geeta Gulati
- Department of Cardiology, Oslo University Hospital, Postbox 4950, Ullevål, Nydalen, 0424 Oslo, Norway
- Department of Research, Akershus University Hospital, Lørenskog and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
61
|
Capra D, Monti CB, Luporini AG, Lombardi F, Gumina C, Sironi A, Asti ELG, Bonavina L, Secchi F, Sardanelli F. Computed tomography-derived myocardial extracellular volume: an early biomarker of cardiotoxicity in esophageal cancer patients undergoing radiation therapy. Insights Imaging 2020; 11:120. [PMID: 33226481 PMCID: PMC7683763 DOI: 10.1186/s13244-020-00922-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/08/2020] [Indexed: 12/12/2022] Open
Abstract
Objectives We aimed to assess extracellular volume (ECV) through non-gated, contrast-enhanced computed tomography (CT) before and after radiation therapy (RT) in patients with esophageal cancer (EC). Materials and methods EC patients who had undergone CT before and after RT were retrospectively assessed. Patients with preexisting cardiovascular disease or with heavily artifacted CT were excluded. ECV was calculated using density values for the myocardial septum and blood pool. Data were reported as mean and standard deviation or median and interquartile range according to their distribution; t test or Wilcoxon and Pearson r or Spearman ρ were subsequently used. Results Twenty-one patients with stage ≥ IB EC, aged 64 ± 18 years, were included. Mean and maximum RT doses were 21.2 Gy (16.9–24.1) and 42.5 Gy (41.8–49.2), respectively. At baseline (n = 21), hematocrit was 39% ± 4%, ECV 27.9% ± 3.5%; 35 days (30–38) after RT (n = 20), hematocrit was 36% ± 4%, lower than at baseline (p = 0.002), ECV 30.3% ± 8.3%, higher than at baseline (p = 0.081); at follow-up 420 days (244–624) after RT (n = 13), hematocrit was 36% ± 5%, lower than at baseline (p = 0.030), ECV 31.4% ± 4.5%, higher than at baseline (p = 0.011). No patients showed signs of overt cardiotoxicity. ECV early after RT was moderately positively correlated with maximum RT dose (ρ = 0.50, p = 0.036). Conclusions In EC patients, CT-derived myocardial ECV was increased after RT and may thus appear as a potential early biomarker of cardiotoxicity.
Collapse
Affiliation(s)
- Davide Capra
- Department of Biomedical Sciences for Health, Università Degli Studi Di Milano, Via Mangiagalli 31, 20133, Milano, Italy
| | - Caterina Beatrice Monti
- Department of Biomedical Sciences for Health, Università Degli Studi Di Milano, Via Mangiagalli 31, 20133, Milano, Italy.
| | - Alberto Gianluigi Luporini
- Unit of Medical Oncology, IRCCS Policlinico San Donato, Via Morandi 30, 20097, San Donato Milanese, Italy
| | - Fabrizio Lombardi
- Unit of Radiation Oncology, IRCCS Policlinico San Donato, Via Morandi 30, 20097, San Donato Milanese, Italy
| | - Calogero Gumina
- Unit of Radiation Oncology, IRCCS Policlinico San Donato, Via Morandi 30, 20097, San Donato Milanese, Italy
| | - Andrea Sironi
- Division of General and Foregut Surgery, IRCCS Policlinico San Donato, Via Morandi 30, 20097, San Donato Milanese, Italy
| | - Emanuele Luigi Giuseppe Asti
- Division of General and Foregut Surgery, IRCCS Policlinico San Donato, Via Morandi 30, 20097, San Donato Milanese, Italy
| | - Luigi Bonavina
- Department of Biomedical Sciences for Health, Università Degli Studi Di Milano, Via Mangiagalli 31, 20133, Milano, Italy.,Division of General and Foregut Surgery, IRCCS Policlinico San Donato, Via Morandi 30, 20097, San Donato Milanese, Italy
| | - Francesco Secchi
- Department of Biomedical Sciences for Health, Università Degli Studi Di Milano, Via Mangiagalli 31, 20133, Milano, Italy.,Unit of Radiology, IRCCS Policlinico San Donato, Via Morandi 30, 20097, San Donato Milanese, Italy
| | - Francesco Sardanelli
- Department of Biomedical Sciences for Health, Università Degli Studi Di Milano, Via Mangiagalli 31, 20133, Milano, Italy.,Unit of Radiology, IRCCS Policlinico San Donato, Via Morandi 30, 20097, San Donato Milanese, Italy
| |
Collapse
|
62
|
Elevations in High-Sensitive Cardiac Troponin T and N-Terminal Prohormone Brain Natriuretic Peptide Levels in the Serum Can Predict the Development of Anthracycline-Induced Cardiomyopathy. Am J Ther 2020; 27:e142-e150. [PMID: 30648987 DOI: 10.1097/mjt.0000000000000930] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Anthracyclines remain the cornerstone of the treatment in many cancers including lymphomas, leukemia and sarcomas, and breast cancer. The cardiomyopathy that develops from anthracyclines can lead to heart failure and decreased survival. Multiple mechanisms are involved in the pathophysiology of anthracycline-induced heart failure. STUDY QUESTION We hypothesize that anthracycline-induced cardiac (AIC) pathology can be monitored using a panel of blood biomarkers including high-sensitive cardiac troponin T (hs-cTnT) for myocyte necrosis and N-terminal prohormone brain natriuretic peptide (NT-proBNP) for parietal stress. STUDY DESIGN A prospective, institutionally approved study recruited all patients with cancer scheduled to start anthracycline chemotherapy in the Transylvania University cancer clinics. MEASURES AND OUTCOMES Transthoracic 2D echocardiography and the measurements of NT-proBNP and hs-cTnT plasma levels were performed at the beginning of the study and 3 months and 6 months after anthracycline treatment initiation. RESULTS The plasma levels of hs-cTnT at 3 months (rho = 0.439, P = 0.0001) and 6 months (rho = 0.490, P = 0.0001) are correlated with AIC occurrence. For a cutoff value of hs-cTnT at 3 months > 0.008 ng/mL, we obtained 66.7% sensitivity and 67.9% specificity for developing AIC at 6 months, with a 54.5% positive predictive value and a 87.8% negative predictive value. The NT-proBNP serum levels at 3 months (rho = 0.495, P = 0.0001) and 6 months (rho = 0.638, P = 0.0001) are correlated with an AIC diagnosis at 6 months. For a cutoff value of NT-proBNP at 3 months >118.5 pg/mL, we obtained 80% sensitivity and 79.2% specificity for evolution to AIC at 6 months, with 52.2% positive predictive value and 93.3% negative predictive value. CONCLUSIONS In anthracycline-treated cancer patients, the increase in plasma levels of NT-proBNP and of hs-cTnT can predict the development of anthracycline-induced cardiomyopathy. Early identification of at-risk patients will potentially allow for targeted dose reductions and will diminish the number of patients developing cardiac pathology.
Collapse
|
63
|
The immuno-oncological challenge of COVID-19. ACTA ACUST UNITED AC 2020; 1:946-964. [DOI: 10.1038/s43018-020-00122-3] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 09/02/2020] [Indexed: 02/06/2023]
|
64
|
Neves KB, Montezano AC, Lang NN, Touyz RM. Vascular toxicity associated with anti-angiogenic drugs. Clin Sci (Lond) 2020; 134:2503-2520. [PMID: 32990313 DOI: 10.1042/cs20200308] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/15/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023]
Abstract
Over the past two decades, the treatment of cancer has been revolutionised by the highly successful introduction of novel molecular targeted therapies and immunotherapies, including small-molecule kinase inhibitors and monoclonal antibodies that target angiogenesis by inhibiting vascular endothelial growth factor (VEGF) signaling pathways. Despite their anti-angiogenic and anti-cancer benefits, the use of VEGF inhibitors (VEGFi) and other tyrosine kinase inhibitors (TKIs) has been hampered by potent vascular toxicities especially hypertension and thromboembolism. Molecular processes underlying VEGFi-induced vascular toxicities still remain unclear but inhibition of endothelial NO synthase (eNOS), reduced nitric oxide (NO) production, oxidative stress, activation of the endothelin system, and rarefaction have been implicated. However, the pathophysiological mechanisms still remain elusive and there is an urgent need to better understand exactly how anti-angiogenic drugs cause hypertension and other cardiovascular diseases (CVDs). This is especially important because VEGFi are increasingly being used in combination with other anti-cancer dugs, such as immunotherapies (immune checkpoint inhibitors (ICIs)), other TKIs, drugs that inhibit epigenetic processes (histone deacetylase (HDAC) inhibitor) and poly (adenosine diphosphate-ribose) polymerase (PARP) inhibitors, which may themselves induce cardiovascular injury. Here, we discuss vascular toxicities associated with TKIs, especially VEGFi, and provide an up-to-date overview on molecular mechanisms underlying VEGFi-induced vascular toxicity and cardiovascular sequelae. We also review the vascular effects of VEGFi when used in combination with other modern anti-cancer drugs.
Collapse
Affiliation(s)
- Karla B Neves
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, U.K
| | - Augusto C Montezano
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, U.K
| | - Ninian N Lang
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, U.K
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, U.K
| |
Collapse
|
65
|
Impact of Arterial Hypertension on Doxorubicin-Based Chemotherapy-Induced Subclinical Cardiac Damage in Breast Cancer Patients. Cardiovasc Toxicol 2020; 20:321-327. [PMID: 31782105 DOI: 10.1007/s12012-019-09556-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Advances in oncologic therapies have allowed to achieve better outcomes and longer survival in many patients with breast cancer. Anthracyclines are cytotoxic antibiotics widely used in daily oncology practice. However, anthracyclines cause cardiotoxicity which is a limiting factor of its use. Cumulative dose of anthracyclines is the major cause of induced cardiotoxicity. According to previous clinical trials, the major predisposing high-risk factors for anthracycline-based chemotherapy-induced cardiotoxicity are age, body weight, female gender, radiotherapy, and other diseases such as diabetes and hypertension. Experimental studies in animals confirm that hypertension may be a significant factor predisposing anthracycline-based chemotherapy cardiotoxicity. The main objective of our study was to identify the effect of pre-existing arterial hypertension on the development of subclinical cardiac damage during or after doxorubicin-based chemotherapy in breast cancer patients. The study was performed prospectively between March 2016 and January 2017 in the Hospital of Lithuanian University of Health Sciences Kaunas Clinics Department of Oncology and Department of Cardiology. Data of 73 women with breast cancer treated with doxorubicin-based chemotherapy in outpatient clinic were analyzed. Statistically significant association between pre-existing arterial hypertension and left ventricular systolic dysfunction after completion of chemotherapy was observed (P < 0.004). Our study demonstrated that pre-existing arterial hypertension has a very important role in the development of anthracycline-based chemotherapy-induced cardiotoxicity, despite arterial hypertension control quality. Consequently, further studies evaluating impact of other risk factors and how early and sufficient management of arterial hypertension could influence the development of cardiotoxicity are needed to avoid permanent cardiac damage.
