51
|
Liu Y, Tang LL, Liang C, Wu MM, Zhang ZR. Insulin Resistance and Pellino-1 Mediated Decrease in the Activities of Vasodilator Signaling Contributes to Sunitinib-Induced Hypertension. Front Pharmacol 2021; 12:617165. [PMID: 33841146 PMCID: PMC8027079 DOI: 10.3389/fphar.2021.617165] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 01/21/2021] [Indexed: 01/10/2023] Open
Abstract
Antiangiogenic tyrosine kinases inhibitors induce hypertension, which may increase the incidents of cardiovascular complications and limit their use. However, the mechanisms by which usage of TKIs results in hypertension have not been fully understood. Here, we report the potential mechanisms of how sunitinib, a widely used TKI, induces hypertension. Male SD rats were randomly divided into control group and sunitinib-administrated group. We show that sunitinib administration for seven days caused a significant increase in artery blood pressure, along with glycerolipid metabolism abnormalities including decreased food intake and low body weight, hypoglycemia, hyperinsulinemia. Sunitinib administration also resulted in a significant increase in the levels of insulin autoantibody (IAA), cyclic adenosine monophosphate and free fatty acid in serum; whereas, sunitinib administration had no effects on serum glucagon levels. Sunitinib led to the decreased insulin sensitivity as determined by insulin tolerance test (ITT) and glucose tolerance test (GTT), reflecting insulin resistance occurred in sunitinib-treated rats. The results obtained from wire myograph assay in the mesenteric arteries show that endothelium-dependent relaxation, but not endothelium-independent relaxation, was impaired by sunitinib. Furthermore, western blot analysis revealed that the expressions levels of phosphorylated IRS-1, Pellino-1, AKT and eNOS were significantly attenuated by sunitinib in rat mesenteric artery tissues and in the sunitinib-treated primary cultured mesenteric artery endothelial cells. The levels of serum and endothelium-derived nitric oxide were also significantly decreased by sunitinib. Moreover, sunitinib-induced decrease in the expression levels of phosphorylated AKT and eNOS was further reduced by knocking down of Pellino-1 in MAECs. Our results suggest that sunitinib causes vascular dysfunction and hypertension, which are associated with insulin resistance- and Pellino-1-mediated inhibition of AKT/eNOS/NO signaling. Our results may provide a rational for preventing and/or treating sunitinib-induced endothelial dysfunction and hypertension.
Collapse
Affiliation(s)
- Yang Liu
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang key laboratory for Metabolic disorder and cancer related cardiovascular diseases, and Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Liang-Liang Tang
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang key laboratory for Metabolic disorder and cancer related cardiovascular diseases, and Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Chen Liang
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang key laboratory for Metabolic disorder and cancer related cardiovascular diseases, and Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Ming-Ming Wu
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang key laboratory for Metabolic disorder and cancer related cardiovascular diseases, and Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Zhi-Ren Zhang
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang key laboratory for Metabolic disorder and cancer related cardiovascular diseases, and Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China.,NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China
| |
Collapse
|
52
|
Moslehi J, Lichtman AH, Sharpe AH, Galluzzi L, Kitsis RN. Immune checkpoint inhibitor-associated myocarditis: manifestations and mechanisms. J Clin Invest 2021; 131:145186. [PMID: 33645548 DOI: 10.1172/jci145186] [Citation(s) in RCA: 98] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have transformed the treatment of various cancers, including malignancies once considered untreatable. These agents, however, are associated with inflammation and tissue damage in multiple organs. Myocarditis has emerged as a serious ICI-associated toxicity, because, while seemingly infrequent, it is often fulminant and lethal. The underlying basis of ICI-associated myocarditis is not completely understood. While the importance of T cells is clear, the inciting antigens, why they are recognized, and the mechanisms leading to cardiac cell injury remain poorly characterized. These issues underscore the need for basic and clinical studies to define pathogenesis, identify predictive biomarkers, improve diagnostic strategies, and develop effective treatments. An improved understanding of ICI-associated myocarditis will provide insights into the equilibrium between the immune and cardiovascular systems.
Collapse
Affiliation(s)
- Javid Moslehi
- Division of Cardiovascular Medicine and Division of Oncology, Cardio-Oncology Program, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Andrew H Lichtman
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Arlene H Sharpe
- Department of Immunology and Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA.,Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Sandra and Edward Meyer Cancer Center, Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medical College, New York, New York, USA.,Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA.,Université de Paris, Paris, France
| | - Richard N Kitsis
- Departments of Medicine and Cell Biology, Wilf Family Cardiovascular Research Institute, and Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
53
|
Skubalova Z, Rex S, Sukupova M, Zahalka M, Skladal P, Pribyl J, Michalkova H, Weerasekera A, Adam V, Heger Z. Passive Diffusion vs Active pH-Dependent Encapsulation of Tyrosine Kinase Inhibitors Vandetanib and Lenvatinib into Folate-Targeted Ferritin Delivery System. Int J Nanomedicine 2021; 16:1-14. [PMID: 33442247 PMCID: PMC7797358 DOI: 10.2147/ijn.s275808] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 11/07/2020] [Indexed: 12/12/2022] Open
Abstract
Introduction The present study reports on examination of the effects of encapsulating the tyrosine kinase inhibitors (TKIs) vandetanib and lenvatinib into a biomacromolecular ferritin-based delivery system. Methods The encapsulation of TKIs was performed via two strategies: i) using an active reversible pH-dependent reassembly of ferritin´s quaternary structure and ii) passive loading of hydrophobic TKIs through the hydrophobic channels at the junctions of ferritin subunits. After encapsulation, ferritins were surface-functionalized with folic acid promoting active-targeting capabilities. Results The physico-chemical and nanomechanical analyses revealed that despite the comparable encapsulation efficiencies of both protocols, the active loading affects stability and rigidity of ferritins, plausibly due to their imperfect reassembly. Biological experiments with hormone-responsive breast cancer cells (T47-D and MCF-7) confirmed the cytotoxicity of encapsulated and folate-targeted TKIs to folate-receptor positive cancer cells, but only limited cytotoxic effects to healthy breast epithelium. Importantly, the long-term cytotoxic experiments revealed that compared to the pH-dependent encapsulation, the passively-loaded TKIs exert markedly higher anticancer activity, most likely due to undesired influence of harsh acidic environment used for the pH-dependent encapsulation on the TKIs’ structural and functional properties. Conclusion Since the passive loading does not require a reassembly step for which acids are needed, the presented investigation serves as a solid basis for future studies focused on encapsulation of small hydrophobic molecules.
Collapse
Affiliation(s)
- Zuzana Skubalova
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czech Republic.,Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Simona Rex
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czech Republic.,Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Martina Sukupova
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czech Republic
| | - Martin Zahalka
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czech Republic
| | - Petr Skladal
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Jan Pribyl
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Hana Michalkova
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czech Republic.,Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Akila Weerasekera
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czech Republic.,Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Zbynek Heger
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czech Republic.,Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| |
Collapse
|
54
|
Gonciar D, Mocan L, Zlibut A, Mocan T, Agoston-Coldea L. Cardiotoxicity in HER2-positive breast cancer patients. Heart Fail Rev 2021; 26:919-935. [PMID: 33405000 DOI: 10.1007/s10741-020-10072-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/21/2020] [Indexed: 01/22/2023]
Abstract
Due to the recent advances in diagnosis and management of patients with HER2-positive breast cancer, especially through novel HER2-targeted agents, cardiotoxicity becomes an emerging problem. Although chemotherapy significantly increases survival, the risk of cardiovascular disease development is high and still underestimated and could imply treatment discontinuation. Frequently, due to lack of rigorous diagnosis strategies, cardiotoxicity assessment is delayed, and, moreover, the efficacy of current therapy options in restoring heart function is questionable. For a comprehensive risk assessment, it is vital to characterize the clinical spectrum of HER2-targeted agents and anthracyclines, as well as their pathogenic pathways involved in cardiotoxicity. Advanced cardiovascular multimodal imaging and circulating biomarkers plays primary roles in early assessing cardiotoxicity and also in guiding specific preventive measures. Even though the knowledge in this field is rapidly expanding, there are still questions that arise regarding the optimal approach in terms of timing and methods. The aim of the current review aims to providean overview of currently available data.