Collapse
|
66
|
Simões R, Silva LM, de Oliveira AN, Alves MT, Pestana RMC, de Souza IDP, Oliveira HHM, Soares CE, Sabino ADP, Gomes KB. Identification of Clinical and Laboratory Variables Associated with Cardiotoxicity Events Due to Doxorubicin in Breast Cancer Patients: A 1-Year Follow-Up Study. Cardiovasc Toxicol 2020; 21:106-114. [PMID: 32844368 DOI: 10.1007/s12012-020-09600-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 08/14/2020] [Indexed: 11/29/2022]
Abstract
Cardiovascular adverse events in patients with breast cancer undergoing chemotherapy (CT) are frequent due to the high cardiotoxic potential of treatments, especially doxorubicin (DOXO). This study aimed to evaluate the association of plasma levels of various biomarkers with cardiotoxicity in women with breast cancer on DOXO-based chemotherapy. In this single center prospective cohort, 80 breast cancer patients who used DOXO as a first-line treatment for cancer were evaluated. Patients were assessed at three time points: before CT (T0), 1 week after (T1) and 12 months after DOXO treatment (T2). The predominant histological classification was ductal carcinoma, n = 72 (90.0%); the most frequent molecular classification was Human epidermal growth factor receptor-type 2 positive (HER2+), n = 34 (43.0%). In patients submitted to complementary treatment with trastuzumab (n = 23), there was no association with cardio-specific biomarkers. Evaluating the clinical variables and the laboratory parameters in T1 and T2 in relation to T0, the reduction any time of N-terminal-pro hormone B-type natriuretic peptide (NT-proBNP), triglycerides and hematocrit levels showed an association with higher cardiotoxicity risk. In addition, increased levels of troponin I (cTnI) and glycated hemoglobin (HbA1c) showed an independent association with the occurrence of cardiotoxicity. These results suggest that the evaluation of these laboratory tests should be included routinely to identify breast cancer patients under DOXO treatment at cardiotoxicity risk.
Collapse
Affiliation(s)
- Ricardo Simões
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil.,Faculdade de Ciências Médicas de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,Fundação Hospitalar do Estado de Minas Gerais (FHEMIG), Belo Horizonte, Minas Gerais, Brazil.,Instituto de Hipertensão, Belo Horizonte, Minas Gerais, Brazil
| | | | | | - Michelle Teodoro Alves
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Rodrigo Mendonça Cardoso Pestana
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Isabella Dâmaris Passos de Souza
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | | | - Cintia Esteves Soares
- Fundação Hospitalar do Estado de Minas Gerais (FHEMIG), Belo Horizonte, Minas Gerais, Brazil
| | - Adriano de Paula Sabino
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Karina Braga Gomes
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil.
| |
Collapse
|
67
|
Zanuso V, Fregoni V, Gervaso L. Side effects of adjuvant chemotherapy and their impact on outcome in elderly breast cancer patients: a cohort study. Future Sci OA 2020; 6:FSO617. [PMID: 33235809 PMCID: PMC7668125 DOI: 10.2144/fsoa-2020-0076] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Aim: Breast cancer patients over the age of 65 are more likely to suffer chemotherapy side effects, with premature discontinuation, which negatively affects survival. Methods: We conducted a retrospective cohort study enrolling breast cancer patients; dose reductions or interruptions of chemotherapy have been collected, as well as side effects. Progression-free survival was determined by Kaplan–Meier and evaluated for its association with reduction/suspension. The study included 128 women (median age: 71). Results: Nineteen patients experienced cardiotoxicity, while dosage of chemotherapy was reduced in 23 patients (18.0%), and 14 (10.9%) had premature interruptions. Dose reduction/interruptions were associated with numerically worse progression-free survival (78.2 vs 94.8 months; p = 0.10). Conclusion: Reduction/discontinuation of chemotherapy due to side effects affected nearly 30% of our population, potentially worsening outcomes. Breast cancer patients over the age of 65 are more likely to suffer chemotherapy side effects, which negatively affect survival. We conducted a retrospective study of 128 elderly breast cancer patients, collecting changes in chemotherapy doses and schedules, as well side effects. Progression-free survival (PFS) was calculated and evaluated for its association with reduction/suspension. Nineteen patients experienced cardiotoxicity, while dosage of chemotherapy was reduced in 23 patients (18.0%), and 14 (10.9%) had premature interruptions. Dose reduction/interruption were associated with numerically worse PFS (78.2 vs 94.8 months; p = 0.10). Occurrence of reduction/discontinuation of chemotherapy and cardiotoxicity in this population could potentially worsen outcomes.
Collapse
Affiliation(s)
- Valentina Zanuso
- Humanitas Cancer Center, IRCCS Istituto Clinico Humanitas, 20089 Rozzano (MI), Italy.,Humanitas University, 20090 Pieve Emanuele (MI), Italy
| | - Vittorio Fregoni
- ASST Valtellina e Alto Lario, UOC Medicina Generale, 23035 Sondalo (SO), Italy
| | - Lorenzo Gervaso
- European Institute of Oncology (IEO) IRCCS, Gatrointestinal Medical Oncology and Neuroendocrine Tumors, 20141 Milan, Italy.,Molecular Medicine Program, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
68
|
Tocchetti CG, Ameri P, de Boer RA, D’Alessandra Y, Russo M, Sorriento D, Ciccarelli M, Kiss B, Bertrand L, Dawson D, Falcao-Pires I, Giacca M, Hamdani N, Linke WA, Mayr M, van der Velden J, Zacchigna S, Ghigo A, Hirsch E, Lyon AR, Görbe A, Ferdinandy P, Madonna R, Heymans S, Thum T. Cardiac dysfunction in cancer patients: beyond direct cardiomyocyte damage of anticancer drugs: novel cardio-oncology insights from the joint 2019 meeting of the ESC Working Groups of Myocardial Function and Cellular Biology of the Heart. Cardiovasc Res 2020; 116:1820-1834. [DOI: 10.1093/cvr/cvaa222] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/17/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022] Open
Abstract
Abstract
In western countries, cardiovascular (CV) disease and cancer are the leading causes of death in the ageing population. Recent epidemiological data suggest that cancer is more frequent in patients with prevalent or incident CV disease, in particular, heart failure (HF). Indeed, there is a tight link in terms of shared risk factors and mechanisms between HF and cancer. HF induced by anticancer therapies has been extensively studied, primarily focusing on the toxic effects that anti-tumour treatments exert on cardiomyocytes. In this Cardio-Oncology update, members of the ESC Working Groups of Myocardial Function and Cellular Biology of the Heart discuss novel evidence interconnecting cardiac dysfunction and cancer via pathways in which cardiomyocytes may be involved but are not central. In particular, the multiple roles of cardiac stromal cells (endothelial cells and fibroblasts) and inflammatory cells are highlighted. Also, the gut microbiota is depicted as a new player at the crossroads between HF and cancer. Finally, the role of non-coding RNAs in Cardio-Oncology is also addressed. All these insights are expected to fuel additional research efforts in the field of Cardio-Oncology.
Collapse
Affiliation(s)
- Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences, Federico II University, via Pansini 5, 80131 Naples, Italy
- Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Federico II University, Naples, Italy
| | - Pietro Ameri
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine, University of Genova, Genova, Italy
| | - Rudolf A de Boer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, AB31, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Yuri D’Alessandra
- Immunology and Functional Genomics Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Michele Russo
- Department of Translational Medical Sciences, Federico II University, via Pansini 5, 80131 Naples, Italy
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Daniela Sorriento
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Michele Ciccarelli
- Department of Medicine Surgery and Odontology, University of Salerno, Salerno, Italy
| | - Bernadett Kiss
- Department of Pharmacology and Pharmacotherapy, Cardiometabolic Research Group and MTA-SE System Pharmacology Research Group, Semmelweis University, Budapest, Hungary
| | - Luc Bertrand
- IREC Institute, Pole of Cardiovascular Research, Université Catholique de Louvain, Brussels, Belgium
| | - Dana Dawson
- School of Medicine and Dentistry, University of Aberdeen, Aberdeen, UK
| | - Ines Falcao-Pires
- Unidade de Investigação e Desenvolvimento Cardiovascular, Departamento de Cirurgia e Fisiologia, Faculdade de Medicina, Universidade do Porto, Portugal
| | - Mauro Giacca
- Department of Medicine, Surgery and Health Sciences and Cardiovascular Department, Centre for Translational Cardiology, Azienda Sanitaria Universitaria Integrata Trieste, Trieste, Italy
- International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
- King’s British Heart Foundation Centre, King’s College London, London, UK
| | - Nazha Hamdani
- Department of Molecular and Experimental Cardiology, Ruhr Universität Bochum, Bochum, Germany
- Department of Cardiology, St. Joseph Hospital, Ruhr University Bochum, Witten, Germany
| | | | - Manuel Mayr
- King’s British Heart Foundation Centre, King’s College London, London, UK
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam UMC, Vrije Universiteit, Amsterdam Cardiovascular Sciences Institute, Amsterdam, The Netherlands
| | - Serena Zacchigna
- Department of Medicine, Surgery and Health Sciences and Cardiovascular Department, Centre for Translational Cardiology, Azienda Sanitaria Universitaria Integrata Trieste, Trieste, Italy
- International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Alexander R Lyon
- Cardio-Oncology Service, Royal Brompton Hospital, Imperial College London, London, UK
| | - Anikó Görbe
- Department of Pharmacology and Pharmacotherapy, Cardiometabolic Research Group and MTA-SE System Pharmacology Research Group, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Cardiometabolic Research Group and MTA-SE System Pharmacology Research Group, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Rosalinda Madonna
- Institute of Cardiology, University of Pisa, Pisa, Italy
- Center for Cardiovascular Biology and Atherosclerosis Research, McGovern School of Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Stephane Heymans
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht University, Maastricht, The Netherlands
- Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Thomas Thum
- Institute for Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| |
Collapse
|
69
|
Chakouri N, Farah C, Matecki S, Amedro P, Vincenti M, Saumet L, Vergely L, Sirvent N, Lacampagne A, Cazorla O. Screening for in-vivo regional contractile defaults to predict the delayed Doxorubicin Cardiotoxicity in Juvenile Rat. Am J Cancer Res 2020; 10:8130-8142. [PMID: 32724462 PMCID: PMC7381739 DOI: 10.7150/thno.47407] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 06/18/2020] [Indexed: 12/24/2022] Open
Abstract
Anthracyclines are key chemotherapeutic agents used in various adult and pediatric cancers, however, their clinical use is limited due to possible congestive heart failure (HF) caused by acute and irreversible cardiotoxicity. Currently, there is no method to predict the future development of the HF in these patients. In order to identify early biomarkers to predict anthracycline cardiotoxicity in long-term survivors of childhood cancer, this longitudinal study aimed to analyze early and late in-vivo regional myocardial anthracycline-induced cardiotoxicity, related to in-vitro cardiac myocytes dysfunction, in a juvenile rat model. Methods: Young male Wistar rats (4 weeks-old) were treated with different cumulative doses of doxorubicin (7.5, 10 or 12.5 mg/kg) or NaCl (0.9%) once a week for 6 weeks by intravenous injection. Cardiac function was evaluated in-vivo by conventional (left ventricular ejection fraction, LVEF) and regional two-dimensional (2D) speckle tracking echocardiography over the 4 months after the last injection. The animals were assigned to preserved (pEF) or reduced EF (rEF) groups at the end of the protocol and were compared to controls. Results: We observed a preferential contractile dysfunction of the base of the heart, further altered in the posterior segment, even in pEF group. The first regional alterations appeared 1 month after chemotherapy. Functional investigation of cardiomyocytes isolated from the LV base 1 month after doxorubicin treatment showed that early in-vivo contractile alterations were associated with both decreased myofilament Ca2+ sensitivity and length-dependent activation. Changes in post-translational modifications (phosphorylation; S-glutathionylation) and protein degradation of the cardiac myosin binding protein-C may contribute to these alterations. Conclusion: Our data suggest that screening of the contractile defaults of the base of the heart by regional 2D strain echocardiography is useful to detect subclinical myocardial dysfunction prior to the development of delayed anthracycline-induced cardiomyopathy in pediatric onco-cardiology.