Collapse
Affiliation(s)
- Diana Gonciar
- 2nd Department of Internal Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Lucian Mocan
- 3rd Surgery Department, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Nanomedicine, Regional Institute of Gastroenterology and Hepatology, Cluj-Napoca, Romania
| | - Alexandru Zlibut
- 2nd Department of Internal Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Teodora Mocan
- Physiology Department, Iuliu Hațieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Nanomedicine, Regional Institute of Gastroenterology and Hepatology, Cluj-Napoca, Romania
| | - Lucia Agoston-Coldea
- 2nd Department of Internal Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.
| |
Collapse
|
55
|
Iacopo F, Branch M, Cardinale D, Middeldorp M, Sanders P, Cohen JB, Achirica MC, Jaiswal S, Brown SA. Preventive Cardio-Oncology: Cardiovascular Disease Prevention in Cancer Patients and Survivors. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2021. [DOI: 10.1007/s11936-020-00883-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
56
|
Vascular Endothelial Growth Factor (VEGF) Inhibitor Cardiotoxicity: What Should We Know? CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2020. [DOI: 10.1007/s11936-020-00866-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
57
|
Dent SF, Kikuchi R, Kondapalli L, Ismail-Khan R, Brezden-Masley C, Barac A, Fradley M. Optimizing Cardiovascular Health in Patients With Cancer: A Practical Review of Risk Assessment, Monitoring, and Prevention of Cancer Treatment-Related Cardiovascular Toxicity. Am Soc Clin Oncol Educ Book 2020; 40:1-15. [PMID: 32213102 DOI: 10.1200/edbk_286019] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Advances in cancer screening and improved treatment approaches have led to an increase in survivorship and, consequently, recognition of an association between cancer treatments and the development of cardiovascular complications. In addition, as the population becomes proportionally older, comorbid cardiovascular risk factors are more prevalent in the population and compound the risk of developing cancer treatment-related cardiovascular toxicity. Cardio-oncology has emerged as a new subspecialty of medicine that provides a multidisciplinary approach, bringing together oncologists, cardiologists, and allied health care providers who are tasked with optimizing the cardiovascular health of patients exposed to potentially cardiotoxic cancer therapy. Using a case-based approach, practical advice on how to identify, monitor, and treat patients with cancer who are at risk for developing cancer treatment-related cardiovascular dysfunction is discussed. Cardiovascular risk factors (e.g., age, hypertension, diabetes) and cancer therapies (chemotherapy, targeted therapy, radiation) associated with cardiovascular toxicity are presented. Current cardiac monitoring strategies such as two- and three-dimensional echocardiography, cardiac MRI, and biomarkers (troponin and brain natriuretic peptide [BNP]) are discussed. Last, the current literature on pharmacologic (e.g., angiotensin-converting enzyme inhibitors, β-blockers, statins) and lifestyle (diet and exercise) strategies to mitigate cardiovascular toxicity during and following completion of cancer therapy are reviewed.