Collapse
|
70
|
Azizipour N, Avazpour R, Rosenzweig DH, Sawan M, Ajji A. Evolution of Biochip Technology: A Review from Lab-on-a-Chip to Organ-on-a-Chip. MICROMACHINES 2020; 11:E599. [PMID: 32570945 PMCID: PMC7345732 DOI: 10.3390/mi11060599] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 06/09/2020] [Accepted: 06/16/2020] [Indexed: 12/21/2022]
Abstract
Following the advancements in microfluidics and lab-on-a-chip (LOC) technologies, a novel biomedical application for microfluidic based devices has emerged in recent years and microengineered cell culture platforms have been created. These micro-devices, known as organ-on-a-chip (OOC) platforms mimic the in vivo like microenvironment of living organs and offer more physiologically relevant in vitro models of human organs. Consequently, the concept of OOC has gained great attention from researchers in the field worldwide to offer powerful tools for biomedical researches including disease modeling, drug development, etc. This review highlights the background of biochip development. Herein, we focus on applications of LOC devices as a versatile tool for POC applications. We also review current progress in OOC platforms towards body-on-a-chip, and we provide concluding remarks and future perspectives for OOC platforms for POC applications.
Collapse
Affiliation(s)
- Neda Azizipour
- Institut de Génie Biomédical, Polytechnique Montréal, Montreal, QC H3C 3A7, Canada;
| | - Rahi Avazpour
- Department of Chemical Engineering, Polytechnique Montréal, Montreal, QC H3C 3A7, Canada;
| | - Derek H. Rosenzweig
- Department of Surgery, McGill University, Montreal, QC H3G 1A4, Canada;
- Injury, Repair and Recovery Program, Research Institute of McGill University Health Centre, Montreal, QC H3H 2R9, Canada
| | - Mohamad Sawan
- Polystim Neurotech Laboratory, Electrical Engineering Department, Polytechnique Montreal, QC H3T 1J4, Canada
- CenBRAIN Laboratory, School of Engineering, Westlake Institute for Advanced Study, Westlake University, Hangzhou 310024, China
| | - Abdellah Ajji
- Institut de Génie Biomédical, Polytechnique Montréal, Montreal, QC H3C 3A7, Canada;
- NSERC-Industry Chair, CREPEC, Chemical Engineering Department, Polytechnique Montreal, Montreal, QC H3C 3A7, Canada
| |
Collapse
|
71
|
Verberne HJ, Verschure DO. Anthracycline-induced cardiotoxicity: Is there a role for myocardial 123I-mIBG scintigraphy? J Nucl Cardiol 2020; 27:940-942. [PMID: 30603895 DOI: 10.1007/s12350-018-01584-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 12/14/2018] [Indexed: 10/27/2022]
Affiliation(s)
- Hein J Verberne
- Department of Radiology and Nuclear Medicine, F2-238, Amsterdam UMC, Location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| | - Derk O Verschure
- Department of Radiology and Nuclear Medicine, F2-238, Amsterdam UMC, Location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Department of Cardiology, Zaans Medical Center, Zaandam, The Netherlands
| |
Collapse
|
72
|
Anakwue R. Cytotoxic-induced heart failure among breast cancer patients in Nigeria: A call to prevent today's cancer patients from being tomorrow's cardiac patients. Ann Afr Med 2020; 19:1-7. [PMID: 32174608 PMCID: PMC7189886 DOI: 10.4103/aam.aam_24_19] [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] [Indexed: 01/07/2023] Open
Abstract
We report three cases of heart failure (HF) associated with the use of cytotoxic drugs such as anthracycline, cyclophosphamide, and 5-fluorouracil in the treatment of breast cancer in Nigerians. The patients had systolic and diastolic HF: HF with reduced ejection fraction and preserved ejection fraction. The prevalence of breast cancer is increasing across Africa, and cytotoxics are some of the most common and best drugs used during management. The cardiotoxicity caused by these drugs limits their use as chemotherapeutic agents. Cytotoxic-induced HF is a preventable and manageable cause of cardiovascular disease (CVD) in Nigeria and Africa. This article discusses the pathophysiology of cytotoxic-induced HF and presents the risk factors that impair cardiovascular function. The importance of proper assessment and the prophylactic and therapeutic measures in the management of cytotoxic-induced HF are emphasized. The peculiar challenges in the management of cytotoxic-induced HF in Nigeria were also discussed. The need for early involvement of cardiologists by oncologists to improve on the chemotherapeutic and cardiovascular outcome in the management of patients with breast cancer was stressed. Perhaps, it is time to birth a new discipline of cardiooncology in Nigeria.
Collapse
Affiliation(s)
- Raphael Anakwue
- Department of Medicine; Department of Pharmacology and Therapeutics, Faculty of Medical Sciences, College of Medicine, University of Nigeria, Nsukka, Nigeria
| |
Collapse
|
73
|
Gallo S, Spilinga M, Albano R, Ferrauto G, Di Gregorio E, Casanova E, Balmativola D, Bonzano A, Boccaccio C, Sapino A, Comoglio PM, Crepaldi T. Activation of the MET receptor attenuates doxorubicin-induced cardiotoxicity in vivo and in vitro. Br J Pharmacol 2020; 177:3107-3122. [PMID: 32133617 PMCID: PMC7280013 DOI: 10.1111/bph.15039] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 02/11/2020] [Accepted: 02/19/2020] [Indexed: 12/16/2022] Open
Abstract
Background and Purpose Doxorubicin anti‐cancer therapy is associated with cardiotoxicity, resulting from DNA damage response (DDR). Hepatocyte growth factor (HGF) protects cardiomyocytes from injury, but its effective use is compromised by low biodistribution. In this study, we have investigated whether the activation of the HGF receptor—encoded by the Met gene—by an agonist monoclonal antibody (mAb) could protect against doxorubicin‐induced cardiotoxicity. Experimental Approach The mAb (5 mg·kg−1) was injected in vivo into C57BL/6J mice, before doxorubicin (three doses of 7 mg·kg−1). Cardiac functions were evaluated through MRI after treatment termination. Heart histological staining and mRNA levels of genes associated with heart failure (Acta1 and Nppa), inflammation (IL‐6), and fibrosis (Ctgf, Col1a2, Timp1, and Mmp9) were assessed. MAb (100 nM) was administered in vitro to H9c2 cardiomyoblasts before addition of doxorubicin (25 μM). DDR and apoptosis markers were evaluated by quantitative western blotting, flow cytometry, and immunofluorescence. Stattic was used for pharmacological inactivation of STAT3. Key Results In vivo, administration of the mAb alleviated doxorubicin‐induced cardiac dysfunction and fibrosis. In vitro, mAb mimicked the response to HGF by (a) inhibiting histone H2AX phosphorylation at S139, (b) quenching the expression of the DNA repair enzyme PARP1, and (c) reducing the proteolytic activation of caspase 3. The MET‐driven cardioprotection involved, at least in vitro, the phosphorylation of STAT3. Conclusion and Implications The MET agonist mAb provides a new tool for cardioprotection against anthracycline cardiotoxicity.
Collapse
Affiliation(s)
- Simona Gallo
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy
| | - Martina Spilinga
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy.,Department of Oncology, University of Turin, Turin, Italy
| | | | - Giuseppe Ferrauto
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Enza Di Gregorio
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Elena Casanova
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy
| | | | | | - Carla Boccaccio
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy.,Department of Oncology, University of Turin, Turin, Italy
| | - Anna Sapino
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy.,Department of Medical Sciences, University of Turin, Turin, Italy
| | | | - Tiziana Crepaldi
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy.,Department of Oncology, University of Turin, Turin, Italy
| |
Collapse
|
74
|
TRANSLATION MEDICINE, BIOMEDICINE AND MEDICAL BIOTECHNOLOGY: THE TRANSITION TO PERSONALIZED MEDICINE. BIOTECHNOLOGIA ACTA 2020. [DOI: 10.15407/biotech13.02.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
75
|
Cardinale D, Iacopo F, Cipolla CM. Cardiotoxicity of Anthracyclines. Front Cardiovasc Med 2020; 7:26. [PMID: 32258060 PMCID: PMC7093379 DOI: 10.3389/fcvm.2020.00026] [Citation(s) in RCA: 225] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 02/18/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiotoxicity is a feared side effect that may limit the clinical use of anthracyclines. It may indeed affect the quality of life and survival of patients with cancer, regardless of oncological prognosis. This paper provides an overview of anthracycline-induced cardiotoxicity in terms of definition, classification, incidence, risk factors, possible mechanisms, diagnosis, and treatment. We also report effective strategies for preventing cardiotoxicity. In addition, we discuss limiting current approaches, the need for a new classification, and early cardiotoxicity detection and treatment. Probably, anthracycline-induced cardiotoxicity is a continuous phenomenon that starts from myocardial cell injury; it is followed by left ventricular ejection fraction (LVEF) and, if not diagnosed and cured early, progressively leads to symptomatic heart failure. Anthracycline-induced cardiotoxicity can be detected at a preclinical phase. The role of biomarkers, in particular troponins, in identifying subclinical cardiotoxicity and its therapy with angiotensin-converting enzyme inhibitors (mainly enalapril) to prevent LVEF reduction is a recognized and effective strategy. If cardiac dysfunction has already occurred, partial or complete LVEF recovery may still be obtained in case of early detection of cardiotoxicity and prompt heart failure treatment.
Collapse
Affiliation(s)
- Daniela Cardinale
- Cardioncology Unit, European Institute of Oncology, IRCCS, Milan, Italy
| | - Fabiani Iacopo
- Cardioncology Unit, European Institute of Oncology, IRCCS, Milan, Italy
| | | |
Collapse
|
76
|
MicroRNAs in Cancer Treatment-Induced Cardiotoxicity. Cancers (Basel) 2020; 12:cancers12030704. [PMID: 32192047 PMCID: PMC7140035 DOI: 10.3390/cancers12030704] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/12/2020] [Accepted: 03/13/2020] [Indexed: 12/20/2022] Open
Abstract
Cancer treatment has made significant progress in the cure of different types of tumors. Nevertheless, its clinical use is limited by unwanted cardiotoxicity. Aside from the conventional chemotherapy approaches, even the most newly developed, i.e., molecularly targeted therapy and immunotherapy, exhibit a similar frequency and severity of toxicities that range from subclinical ventricular dysfunction to severe cardiomyopathy and, ultimately, congestive heart failure. Specific mechanisms leading to cardiotoxicity still remain to be elucidated. For instance, oxidative stress and DNA damage are considered key players in mediating cardiotoxicity in different treatments. microRNAs (miRNAs) act as key regulators in cell proliferation, cell death, apoptosis, and cell differentiation. Their dysregulation has been associated with adverse cardiac remodeling and toxicity. This review provides an overview of the cardiotoxicity induced by different oncologic treatments and potential miRNAs involved in this effect that could be used as possible therapeutic targets.
Collapse
|
77
|
Aldemir MN, Simsek M, Kara AV, Ozcicek F, Mammadov R, Yazıcı GN, Sunar M, Coskun R, Gulaboglu M, Suleyman H. The effect of adenosine triphosphate on sunitinib-induced cardiac injury in rats. Hum Exp Toxicol 2020; 39:1046-1053. [PMID: 32131635 DOI: 10.1177/0960327120909874] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In this study, we aimed to show the effect of adenosine 5'-triphosphate (ATP) on sunitinib-induced cardiac injury in rats. The rats (n = 30) were divided equally into three groups as sunitinib group (SG), sunitinib plus ATP group (SAG), and healthy group (HG); 2 mg/kg ATP was injected intraperitoneally (ip) to the SAG group. Same volume normal saline as solvent was administered ip to the other two groups. After 1 h, 25 mg/kg sunitinib was applied orally via catheter to stomach in the SAG and SG groups. This procedure was repeated once daily for 5 weeks. At the end of this period, all animals were sacrificed and their cardiac tissue was removed. Malondialdehyde (MDA), total glutathione (tGSH), tumor necrosis factor α (TNF-α), and nuclear factor κB (NF-κB) levels in rats' cardiac tissues and troponin I (Tp-I) levels in rats' blood samples were evaluated. Histopathological analysis was also performed in cardiac tissues of the animals. MDA, TNF-α, NF-κB, and Tp-I levels were higher in the SG group compared to the SAG and HG groups (p < 0.001). tGSH levels of the SG group were lower than the SAG and HG groups (p < 0.001). The structure and morphology of cardiac muscle fibers and blood vessels were normal in the control group. In the SG group, obvious cardiac muscle tissue damage with dilated myofibers, locally atrophic myofibers, and congested blood vessels were observed. In the SAG group, marked amelioration in these findings was observed. We showed this for the first time that ATP administration exerts a protective effect against cardiac effects of sunitinib.
Collapse
Affiliation(s)
- M N Aldemir
- Department of Medical Oncology, Faculty of Medicine, Yuzuncu Yil University, Van, Turkey
| | - M Simsek
- Department of Medical Oncology, Yozgat City Hospital, Yozgat, Turkey
| | - A V Kara
- Department of Nephrology, Faculty of Medicine, Erzincan University, Erzincan, Turkey
| | - F Ozcicek
- Department of Internal Medicine, Faculty of Medicine, Erzincan University, Erzincan, Turkey
| | - R Mammadov
- Department Pharmacology, Faculty of Medicine, Erzincan University, Erzincan, Turkey
| | - G N Yazıcı
- Department of Histology and Embryology, Faculty of Medicine, Erzincan University, Erzincan, Turkey
| | - M Sunar
- Department of Anatomy, Faculty of Medicine, Erzincan University, Erzincan, Turkey
| | - R Coskun
- Department of Cardiology, Faculty of Medicine, Erzincan University, Erzincan, Turkey
| | - M Gulaboglu
- Department of Biochemistry, Faculty of Pharmacy, Ataturk University, Erzurum, Turkey
| | - H Suleyman
- Department of Pharmacology, Faculty of Medicine, Erzincan University, Erzincan, Turkey
| |
Collapse
|
78
|
Wang B, Yu Y, Zhang Y, Hao X, Zhao H, Yang S, Sun Q, Wang Y. Speckle tracking echocardiography in the early detection and prediction of anthracycline cardiotoxicity in diffuse large B-cell lymphoma treated with (R)-CHOP regimen. Echocardiography 2020; 37:421-428. [PMID: 32112477 DOI: 10.1111/echo.14622] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 01/20/2020] [Accepted: 02/06/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Despite speckle tracking echocardiography (STE) has emerged as a sensitive technique for identifying myocardial dysfunction, there are little data available on the appropriate timing to perform STE in the serial assessment after anthracycline administration. Moreover, further uncertainty is increased in the context of STE application in diffuse large B-cell lymphoma (DLBCL) research, as most recommendations are inferred by studies conducted primarily in breast cancer. OBJECTIVE This study aimed to determine whether early measurement of strain parameters derived by STE could predict the development of cardiotoxicity. METHODS Sixty-five patients were included in the final analysis. The patients were evaluated at baseline, after the third cycle and sixth-eighth cycle, and during follow-up. Global longitudinal strain (GLS) was analyzed using STE, and left ventricular ejection fraction (LVEF) was calculated by real time 3D echocardiography (RT3DE). RESULTS There was a significant decrease in GLS after the third cycle of chemotherapy and remained decreased during subsequent follow-up, whereas LVEF decreased only at follow-up. A percentage reduction in GLS of 13.8% between baseline and the third cycle of chemotherapy was the best predictor of further LVEF reduction. CONCLUSION Earlier monitoring timing and more accurate assessment methods might be helpful in the prevention of irreversible heart failure.
Collapse
Affiliation(s)
- Baozhen Wang
- Department of Ultrasound, Cangzhou People's Hospital, Cangzhou, China
| | - Yang Yu
- Medical Imaging Center, Cangzhou People's Hospital, Cangzhou, China
| | - Yue Zhang
- Department of Ultrasound, Cangzhou People's Hospital, Cangzhou, China
| | - Xiaoyi Hao
- Department of Ultrasound, Cangzhou People's Hospital, Cangzhou, China
| | - Hong Zhao
- Department of Ultrasound, Cangzhou People's Hospital, Cangzhou, China
| | - Shan Yang
- Department of Ultrasound, Cangzhou People's Hospital, Cangzhou, China
| | - Qianqian Sun
- Department of Ultrasound, Cangzhou People's Hospital, Cangzhou, China
| | - Yue Wang
- Medical Imaging Center, Cangzhou People's Hospital, Cangzhou, China
| |
Collapse
|
79
|
Curigliano G, Lenihan D, Fradley M, Ganatra S, Barac A, Blaes A, Herrmann J, Porter C, Lyon AR, Lancellotti P, Patel A, DeCara J, Mitchell J, Harrison E, Moslehi J, Witteles R, Calabro MG, Orecchia R, de Azambuja E, Zamorano JL, Krone R, Iakobishvili Z, Carver J, Armenian S, Ky B, Cardinale D, Cipolla CM, Dent S, Jordan K. Management of cardiac disease in cancer patients throughout oncological treatment: ESMO consensus recommendations. Ann Oncol 2020; 31:171-190. [PMID: 31959335 PMCID: PMC8019325 DOI: 10.1016/j.annonc.2019.10.023] [Citation(s) in RCA: 567] [Impact Index Per Article: 113.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 12/13/2022] Open
Abstract
Cancer and cardiovascular (CV) disease are the most prevalent diseases in the developed world. Evidence increasingly shows that these conditions are interlinked through common risk factors, coincident in an ageing population, and are connected biologically through some deleterious effects of anticancer treatment on CV health. Anticancer therapies can cause a wide spectrum of short- and long-term cardiotoxic effects. An explosion of novel cancer therapies has revolutionised this field and dramatically altered cancer prognosis. Nevertheless, these new therapies have introduced unexpected CV complications beyond heart failure. Common CV toxicities related to cancer therapy are defined, along with suggested strategies for prevention, detection and treatment. This ESMO consensus article proposes to define CV toxicities related to cancer or its therapies and provide guidance regarding prevention, screening, monitoring and treatment of CV toxicity. The majority of anticancer therapies are associated with some CV toxicity, ranging from asymptomatic and transient to more clinically significant and long-lasting cardiac events. It is critical however, that concerns about potential CV damage resulting from anticancer therapies should be weighed against the potential benefits of cancer therapy, including benefits in overall survival. CV disease in patients with cancer is complex and treatment needs to be individualised. The scope of cardio-oncology is wide and includes prevention, detection, monitoring and treatment of CV toxicity related to cancer therapy, and also ensuring the safe development of future novel cancer treatments that minimise the impact on CV health. It is anticipated that the management strategies discussed herein will be suitable for the majority of patients. Nonetheless, the clinical judgment of physicians remains extremely important; hence, when using these best clinical practices to inform treatment options and decisions, practitioners should also consider the individual circumstances of their patients on a case-by-case basis.