Collapse
Affiliation(s)
- Susan F Dent
- Division of Medical Oncology, Duke Cancer Institute, Duke University, Durham, NC
| | - Robin Kikuchi
- Division of Medical Oncology, Duke Cancer Institute, Duke University, Durham, NC
| | - Lavanya Kondapalli
- University of Colorado Health Cancer, University of Colorado, Aurora, CO
| | | | | | - Ana Barac
- MedStar Heart and Vascular Institute, Georgetown University, Washington, DC
| | - Michael Fradley
- Moffitt Cancer Center, University of South Florida, Tampa, FL
| |
Collapse
|
58
|
Bottinor W, Hundley WG. Cancer and Cardiovascular Disease: Finding the Signal Through the Noise. JACC: CARDIOONCOLOGY 2020; 2:578-580. [PMID: 34396269 PMCID: PMC8352326 DOI: 10.1016/j.jaccao.2020.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Wendy Bottinor
- Department of Internal Medicine, Division of Cardiovascular Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - W Gregory Hundley
- Department of Internal Medicine, Division of Cardiovascular Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
59
|
Alexandre J, Cautela J, Ederhy S, Damaj GL, Salem JE, Barlesi F, Farnault L, Charbonnier A, Mirabel M, Champiat S, Cohen-Solal A, Cohen A, Dolladille C, Thuny F. Cardiovascular Toxicity Related to Cancer Treatment: A Pragmatic Approach to the American and European Cardio-Oncology Guidelines. J Am Heart Assoc 2020; 9:e018403. [PMID: 32893704 PMCID: PMC7727003 DOI: 10.1161/jaha.120.018403] [Citation(s) in RCA: 139] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The considerable progress made in the field of cancer treatment has led to a dramatic improvement in the prognosis of patients with cancer. However, toxicities resulting from these treatments represent a cost that can be harmful to short- and long-term outcomes. Adverse events affecting the cardiovascular system are one of the greatest challenges in the overall management of patients with cancer, as they can compromise the success of the optimal treatment against the tumor. Such adverse events are associated not only with older chemotherapy drugs such as anthracyclines but also with many targeted therapies and immunotherapies. Recognizing this concern, several American and European governing societies in oncology and cardiology have published guidelines on the cardiovascular monitoring of patients receiving potentially cardiotoxic cancer therapies, as well as on the management of cardiovascular toxicities. However, the low level of evidence supporting these guidelines has led to numerous discrepancies, leaving clinicians without a consensus strategy to apply. A cardio-oncology expert panel from the French Working Group of Cardio-Oncology has undertaken an ambitious effort to analyze and harmonize the most recent American and European guidelines to propose roadmaps and decision algorithms that would be easy for clinicians to use in their daily practice. In this statement, the experts addressed the cardiovascular monitoring strategies for the cancer drugs associated with the highest risk of cardiovascular toxicities, as well as the management of such toxicities.
Collapse
Affiliation(s)
- Joachim Alexandre
- PICARO Cardio-Oncology Program Department of Pharmacology Department of Cardiology Caen Hospital Medical School Caen-Normandy University Caen France
| | - Jennifer Cautela
- Unit of Heart Failure and Valvular Heart Diseases Department of Cardiology Nord Hospital Center for CardioVascular and Nutrition Research (C2VN) University Mediterranean Center of Cardio-Oncology (MEDI-CO Center) Assistance Publique - Hôpitaux de MarseilleAix-Marseille University Marseille France.,Mediterranean Group of Cardio-Oncology (gMEDICO) Marseille France
| | - Stéphane Ederhy
- UNICO-GRECO Cardio-Oncology Program Department of Cardiology Saint-Antoine Hospital Tenon Hospital Inserm 856 Assistance Publique - Hôpitaux de ParisSorbonne University Paris France
| | - Ghandi Laurent Damaj
- Department of Hematology Caen Hospital Medical School Caen-Normandy University Caen France
| | - Joe-Elie Salem
- UNICO-GRECO Cardio-Oncology Program Department of Pharmacology Centre d'Investigation Clinique Paris-Est Pitié-Salpêtrière Hospital Assistance Publique - Hôpitaux de ParisSorbonne University Paris France
| | - Fabrice Barlesi
- Drug Development Department (DITEP) Gustave RoussyParis-Saclay University Villejuif France
| | - Laure Farnault
- Departement of Hematology Conception HospitalAssistance Publique - Hôpitaux de MarseilleAix-Marseille University Marseille France
| | - Aude Charbonnier
- Departement of Hematology Paoli-Calmettes Cancer InstituteAix-Marseille University Marseille France
| | - Mariana Mirabel