Collapse
Affiliation(s)
- G. Curigliano
- European Institute of Oncology IRCCS, Milan
- Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy
| | - D. Lenihan
- Cardiovascular Division, Cardio-Oncology Center of Excellence, Washington University Medical Center, St. Louis
| | - M. Fradley
- Cardio-oncology Program, Division of Cardiovascular Medicine, Morsani College of Medicine and H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, Tampa
| | - S. Ganatra
- Cardio-Oncology Program, Lahey Medical Center, Burlington
| | - A. Barac
- Cardio-Oncology Program, Medstar Heart and Vascular Institute and MedStar Georgetown Cancer Institute, Georgetown University Hospital, Washington DC
| | - A. Blaes
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis
| | | | - C. Porter
- University of Kansas Medical Center, Lawrence, USA
| | - A. R. Lyon
- Royal Brompton Hospital and Imperial College, London, UK
| | - P. Lancellotti
- GIGA Cardiovascular Sciences, Acute Care Unit, Heart Failure Clinic, CHU Sart Tilman, University Hospital of Liège, Liège, Belgium
| | - A. Patel
- Morsani College of Medicine, University of South Florida, Tampa
| | - J. DeCara
- Medicine Section of Cardiology, University of Chicago, Chicago
| | - J. Mitchell
- Washington University Medical Center, St. Louis
| | - E. Harrison
- HCA Memorial Hospital and University of South Florida, Tampa
| | - J. Moslehi
- Vanderbilt University School of Medicine, Nashville
| | - R. Witteles
- Division of Cardiovascular Medicine, Falk CVRC, Stanford University School of Medicine, Stanford, USA
| | - M. G. Calabro
- Department of Anesthesia and Intensive Care, IRCCS, San Raffaele Scientific Institute, Milan, Italy
| | | | - E. de Azambuja
- Institut Jules Bordet and L’Université Libre de Bruxelles, Brussels, Belgium
| | | | - R. Krone
- Division of Cardiology, Washington University, St. Louis, USA
| | - Z. Iakobishvili
- Clalit Health Services, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - J. Carver
- Division of Cardiology, Abramson Cancer Center, Hospital of the University of Pennsylvania, Philadelphia
| | - S. Armenian
- Department of Population Sciences, City of Hope Comprehensive Cancer Center, Duarte
| | - B. Ky
- University of Pennsylvania School of Medicine, Philadelphia, USA
| | - D. Cardinale
- Cardioncology Unit, European Institute of Oncology, IRCCS, Milan
| | - C. M. Cipolla
- Cardiology Department, European Institute of Oncology, IRCCS, Milan, Italy
| | - S. Dent
- Duke Cancer Institute, Duke University, Durham, USA
| | - K. Jordan
- Department of Medicine V, Hematology, Oncology and Rheumatology, University of Heidelberg, Heidelberg, Germany
| | - ESMO Guidelines Committee
- Correspondence to: ESMO Guidelines Committee, ESMO Head Office, Via Ginevra 4, CH-6900 Lugano, Switzerland, (ESMO Guidelines Committee)
| |
Collapse
|
80
|
Al-Sadawi M, Singh K, Capric V, Mohiuddin A, Haddadin M, Nunez A, Shaikh S, Bukharovich I, McFarlane SI. Incidence and Associated Risk Factors of Chemotherapy-Induced Cardiomyopathy in the African American and Afro-Caribbean Populations. INTERNATIONAL JOURNAL OF CLINICAL RESEARCH & TRIALS 2020; 5:154. [PMID: 33511317 PMCID: PMC7839991 DOI: 10.15344/2456-8007/2020/154] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND Chemotherapy-induced cardiomyopathy (CICM) and heart failure are major complications of cancer therapeutics and can result in significant morbidity and mortality. There is limited data on the incidence and risk factors of CICM in African American and Afro-Caribbean patients. METHODS We performed a retrospective chart review to evaluate the baseline characteristics that may predispose to CICM. Patients were African American and Afro-Caribbean ethnicity. Data was collected between 2014 to 2018. Patients had transthoracic echocardiogram (TTE) or multigated acquisition scan (MUGA) prior to cancer therapy and every 3 months thereafter, until the end of the regimen. CICM was defined as a ≥16% reduction in LVEF or ≥10% reduction in LVEF to a value <50%. RESULTS A total of 230 patients were studied, with a mean age of 54±12 years with 91% were females, BMI 30±4, 81% were taking anthracyclines, 87% were on Trastuzumab while 5% were receiving both medications. The prevalence of comorbidities was as follows: hypertension 8%, diabetes mellitus 8%, ESRD 8%, dyslipidemia 8%, CAD 7%. The incidence of CICM was 7% overall, while it was 6% and 8% for patients taking Anthracyclines and Trastuzumab, respectively. CICM was associated with dyslipidemia (r= .22, p= .001), hypertension (r= .12, p= .05), baseline ejection fraction (r= -.21, p= .001) and concomitant use of radiation therapy (r= .147, p= .02), but not with age, gender, beta blocker use, angiotensin converting enzyme inhibitor use, number of chemotherapy cycles or stage of the malignancy. On multivariate analysis CICM was independently associated with baseline ejection fraction (β= -.193, P= .003) and dyslipidemia (β= -.20, P= .003). CONCLUSION The incidence of CICM in African Americans and Afro-Caribbean is higher than reported in the general population. Dyslipidemia and baseline ejection fraction were seen as the major risk factors associated with the higher incidence of CICM.
Collapse
Affiliation(s)
- Mohammed Al-Sadawi
- Department of Internal Medicine, State University of New York: Downstate Medical Center, Brooklyn, NY 11203, United States
| | - Kurnvir Singh
- Department of Internal Medicine, State University of New York: Downstate Medical Center, Brooklyn, NY 11203, United States
| | - Violeta Capric
- Department of Internal Medicine, State University of New York: Downstate Medical Center, Brooklyn, NY 11203, United States
| | - Amena Mohiuddin
- Department of Internal Medicine, State University of New York: Downstate Medical Center, Brooklyn, NY 11203, United States
| | - Michael Haddadin
- Department of Internal Medicine, State University of New York: Downstate Medical Center, Brooklyn, NY 11203, United States
| | - Arismendy Nunez
- Department of Cardiovascular Medicine, State University of New York: Downstate Medical Center, Brooklyn, NY 11203, United States
| | - Shakil Shaikh
- Department of Cardiovascular Medicine, State University of New York: Downstate Medical Center, Brooklyn, NY 11203, United States
| | - Inna Bukharovich
- Department of Cardiovascular Medicine, Kings County Hospital Center, Brooklyn, NY 11203, United States
| | - Samy I. McFarlane
- Department of Internal Medicine, State University of New York: Downstate Medical Center, Brooklyn, NY 11203, United States,Corresponding Author: Prof. Samy I. McFarlane, College of Medicine, Department of Medicine, Division of Endocrinology, Internal Medicine Residency Program Director, State University of New York, Downstate Medical Center, 450 Clarkson Ave, Box 50, Brooklyn, New York, 11203-2098, USA. Phone 718-270-6707, Fax 718-270-4488;
| |
Collapse
|
81
|
Kang Y, Assuncao BL, Denduluri S, McCurdy S, Luger S, Lefebvre B, Carver J, Scherrer-Crosbie M. Symptomatic Heart Failure in Acute Leukemia Patients Treated With Anthracyclines. JACC: CARDIOONCOLOGY 2019; 1:208-217. [PMID: 32905430 PMCID: PMC7472996 DOI: 10.1016/j.jaccao.2019.10.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Objectives The purpose of this study was to investigate the occurrence and develop a risk score for heart failure (HF) in acute leukemia. Background Knowledge is scarce regarding the incidence and risk factors of symptomatic HF in patients with acute leukemia. Methods Baseline clinical and echocardiographic parameters, including indices of cardiac function (left ventricular ejection fraction and myocardial strain [global longitudinal strain; GLS]), were obtained in 450 patients with acute leukemia treated with anthracyclines, before chemotherapy initiation. Potential risk factors for HF were evaluated using Fine and Gray’s regression analysis, and from this, a 21-point risk score was generated. Results Forty patients (8.9%) developed HF. The HF risk score included a baseline GLS >−15% (indicative of greater impairment) (6 points), baseline left ventricular ejection fraction <50%, pre-existing cardiovascular disease, acute myeloid leukemia (4 points each), cumulative anthracycline dose ≥250 mg/m2 (2 points), and age >60 years (1 point). Patients were stratified into low (score 0 to 6), moderate (score 7 to 13), and high risk (score 14 to 21). The estimated 1-year cumulative incidence of HF for low-, moderate-, and high-risk groups was 1.0%, 13.6%, and 35.0%, respectively (p < 0.001). The HF risk score was also predictive of all-cause mortality (p < 0.001). After adjustment for age and leukemia type, however, only GLS was significantly associated with all-cause mortality (hazard ratio: 1.73; 95% confidence interval: 1.30 to 2.31; p < 0.001). Conclusions We developed a baseline risk score to determine risk of HF in patients with acute leukemia. Additional studies are needed to determine the external validity of these findings.
Collapse
Affiliation(s)
- Yu Kang
- Division of Cardiovascular Diseases, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Bruna Leal Assuncao
- Departamento de Medicina, Universidade Federal de Sao Paulo, UNIFESP, Sao Paulo-SP, Brazil
| | - Srinivas Denduluri
- Division of Cardiovascular Diseases, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Shannon McCurdy
- Division of Hematology and Oncology Diseases, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Selina Luger
- Division of Hematology and Oncology Diseases, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Bénédicte Lefebvre
- Division of Cardiovascular Diseases, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Joseph Carver
- Division of Cardiovascular Diseases, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Marielle Scherrer-Crosbie
- Division of Cardiovascular Diseases, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Address for correspondence: Dr. Marielle Scherrer-Crosbie, Division of Cardiovascular Diseases, Department of Medicine, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, Pennsylvania 19104, USA. @mariellesc1
| |
Collapse
|
82
|
miR-200a Attenuated Doxorubicin-Induced Cardiotoxicity through Upregulation of Nrf2 in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:1512326. [PMID: 31781322 PMCID: PMC6875222 DOI: 10.1155/2019/1512326] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 08/07/2019] [Accepted: 08/31/2019] [Indexed: 02/07/2023]
Abstract
Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) was closely involved in doxorubicin- (DOX-) induced cardiotoxicity. MicroRNA-200a (miR-200a) could target Keap1 mRNA and promote degradation of Keap1 mRNA, resulting in Nrf2 activation. However, the role of miR-200a in DOX-related cardiotoxicity remained unclear. Our study is aimed at investigating the effect of miR-200a on DOX-induced cardiotoxicity in mice. For cardiotropic expression, male mice received an injection of an adeno-associated virus 9 (AAV9) system carrying miR-200a or miR-scramble. Four weeks later, mice received a single intraperitoneal injection of DOX at 15 mg/kg. In our study, we found that miR-200a mRNA was the only microRNA that was significantly decreased in DOX-treated mice and H9c2 cells. miR-200a supplementation blocked whole-body wasting and heart atrophy caused by acute DOX injection, decreased the levels of cardiac troponin I and the N-terminal probrain natriuretic peptide, and improved cardiac and adult cardiomyocyte contractile function. Moreover, miR-200a reduced oxidative stress and cardiac apoptosis without affecting matrix metalloproteinase and inflammatory factors in mice with acute DOX injection. miR-200a also attenuated DOX-induced oxidative injury and cell loss in vitro. As expected, we found that miR-200a activated Nrf2 and Nrf2 deficiency abolished the protection provided by miR-200a supplementation in mice. miR-200a also provided cardiac benefits in a chronic model of DOX-induced cardiotoxicity. In conclusion, miR-200a protected against DOX-induced cardiotoxicity via activation of the Nrf2 signaling pathway. Our data suggest that miR-200a may represent a new cardioprotective strategy against DOX-induced cardiotoxicity.
Collapse
|
83
|
Hohmann C, Baldus S, Pfister R. [Curing cancer and protecting the heart : Challenges in cardio-oncology in the era of modern tumor treatment]. Herz 2019; 44:175-188. [PMID: 30847511 DOI: 10.1007/s00059-019-4787-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Recent advances in the medical oncological treatment options for cancer have led to a clear improvement in the survival rate worldwide; however, many of the recently developed new drugs are directly or indirectly associated with cardiovascular side effects. Cardiovascular diseases are already the most frequent non-cancerous cause of death in tumor patients. Prevention, early detection of these complications, correct management and timely initiation of specific cardiac medical treatment are the key for an improvement of the cardiovascular prognosis. This article provides an overview and comprehensive summary of the possible cardiotoxic side effects of important oncological therapies and offers possible practical strategies with respect to risk stratification, cardiological follow-up care and management approaches for chemotherapy-induced left ventricular dysfunction.
Collapse
Affiliation(s)
- C Hohmann
- Klinik III für Innere Medizin, Herzzentrum, Universität zu Köln, Kerpener Str. 62, 50937, Köln, Deutschland
| | - S Baldus
- Klinik III für Innere Medizin, Herzzentrum, Universität zu Köln, Kerpener Str. 62, 50937, Köln, Deutschland
| | - R Pfister
- Klinik III für Innere Medizin, Herzzentrum, Universität zu Köln, Kerpener Str. 62, 50937, Köln, Deutschland.
| |
Collapse
|
84
|
Agha A, Zarifa A, Kim P, Iliescu C, Gladish G, Hassan S, Palaskas N, Durand JB, Lu Y, Lopez-Mattei J. The Role of Cardiovascular Imaging and Serum Biomarkers in Identifying Cardiotoxicity Related to Cancer Therapeutics. Methodist Debakey Cardiovasc J 2019; 15:258-266. [PMID: 31988686 DOI: 10.14797/mdcj-15-4-258] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Innovations and discoveries in cancer therapeutics have improved survival rates in patients with various types of malignancies. At the same time, physicians are identifying an increased number of patients with treatment-related cardiotoxicity. It is imperative that physicians recognize early treatment-related adverse effects to determine the safest therapeutic options for patients with cancer. This manuscript evaluates the role of cardiovascular imaging and biomarkers in identifying cardiotoxicity trigged by various chemotherapeutic agents and summarizes expert consensus statements regarding cardiotoxicity monitoring.
Collapse
Affiliation(s)
- Ali Agha
- MCGOVERN MEDICAL SCHOOL, UNIVERSITY OF TEXAS HEALTH SCIENCE CENTER AT HOUSTON, HOUSTON, TEXAS
| | | | - Peter Kim
- UNIVERSITY OF TEXAS MD ANDERSON CANCER CENTER, HOUSTON, TEXAS
| | - Cezar Iliescu
- UNIVERSITY OF TEXAS MD ANDERSON CANCER CENTER, HOUSTON, TEXAS
| | - Greg Gladish
- UNIVERSITY OF TEXAS MD ANDERSON CANCER CENTER, HOUSTON, TEXAS
| | - Saamir Hassan
- UNIVERSITY OF TEXAS MD ANDERSON CANCER CENTER, HOUSTON, TEXAS
| | | | - Jean B Durand
- UNIVERSITY OF TEXAS MD ANDERSON CANCER CENTER, HOUSTON, TEXAS
| | - Yang Lu
- UNIVERSITY OF TEXAS MD ANDERSON CANCER CENTER, HOUSTON, TEXAS
| | | |
Collapse
|
85
|
Timóteo AT, Moura Branco L, Filipe F, Galrinho A, Rio P, Portugal G, Oliveira S, Ferreira RC. Cardiotoxicity in breast cancer treatment: What about left ventricular diastolic function and left atrial function? Echocardiography 2019; 36:1806-1813. [PMID: 31573712 DOI: 10.1111/echo.14487] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 09/08/2019] [Indexed: 11/29/2022] Open
Abstract
AIMS Cardiotoxicity is a possible complication of cancer treatment, particularly with anthracyclines and anti-HER2 drugs. Systolic dysfunction has already been described. Diastolic dysfunction and left atrial function are less studied. We sought to analyze the impact of cardiotoxic treatments on left ventricular diastolic function and left atrial (LA) function. METHODS AND RESULTS Retrospective study of 100 patients (all women, with a mean age of 54 ± 12 years) with three exams in the span of 1 year during treatment for breast cancer. Patients with previous cancer treatment, coronary artery disease, significant valvular disease, and atrial arrhythmias were excluded. Diastolic dysfunction was classified according to international guidelines and left atrial strain was analyzed by two-dimensional speckle tracking. In our sample, 74% received anthracyclines, 83% anti-HER2, and 76% radiation treatment. In the follow-up, 20% developed new or worsening diastolic dysfunction. Age was the only independent predictor (OR 1.93, 95% CI 1.04-3.58, P = .037). In left atrial function, only the contractile function was significantly reduced in 20.8% of the patients and age was also the only independent predictor, but with a protective effect (OR 0.51, 95% CI 0.28-0.91, P = .023). CONCLUSIONS During breast cancer treatment, 20% of the patients develop new or worsening diastolic dysfunction, being age the main determinant, suggesting higher impact of chemotherapy in older patients. Contractile left atrial function is also compromised but, in this case, age seems to be protective. Our results support a stricter surveillance in older patients together to eventually adjust chemotherapy regimens.
Collapse
Affiliation(s)
- Ana Teresa Timóteo
- Cardiology Department, Santa Marta Hospital, Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
| | - Luisa Moura Branco
- Cardiology Department, Santa Marta Hospital, Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
| | - Frederico Filipe
- Oncology Department, Santo António Capuchos Hospital, Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
| | - Ana Galrinho
- Cardiology Department, Santa Marta Hospital, Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
| | - Pedro Rio
- Cardiology Department, Santa Marta Hospital, Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
| | - Guilherme Portugal
- Cardiology Department, Santa Marta Hospital, Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
| | - Sónia Oliveira
- Oncology Department, Santo António Capuchos Hospital, Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
| | - Rui Cruz Ferreira
- Cardiology Department, Santa Marta Hospital, Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
| |
Collapse
|
86
|
Lin K, Lengacher C. Anthracycline Chemotherapy–Induced Cardiotoxicity in Breast Cancer Survivors: A Systematic Review. Oncol Nurs Forum 2019; 46:E145-E158. [DOI: 10.1188/19.onf.e145-e158] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
87
|
Abstract
Cardiovascular magnetic resonance (CMR) imaging is useful to identify systolic dysfunction, particularly when echocardiographic imaging is not acceptable because of poor acoustic windows or when left ventricular ejection fraction (LVEF) is inconclusive by other modalities and an accurate LVEF measurement is needed. Of particular advantage in cardio-oncology is CMR's capability to perform tissue characterization to noninvasively identify changes in pathologic conditions related to cancer therapy or to discriminate causes of disease that may confound presentation in cardio-oncology patients. For these reasons, there is an increasing use of CMR in the screening and surveillance of cardio-oncology patients.
Collapse
Affiliation(s)
- Jennifer Hawthorne Jordan
- Department of Biomedical Engineering, Virginia Commonwealth University, Pauley Heart Center, Virginia Commonwealth University Health Sciences, 8-119B, 1200 East Broad Street, Richmond, VA 23298, USA.
| | - William Gregory Hundley
- Pauley Heart Center, Virginia Commonwealth University Health Sciences, 8-124, 1200 East Broad Street, Richmond, VA 23298, USA
| |
Collapse
|
88
|
Patient-reported health problems and healthcare use after treatment for early-stage breast cancer. Breast 2019; 46:4-11. [DOI: 10.1016/j.breast.2019.03.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 03/15/2019] [Accepted: 03/28/2019] [Indexed: 12/14/2022] Open
|
89
|
Emaus MJ, Išgum I, van Velzen SGM, van den Bongard HJGD, Gernaat SAM, Lessmann N, Sattler MGA, Teske AJ, Penninkhof J, Meijer H, Pignol JP, Verkooijen HM. Bragatston study protocol: a multicentre cohort study on automated quantification of cardiovascular calcifications on radiotherapy planning CT scans for cardiovascular risk prediction in patients with breast cancer. BMJ Open 2019; 9:e028752. [PMID: 31352417 PMCID: PMC6661654 DOI: 10.1136/bmjopen-2018-028752] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
INTRODUCTION Cardiovascular disease (CVD) is an important cause of death in breast cancer survivors. Some breast cancer treatments including anthracyclines, trastuzumab and radiotherapy can increase the risk of CVD, especially for patients with pre-existing CVD risk factors. Early identification of patients at increased CVD risk may allow switching to less cardiotoxic treatments, active surveillance or treatment of CVD risk factors. One of the strongest independent CVD risk factors is the presence and extent of coronary artery calcifications (CAC). In clinical practice, CAC are generally quantified on ECG-triggered cardiac CT scans. Patients with breast cancer treated with radiotherapy routinely undergo radiotherapy planning CT scans of the chest, and those scans could provide the opportunity to routinely assess CAC before a potentially cardiotoxic treatment. The Bragatston study aims to investigate the association between calcifications in the coronary arteries, aorta and heart valves (hereinafter called 'cardiovascular calcifications') measured automatically on planning CT scans of patients with breast cancer and CVD risk. METHODS AND ANALYSIS In a first step, we will optimise and validate a deep learning algorithm for automated quantification of cardiovascular calcifications on planning CT scans of patients with breast cancer. Then, in a multicentre cohort study (University Medical Center Utrecht, Utrecht, Erasmus MC Cancer Institute, Rotterdam and Radboudumc, Nijmegen, The Netherlands), the association between cardiovascular calcifications measured on planning CT scans of patients with breast cancer (n≈16 000) and incident (non-)fatal CVD events will be evaluated. To assess the added predictive value of these calcifications over traditional CVD risk factors and treatment characteristics, a case-cohort analysis will be performed among all cohort members diagnosed with a CVD event during follow-up (n≈200) and a random sample of the baseline cohort (n≈600). ETHICS AND DISSEMINATION The Institutional Review Boards of the participating hospitals decided that the Medical Research Involving Human Subjects Act does not apply. Findings will be published in peer-reviewed journals and presented at conferences. TRIAL REGISTRATION NUMBER NCT03206333.