- Unit of Cardio-Oncology and Prevention European Georges Pompidou HospitalAssistance Publique - Hôpitaux de ParisSorbonne University Paris France
| | - Stéphane Champiat
- Drug Development Department (DITEP) Gustave RoussyParis-Saclay University Villejuif France
| | - Alain Cohen-Solal
- Department of Cardiology Lariboisière Hospital UMR-S 942 Assistance Publique - Hôpitaux de ParisParis University Paris France
| | - Ariel Cohen
- UNICO-GRECO Cardio-Oncology Program Department of Cardiology Saint-Antoine Hospital Tenon Hospital Inserm 856 Assistance Publique - Hôpitaux de ParisSorbonne University Paris France
| | - Charles Dolladille
- PICARO Cardio-Oncology Program Department of Pharmacology Department of Cardiology Caen Hospital Medical School Caen-Normandy University Caen France
| | - Franck Thuny
- Unit of Heart Failure and Valvular Heart Diseases Department of Cardiology Nord Hospital Center for CardioVascular and Nutrition Research (C2VN) University Mediterranean Center of Cardio-Oncology (MEDI-CO Center) Assistance Publique - Hôpitaux de MarseilleAix-Marseille University Marseille France.,Mediterranean Group of Cardio-Oncology (gMEDICO) Marseille France
| |
Collapse
|
60
|
Bottinor W, Parikh A, Jahangir E. Emerging cancer therapies and cardiovascular risk. J Thromb Thrombolysis 2020; 51:837-845. [PMID: 32886244 DOI: 10.1007/s11239-020-02263-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The cardiovascular (CV) toxicity profiles of traditional cancer therapies such as anthracyclines and radiation therapy are familiar to many cardiologists. With the development and widespread use of additional cancer therapeutics, CV toxicities related to these agents are becoming more common. Cardiovascular specialists are often integrated into the care team for individuals with cancer and knowledge of the CV toxicities of cancer therapeutics has become essential. In this review, we provide a clinically focused summary of the current data regarding CV toxicities of common cancer therapies and identify potential management strategies for the CV specialist.
Collapse
Affiliation(s)
- Wendy Bottinor
- Division of Cardiovascular Medicine, Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Gateway bldg. 1200 E Marshall St, Richmond, VA, 23298, USA. .,Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA.
| | - Amar Parikh
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Eiman Jahangir
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
61
|
Miller JM, Meki MH, Ou Q, George SA, Gams A, Abouleisa RRE, Tang XL, Ahern BM, Giridharan GA, El-Baz A, Hill BG, Satin J, Conklin DJ, Moslehi J, Bolli R, Ribeiro AJS, Efimov IR, Mohamed TMA. Heart slice culture system reliably demonstrates clinical drug-related cardiotoxicity. Toxicol Appl Pharmacol 2020; 406:115213. [PMID: 32877659 DOI: 10.1016/j.taap.2020.115213] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/20/2020] [Accepted: 08/22/2020] [Indexed: 02/07/2023]
Abstract
The limited availability of human heart tissue and its complex cell composition are major limiting factors for the reliable testing of drug efficacy and toxicity. Recently, we developed functional human and pig heart slice biomimetic culture systems that preserve the viability and functionality of 300 μm heart slices for up to 6 days. Here, we tested the reliability of this culture system for testing the cardiotoxicity of anti-cancer drugs. We tested three anti-cancer drugs (doxorubicin, trastuzumab, and sunitinib) with known different mechanisms of cardiotoxicity at three concentrations and assessed the effect of these drugs on heart slice viability, structure, function and gene expression. Slices incubated with any of these drugs for 48 h showed diminished in viability as well as loss of cardiomyocyte structure and function. Mechanistically, RNA sequencing of doxorubicin-treated tissues demonstrated a significant downregulation of cardiac genes and upregulation of oxidative stress responses. Trastuzumab treatment downregulated cardiac muscle contraction-related genes consistent with its clinically known effect on cardiomyocytes. Interestingly, sunitinib treatment resulted in significant downregulation of angiogenesis-related genes, in line with its mechanism of action. Similar to hiPS-derived-cardiomyocytes, heart slices recapitulated the expected toxicity of doxorubicin and trastuzumab, however, slices were superior in detecting sunitinib cardiotoxicity and mechanism in the clinically relevant concentration range of 0.1-1 μM. These results indicate that heart slice culture models have the potential to become a reliable platform for testing and elucidating mechanisms of drug cardiotoxicity.
Collapse
Affiliation(s)
- Jessica M Miller
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, USA; Department of Bioengineering, University of Louisville, KY, USA
| | - Moustafa H Meki
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, USA; Department of Bioengineering, University of Louisville, KY, USA
| | - Qinghui Ou
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, USA
| | - Sharon A George
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA
| | - Anna Gams
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA
| | - Riham R E Abouleisa
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, USA
| | - Xian-Liang Tang
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, USA
| | - Brooke M Ahern
- Department of Physiology, University of Kentucky, KY, USA
| | | | - Ayman El-Baz
- Department of Bioengineering, University of Louisville, KY, USA
| | - Bradford G Hill
- Envirome Institute, Diabetes and Obesity Center, Department of Medicine, University of Louisville, KY, USA
| | - Jonathan Satin
- Department of Physiology, University of Kentucky, KY, USA
| | - Daniel J Conklin
- Envirome Institute, Diabetes and Obesity Center, Department of Medicine, University of Louisville, KY, USA
| | - Javid Moslehi
- Division of Cardiology, Cardio-Oncology Program, Vanderbilt University Medical Center, 2220 Pierce Avenue, Nashville, USA
| | - Roberto Bolli
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, USA
| | - Alexandre J S Ribeiro
- U.S. Food and Drug Administration, Center for Drug Evaluation and Research, Office of Translational Science, Office of Clinical Pharmacology, Division of Applied Regulatory Science, Silver Spring, MD, USA.
| | - Igor R Efimov
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA.
| | - Tamer M A Mohamed
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, USA; Department of Bioengineering, University of Louisville, KY, USA; Envirome Institute, Diabetes and Obesity Center, Department of Medicine, University of Louisville, KY, USA; Department of Pharmacology and Toxicology, University of Louisville, KY, USA; Institute of Cardiovascular Sciences, University of Manchester, UK; Faculty of Pharmacy, Zagazig University, Egypt.
| |
Collapse
|
62
|
Van Leeuwen MT, Luu S, Gurney H, Brown MR, Pearson SA, Webber K, Hunt L, Hong S, Delaney GP, Vajdic CM. Cardiovascular Toxicity of Targeted Therapies for Cancer: An Overview of Systematic Reviews. JNCI Cancer Spectr 2020; 4:pkaa076. [PMID: 33392444 PMCID: PMC7768929 DOI: 10.1093/jncics/pkaa076] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 07/01/2020] [Accepted: 08/18/2020] [Indexed: 12/16/2022] Open
Abstract
Background Several targeted therapies for cancer have been associated with cardiovascular toxicity. The evidence for this association has not been synthesized systematically nor has the quality of evidence been considered. We synthesized systematic review evidence of cardiovascular toxicity of individual targeted agents. Methods We searched MEDLINE, Embase, and the Cochrane Database of Systematic Reviews for systematic reviews with meta-analyses of cardiovascular outcomes for individual agents published to May 2020. We selected reviews according to prespecified eligibility criteria (International Prospective Register of Systematic Reviews CRD42017080014). We classified evidence of cardiovascular toxicity as sufficient, probable, possible, or indeterminate for specific cardiovascular outcomes based on statistical significance, study quality, and size. Results From 113 systematic reviews, we found at least probable systematic review evidence of cardiovascular toxicity for 18 agents, including high- and all-grade hypertension for bevacizumab, ramucirumab, axitinib, cediranib, pazopanib, sorafenib, sunitinib, vandetanib, aflibercept, abiraterone, and enzalutamide, and all-grade hypertension for nintedanib; high- and all-grade arterial thromboembolism (includes cardiac and/or cerebral events) for bevacizumab and abiraterone, high-grade arterial thromboembolism for trastuzumab, and all-grade arterial thromboembolism for sorafenib and tamoxifen; high- and all-grade venous thromboembolism (VTE) for lenalidomide and thalidomide, high-grade VTE for cetuximab and panitumumab, and all-grade VTE for bevacizumab; high- and all-grade left ventricular ejection fraction decline or congestive heart failure for bevacizumab and trastuzumab, and all-grade left ventricular ejection fraction decline/congestive heart failure for pazopanib and sunitinib; and all-grade corrected QT interval prolongation for vandetanib. Conclusions Our review provides an accessible summary of the cardiovascular toxicity of targeted therapy to assist clinicians and patients when managing cardiovascular health.