Collapse
Affiliation(s)
- Marleen J Emaus
- Imaging Division, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ivana Išgum
- Image Sciences Institute, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sanne G M van Velzen
- Image Sciences Institute, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Sofie A M Gernaat
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Nikolas Lessmann
- Image Sciences Institute, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Margriet G A Sattler
- Department of Radiation Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Arco J Teske
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Joan Penninkhof
- Department of Radiation Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Hanneke Meijer
- Department of Radiation Oncology, Radboudumc, Nijmegen, The Netherlands
| | - Jean-Philippe Pignol
- Department of Radiation Oncology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Helena M Verkooijen
- Imaging Division, University Medical Center Utrecht, Utrecht, The Netherlands
- Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
90
|
Cherata DA, Donoiu I, Diaconu R, Glodeanu A, Cârstea D, Militaru C, Istrătoaie O. Longitudinal strain analysis allows the identification of subclinical deterioration of right ventricular function in patients with cancer therapy-related left ventricular dysfunction. Discoveries (Craiova) 2019; 7:e94. [PMID: 32309612 PMCID: PMC7093070 DOI: 10.15190/d.2019.7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND This study was designed to assess right ventricular systolic function in cancer patients. METHODS AND RESULTS 68 consecutive patients receiving potentially cardiotoxic agents were followed for 6 months in a single-center, observational, cohort-study. Left ventricle and free-wall right ventricular longitudinal strain were analyzed prior and after 6 months of treatment, using a vendor-independent software, together with left ventricle ejection fraction, tricuspid annulus plane systolic excursion and right ventricular fractional area change. Cancer therapy-related cardiac dysfunction was defined as a left ventricle ejection fraction drop of >10% to <53%. Both left ventricle ejection fraction (59±7% vs. 55±8%, p<0.0001) and left ventricle longitudinal strain (-19.7±2.5% vs. -17.1±2.6%, p<0.0001) were reduced at follow up, along with free-wall right ventricular longitudinal strain (-24.9±4.5% vs. -21.6±4.9%, p<0.0001). Cancer therapy-related cardiac dysfunction was detected in 20 patients (29%). In 15 out of these 20 patients (75%), a concomitant relative reduction in free-wall right ventricular longitudinal strain magnitude by 17±7% was detected. Moreover, there was a significant correlation between left ventricle and free-wall right ventricular longitudinal strain at follow-up examinations (r=0.323, p<0.0001). A relative drop of right ventricular longitudinal strain >17% had a sensitivity of 55% and a specificity of 70% (AUC=0.75, 0.7-0.8, 95% CI) to identify patients with cancer treatment related cardiac dysfunction. Neither tricuspid annulus plane systolic excursion (24±5 vs. 23±4 mm, p=0.07), nor right ventricular fractional area change (45±8% vs. 44±7%, p=0.6) showed any significant change between examinations. CONCLUSIONS Longitudinal strain analysis allows the identification of subclinical right ventricular dysfunction appearing in the course of cancer treatment when conventional indices of right ventricular dysfunction function are unaffected.
Collapse
Affiliation(s)
| | - Ionuț Donoiu
- Department of Cardiology, University of Medicine and Pharmacy, Craiova, Romania
| | - Rodica Diaconu
- Department of Cardiology, University of Medicine and Pharmacy, Craiova, Romania
| | - Adina Glodeanu
- Department of Cardiology, "Filantropia" Municipal Hospital, Craiova, Romania.,Department of Medical Semiology, University of Medicine and Pharmacy, Craiova, Romania
| | - Doina Cârstea
- Department of Cardiology, "Filantropia" Municipal Hospital, Craiova, Romania.,Department of Medical Semiology, University of Medicine and Pharmacy, Craiova, Romania
| | - Constantin Militaru
- Department of Cardiology, University of Medicine and Pharmacy, Craiova, Romania
| | - Octavian Istrătoaie
- Department of Cardiology, University of Medicine and Pharmacy, Craiova, Romania
| |
Collapse
|
91
|
Tocchetti CG, Cadeddu C, Di Lisi D, Femminò S, Madonna R, Mele D, Monte I, Novo G, Penna C, Pepe A, Spallarossa P, Varricchi G, Zito C, Pagliaro P, Mercuro G. From Molecular Mechanisms to Clinical Management of Antineoplastic Drug-Induced Cardiovascular Toxicity: A Translational Overview. Antioxid Redox Signal 2019; 30:2110-2153. [PMID: 28398124 PMCID: PMC6529857 DOI: 10.1089/ars.2016.6930] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Significance: Antineoplastic therapies have significantly improved the prognosis of oncology patients. However, these treatments can bring to a higher incidence of side-effects, including the worrying cardiovascular toxicity (CTX). Recent Advances: Substantial evidence indicates multiple mechanisms of CTX, with redox mechanisms playing a key role. Recent data singled out mitochondria as key targets for antineoplastic drug-induced CTX; understanding the underlying mechanisms is, therefore, crucial for effective cardioprotection, without compromising the efficacy of anti-cancer treatments. Critical Issues: CTX can occur within a few days or many years after treatment. Type I CTX is associated with irreversible cardiac cell injury, and it is typically caused by anthracyclines and traditional chemotherapeutics. Type II CTX is generally caused by novel biologics and more targeted drugs, and it is associated with reversible myocardial dysfunction. Therefore, patients undergoing anti-cancer treatments should be closely monitored, and patients at risk of CTX should be identified before beginning treatment to reduce CTX-related morbidity. Future Directions: Genetic profiling of clinical risk factors and an integrated approach using molecular, imaging, and clinical data may allow the recognition of patients who are at a high risk of developing chemotherapy-related CTX, and it may suggest methodologies to limit damage in a wider range of patients. The involvement of redox mechanisms in cancer biology and anticancer treatments is a very active field of research. Further investigations will be necessary to uncover the hallmarks of cancer from a redox perspective and to develop more efficacious antineoplastic therapies that also spare the cardiovascular system.
Collapse
Affiliation(s)
| | - Christian Cadeddu
- 2 Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Daniela Di Lisi
- 3 Biomedical Department of Internal Medicine, University of Palermo, Palermo, Italy
| | - Saveria Femminò
- 4 Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Rosalinda Madonna
- 5 Center of Aging Sciences and Translational Medicine - CESI-MeT, "G. d'Annunzio" University, Chieti, Italy.,6 Department of Internal Medicine, The Texas Heart Institute and Center for Cardiovascular Biology and Atherosclerosis Research, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Donato Mele
- 7 Cardiology Unit, Emergency Department, University Hospital of Ferrara, Ferrara, Italy
| | - Ines Monte
- 8 Department of General Surgery and Medical-Surgery Specialities, University of Catania, Catania, Italy
| | - Giuseppina Novo
- 3 Biomedical Department of Internal Medicine, University of Palermo, Palermo, Italy
| | - Claudia Penna
- 4 Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Alessia Pepe
- 9 U.O.C. Magnetic Resonance Imaging, Fondazione Toscana G. Monasterio C.N.R., Pisa, Italy
| | - Paolo Spallarossa
- 10 Clinic of Cardiovascular Diseases, IRCCS San Martino IST, Genova, Italy
| | - Gilda Varricchi
- 1 Department of Translational Medical Sciences, Federico II University, Naples, Italy.,11 Center for Basic and Clinical Immunology Research (CISI) - Federico II University, Naples, Italy
| | - Concetta Zito
- 12 Division of Cardiology, Clinical and Experimental Department of Medicine and Pharmacology, Policlinico "G. Martino" University of Messina, Messina, Italy
| | - Pasquale Pagliaro
- 4 Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Giuseppe Mercuro
- 2 Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| |
Collapse
|
92
|
Abstract
Doxorubicin-induced cardiotoxicity in childhood cancer survivors is a growing problem. The population of patients at risk for cardiovascular disease is steadily increasing, as five-year survival rates for all types of childhood cancers continue to improve. Doxorubicin affects the developing heart differently from the adult heart and in a subset of exposed patients, childhood exposure leads to late, irreversible cardiomyopathy. Notably, the prevalence of late-onset toxicity is increasing in parallel with improved survival. By the year 2020, it is estimated that there will be 500,000 childhood cancer survivors and over 50,000 of them will suffer from doxorubicin-induced cardiotoxicity. The majority of the research to-date, concentrated on childhood cancer survivors, has focused mostly on clinical outcomes through well-designed epidemiological and retrospective cohort studies. Preclinical studies have elucidated many of the cellular mechanisms that elicit acute toxicity in cardiomyocytes. However, more research is needed in the areas of early- and late-onset cardiotoxicity and more importantly improving the scientific understanding of how other cells present in the cardiac milieu are impacted by doxorubicin exposure. The overall goal of this review is to succinctly summarize the major clinical and preclinical studies focused on doxorubicin-induced cardiotoxicity. As the prevalence of patients affected by doxorubicin exposure continues to increase, it is imperative that the major gaps in existing research are identified and subsequently utilized to develop appropriate research priorities for the coming years. Well-designed preclinical research models will enhance our understanding of the pathophysiology of doxorubicin-induced cardiotoxicity and directly lead to better diagnosis, treatment, and prevention. © 2019 American Physiological Society. Compr Physiol 9:905-931, 2019.
Collapse
Affiliation(s)
- Trevi R. Mancilla
- University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
| | - Brian Iskra
- University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
| | - Gregory J. Aune
- University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
| |
Collapse
|
93
|
Schumacher AN, Shackelford DYK, Brown JM, Hayward R. Validation of the 6-min Walk Test for Predicting Peak V˙O2 in Cancer Survivors. Med Sci Sports Exerc 2019; 51:271-277. [PMID: 30239495 DOI: 10.1249/mss.0000000000001790] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE To assess the quality of the relationship between V˙O2peak estimated from patient outcomes on the 6-min walk test (6MWT) and the V˙O2peak calculated from patient outcomes on the University of Northern Colorado Cancer Rehabilitation Institute (UNCCRI) treadmill protocol. METHODS Cancer survivors (N = 187) completed the UNCCRI treadmill protocol and a 6MWT 1 wk apart in randomized order to obtain V˙O2peak. Values from the UNCCRI treadmill protocol were compared against four common 6MWT V˙O2peak prediction equations. RESULTS All four 6MWT prediction equations significantly (P < 0.001) underestimated V˙O2peak with predicted values ranging from 8.0 ± 4.1 mL·kg·min to 18.6 ± 3.1 mL·kg·min, whereas the UNCCRI treadmill protocol yielded a significantly higher value of 23.9 ± 7.6 mL·kg·min. A positive strong correlation occurred between estimated V˙O2peak derived from the UNCCRI treadmill protocol and only one of the V˙O2peak values derived from the 6MWT prediction equations (r = 0.81), and all four equations consistently underpredicted V˙O2peak. CONCLUSIONS These findings suggest that the 6MWT is not a valid test for predicting V˙O2peak in the cancer population due to its consistent underestimation of V˙O2peak regardless of the prediction equation. Obtaining an accurate and valid V˙O2peak value is necessary to correctly prescribe an individualized exercise rehabilitation regimen for cancer survivors. It is recommended that clinicians avoid the 6MWT and instead implement treadmill testing to volitional fatigue to quantify V˙O2peak in cancer survivors.