Collapse
Affiliation(s)
- Marina T Van Leeuwen
- Centre for Big Data Research in Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Steven Luu
- Centre for Big Data Research in Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Howard Gurney
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Martin R Brown
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Sallie-Anne Pearson
- Centre for Big Data Research in Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Kate Webber
- Department of Oncology, Monash Health, Clayton, Victoria, Australia.,School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Lee Hunt
- Cancer Voices NSW, Milsons Point, New South Wales, Australia
| | - Soojung Hong
- Centre for Big Data Research in Health, University of New South Wales, Sydney, New South Wales, Australia.,Division of Oncology-Haematology, Department of Internal Medicine, National Health Insurance Service Ilsan Hospital, Goyang, Republic of Korea
| | - Geoffrey P Delaney
- Liverpool Cancer Therapy Centre, Liverpool, New South Wales, Australia.,Collaboration for Cancer Outcomes Research and Evaluation, Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia.,South Western Sydney Clinical School, University of New South Wales, Liverpool, New South Wales, Australia
| | - Claire M Vajdic
- Centre for Big Data Research in Health, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
63
|
Waliany S, Lee D, Witteles RM, Neal JW, Nguyen P, Davis MM, Salem JE, Wu SM, Moslehi JJ, Zhu H. Immune Checkpoint Inhibitor Cardiotoxicity: Understanding Basic Mechanisms and Clinical Characteristics and Finding a Cure. Annu Rev Pharmacol Toxicol 2020; 61:113-134. [PMID: 32776859 DOI: 10.1146/annurev-pharmtox-010919-023451] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Immune checkpoint inhibitors (ICIs) attenuate mechanisms of self-tolerance in the immune system, enabling T cell responses to cancerous tissues and revolutionizing care for cancer patients. However, by loweringbarriers against self-reactivity, ICIs often result in varying degrees of autoimmunity. Cardiovascular complications, particularly myocarditis but also arrhythmias, pericarditis, and vasculitis, have emerged as significant complications associated with ICIs. In this review, we examine the clinical aspects and basic science principles that underlie ICI-associated myocarditis and other cardiovascular toxicities. In addition, we discuss current therapeutic approaches. We believe a better mechanistic understanding of ICI-associated toxicities can lead to improved patient outcomes by reducing treatment-related morbidity.