Collapse
Affiliation(s)
| | | | | | - Reid Hayward
- School of Sport and Exercise Science, and the University of Northern Colorado Cancer Rehabilitation Institute, University of Northern Colorado, Greeley, CO
| |
Collapse
|
94
|
Abstract
The emergence of various targeted anticancer agents has led us to uncharted territory secondary to their cardiotoxic potential with many burning questions, which in turn has led to the evolution of the cardio-oncology field. These targeted agents differ in their cardiovascular complication (CVC) potential even within the same class and it is very difficult to design screening tests that can predict CVCs. Moreover, there is a need for more research to answer many crucial questions, since these toxicities are unanticipated and can lead to poor overall survival of cancer patients. We still do not clearly understand the mechanism for such toxicity, risk factors, and natural history. A better understanding of the underlying risk factors and identification of biomarkers would help us develop protocols for appropriate monitoring strategies which in turn would help capture these toxicities at early stages. In this succinct review, we try to focus on CVC definition, summarize some published research, and point to areas of unmet need in this new field.
Collapse
Affiliation(s)
- Chintan P Shah
- Department of Medicine, Hematology Oncology Division, University of Florida, 1600 SW Archer Road, Gainesville, 32608, FL, USA
| | - Jan S Moreb
- Department of Medicine, Hematology Oncology Division, University of Florida, Gainesville, FL, USA
| |
Collapse
|
95
|
Cuomo A, Rodolico A, Galdieri A, Russo M, Campi G, Franco R, Bruno D, Aran L, Carannante A, Attanasio U, Tocchetti CG, Varricchi G, Mercurio V. Heart Failure and Cancer: Mechanisms of Old and New Cardiotoxic Drugs in Cancer Patients. Card Fail Rev 2019; 5:112-118. [PMID: 31179022 PMCID: PMC6545979 DOI: 10.15420/cfr.2018.32.2] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 01/30/2019] [Indexed: 02/07/2023] Open
Abstract
Although there have been many improvements in prognosis for patients with cancer, anticancer therapies are burdened by the risk of cardiovascular toxicity. Heart failure is one of the most dramatic clinical expressions of cardiotoxicity, and it may occur acutely or appear years after treatment. This article reviews the main mechanisms and clinical presentations of left ventricular dysfunction induced by some old and new cardiotoxic drugs in cancer patients, referring to the most recent advances in the field. The authors describe the mechanisms of cardiotoxicity induced by anthracyclines, which can lead to cardiovascular problems in up to 48% of patients who take them. The authors also describe mechanisms of cardiotoxicity induced by biological drugs that produce left ventricular dysfunction through secondary mechanisms. They outline the recent advances in immunotherapies, which have revolutionised anticancer therapies.
Collapse
Affiliation(s)
- Alessandra Cuomo
- Department of Translational Medical Sciences, Federico II University Naples, Italy
| | - Alessio Rodolico
- Department of Translational Medical Sciences, Federico II University Naples, Italy
| | - Amalia Galdieri
- Department of Translational Medical Sciences, Federico II University Naples, Italy
| | - Michele Russo
- Department of Translational Medical Sciences, Federico II University Naples, Italy
| | - Giacomo Campi
- Department of Translational Medical Sciences, Federico II University Naples, Italy
| | - Riccardo Franco
- Department of Translational Medical Sciences, Federico II University Naples, Italy
| | - Dalila Bruno
- Department of Translational Medical Sciences, Federico II University Naples, Italy
| | - Luisa Aran
- Department of Translational Medical Sciences, Federico II University Naples, Italy
| | - Antonio Carannante
- Department of Translational Medical Sciences, Federico II University Naples, Italy
| | - Umberto Attanasio
- Department of Translational Medical Sciences, Federico II University Naples, Italy
| | - Carlo G Tocchetti
- Department of Translational Medical Sciences, Federico II University Naples, Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences, Federico II University Naples, Italy
| | - Valentina Mercurio
- Department of Translational Medical Sciences, Federico II University Naples, Italy
| |
Collapse
|
96
|
Jacobse JN, Duane FK, Boekel NB, Schaapveld M, Hauptmann M, Hooning MJ, Seynaeve CM, Baaijens MHA, Gietema JA, Darby SC, van Leeuwen FE, Aleman BMP, Taylor CW. Radiation Dose-Response for Risk of Myocardial Infarction in Breast Cancer Survivors. Int J Radiat Oncol Biol Phys 2019; 103:595-604. [PMID: 30385276 PMCID: PMC6361769 DOI: 10.1016/j.ijrobp.2018.10.025] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 10/07/2018] [Accepted: 10/19/2018] [Indexed: 12/17/2022]
Abstract
PURPOSE Previous reports suggest that radiation therapy for breast cancer (BC) can cause ischemic heart disease, with the radiation-related risk increasing linearly with mean whole heart dose (MWHD). This study aimed to validate these findings in younger BC patients and to investigate additional risk factors for radiation-related myocardial infarction (MI). METHODS AND MATERIALS A nested case-control study was conducted within a cohort of BC survivors treated during 1970 to 2009. Cases were 183 patients with MI as their first heart disease after BC. One control per case was selected and matched on age and BC diagnosis date. Information on treatment and cardiovascular risk factors was abstracted from medical and radiation charts. Cardiac doses were estimated for each woman by reconstructing her regimen using modern 3-dimensional computed tomography planning on a typical patient computed tomography scan. RESULTS Median age at BC of cases and controls was 50.2 years (interquartile range, 45.7-54.7). Median time to MI was 13.6 years (interquartile range, 9.9-18.1). Median MWHD was 8.9 Gy (range, 0.3-35.2 Gy). MI rate increased linearly with increasing MWHD (excess rate ratio [ERR] per Gy, 6.4%; 95% confidence interval, 1.3%-16.0%). Patients receiving ≥20 Gy MWHD had a 3.4-fold (95% confidence interval, 1.5-7.6) higher MI rate than unirradiated patients. ERRs were higher for younger women, with borderline significance (ERR<45years, 24.2%/Gy; ERR≥50years, 2.5%/Gy; Pinteraction = .054). Whole heart dose-volume parameters did not modify the dose-response relationship significantly. CONCLUSIONS MI rate after radiation for BC increases linearly with MWHD. Reductions in MWHD are expected to contribute to better cardiovascular health of BC survivors.
Collapse
Affiliation(s)
- Judy N Jacobse
- Department of Psychosocial Research and Epidemiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Frances K Duane
- Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Naomi B Boekel
- Department of Psychosocial Research and Epidemiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Michael Schaapveld
- Department of Psychosocial Research and Epidemiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Michael Hauptmann
- Department of Biostatistics, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Maartje J Hooning
- Department of Medical Oncology, Erasmus MC, Cancer Institute, Rotterdam, The Netherlands
| | - Caroline M Seynaeve
- Department of Medical Oncology, Erasmus MC, Cancer Institute, Rotterdam, The Netherlands
| | - Margreet H A Baaijens
- Department of Radiation Oncology, Erasmus MC, Cancer Institute, Rotterdam, The Netherlands
| | - Jourik A Gietema
- Department of Medical Oncology, University Medical Center Groningen, Groningen, The Netherlands
| | - Sarah C Darby
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Flora E van Leeuwen
- Department of Psychosocial Research and Epidemiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Berthe M P Aleman
- Department of Radiation Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - Carolyn W Taylor
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
97
|
|
98
|
Attenuation of doxorubicin-induced cardiotoxicity in a human in vitro cardiac model by the induction of the NRF-2 pathway. Biomed Pharmacother 2019; 112:108637. [PMID: 30798127 DOI: 10.1016/j.biopha.2019.108637] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 01/28/2019] [Accepted: 01/28/2019] [Indexed: 01/06/2023] Open
Abstract
Dose-dependent cardiotoxicity is the leading adverse reaction seen in cancer patients treated with doxorubicin. Currently, dexrazoxane is the only approved drug that can partially protect against this toxicity in patients, however, its administration is restricted to those patients receiving a high cumulative dose of anthracyclines. Investigations into the mechanisms of cardiotoxicity and efforts to improve cardioprotective strategies have been hindered by the limited availability of a phenotypically relevant in vitro adult human cardiac model system. Here, we adapted a readily reproducible, functional 3D human multi-cell type cardiac system to emulate patient responses seen with doxorubicin and dexrazoxane. We show that administration of two NRF2 gene inducers namely the semi-synthetic triterpenoid Bardoxolone methyl, and the isothiocyanate sulfurophane, result in cardioprotection against doxorubicin toxicity comparable to dexrazoxane as evidenced by an increase in cell viability and a decrease in the production of reactive oxygen species. We further show a synergistic attenuation of cardiotoxicity when the NRF2 inducers and dexrazoxane are used in tandem. Taken together, our data indicate that the 3D spheroid is a suitable model to investigate drug induced cardiotoxicity and we reveal an essential role of the NRF2 pathway in cardioprotection providing a novel pharmacological mechanism and intervention route towards the alleviation of doxorubicin-induced toxicity.
Collapse
|
99
|
Massey SC, Hawkins-Daarud A, Gallaher J, Anderson ARA, Canoll P, Swanson KR. Lesion Dynamics Under Varying Paracrine PDGF Signaling in Brain Tissue. Bull Math Biol 2019; 81:1645-1664. [PMID: 30796683 DOI: 10.1007/s11538-019-00587-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 02/12/2019] [Indexed: 01/08/2023]
Abstract
Paracrine PDGF signaling is involved in many processes in the body, both normal and pathological, including embryonic development, angiogenesis, and wound healing as well as liver fibrosis, atherosclerosis, and cancers. We explored this seemingly dual (normal and pathological) role of PDGF mathematically by modeling the release of PDGF in brain tissue and then varying the dynamics of this release. Resulting simulations show that by varying the dynamics of a PDGF source, our model predicts three possible outcomes for PDGF-driven cellular recruitment and lesion growth: (1) localized, short duration of growth, (2) localized, chronic growth, and (3) widespread chronic growth. Further, our model predicts that the type of response is much more sensitive to the duration of PDGF exposure than the maximum level of that exposure. This suggests that extended duration of paracrine PDGF signal during otherwise normal processes could potentially lead to lesions having a phenotype consistent with pathologic conditions.
Collapse
Affiliation(s)
- Susan Christine Massey
- Precision Neurotherapeutics Innovation Program, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ, 85054, USA.
| | - Andrea Hawkins-Daarud
- Precision Neurotherapeutics Innovation Program, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ, 85054, USA
| | - Jill Gallaher
- Integrative Mathematical Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | | | - Peter Canoll
- Division of Neuropathology, Department of Pathology and Cell Biology, Columbia University School of Medicine, New York, NY, USA
| | - Kristin R Swanson
- Precision Neurotherapeutics Innovation Program, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ, 85054, USA
| |
Collapse
|
100
|
El-Said NT, Mohamed EA, Taha RA. Irbesartan suppresses cardiac toxicity induced by doxorubicin via regulating the p38-MAPK/NF-κB and TGF-β1 pathways. Naunyn Schmiedebergs Arch Pharmacol 2019; 392:647-658. [DOI: 10.1007/s00210-019-01624-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 01/28/2019] [Indexed: 12/19/2022]
|