Collapse
Affiliation(s)
- Sarah Waliany
- Department of Medicine, Stanford University, Stanford, California 94305, USA;
| | - Daniel Lee
- Stanford Cardiovascular Institute, Stanford University, Stanford, California 94305, USA
| | - Ronald M Witteles
- Department of Medicine, Stanford University, Stanford, California 94305, USA; .,Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Joel W Neal
- Department of Medicine, Stanford University, Stanford, California 94305, USA; .,Division of Oncology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Patricia Nguyen
- Department of Medicine, Stanford University, Stanford, California 94305, USA; .,Stanford Cardiovascular Institute, Stanford University, Stanford, California 94305, USA.,Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Mark M Davis
- Department of Microbiology and Immunology and Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California 94305, USA.,Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Joe-Elie Salem
- Sorbonne Université, INSERM, CIC-1901 Paris-Est, CLIP² Galilée, UNICO-GRECO Cardio-Oncology Program, and Department of Pharmacology, Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris, F-75013 Paris, France.,Cardio-Oncology Program, Vanderbilt University Medical Center, Nashville, Tennessee 37203, USA; .,Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37203, USA
| | - Sean M Wu
- Department of Medicine, Stanford University, Stanford, California 94305, USA; .,Stanford Cardiovascular Institute, Stanford University, Stanford, California 94305, USA.,Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Javid J Moslehi
- Cardio-Oncology Program, Vanderbilt University Medical Center, Nashville, Tennessee 37203, USA; .,Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37203, USA
| | - Han Zhu
- Department of Medicine, Stanford University, Stanford, California 94305, USA; .,Stanford Cardiovascular Institute, Stanford University, Stanford, California 94305, USA.,Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| |
Collapse
|
64
|
Denlinger CS, Sanft T, Moslehi JJ, Overholser L, Armenian S, Baker KS, Broderick G, Demark-Wahnefried W, Friedman DL, Goldman M, Henry NL, Hill-Kayser C, Hudson M, Khakpour N, Koura D, McDonough AL, Melisko M, Mooney K, Moore HCF, Moryl N, O'Connor T, Paskett ED, Patel C, Peterson L, Pirl W, Rodriguez MA, Ruddy KJ, Shockney L, Smith S, Syrjala KL, Tevaarwerk A, Zee P, McMillian NR, Freedman-Cass DA. NCCN Guidelines Insights: Survivorship, Version 2.2020. J Natl Compr Canc Netw 2020; 18:1016-1023. [PMID: 32755975 DOI: 10.6004/jnccn.2020.0037] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The NCCN Guidelines for Survivorship provide screening, evaluation, and treatment recommendations for consequences of adult-onset cancer and its treatment, with the goal of helping healthcare professionals who work with survivors, including those in primary care. The guidelines also provide recommendations to help clinicians promote physical activity, weight management, and proper immunizations in survivors and facilitate care coordination to ensure that all of the survivors' needs are addressed. These NCCN Guidelines Insights summarize additions and changes made to the guidelines in 2020 regarding cardiovascular disease risk assessment and screening for subsequent primary malignancies.
Collapse
Affiliation(s)
| | - Tara Sanft
- Yale Cancer Center/Smilow Cancer Hospital
| | | | | | | | - K Scott Baker
- Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | | | | | | | - Mindy Goldman
- UCSF Helen Diller Family Comprehensive Cancer Center
| | | | | | - Melissa Hudson
- St. Jude Children's Research Hospital/The University of Tennessee Health Science Center
| | | | | | | | | | - Kathi Mooney
- Huntsman Cancer Institute at the University of Utah
| | - Halle C F Moore
- Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | | | | | - Electra D Paskett
- The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute
| | | | - Lindsay Peterson
- Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | | | | | | | - Lillie Shockney
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | | | - Karen L Syrjala
- Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | | | - Phyllis Zee
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University; and
| | | | | |
Collapse
|
65
|
Sadler D, Chaulagain C, Alvarado B, Cubeddu R, Stone E, Samuel T, Bastos B, Grossman D, Fu CL, Alley E, Nagarajan A, Nguyen T, Ahmed W, Elson L, Nahleh Z. Practical and cost-effective model to build and sustain a cardio-oncology program. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2020; 6:9. [PMID: 32690995 PMCID: PMC7363993 DOI: 10.1186/s40959-020-00063-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/03/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Cardio-Oncology (CO) is a new subspecialty that thrives mostly in large academic quaternary centers. This study describes how to establish a successful cardio-oncology program, with limited resources, in order to effectively manage the unique care required by this patient population. METHODS Clinical data was collected from 25 consecutive months. There were four foundational elements to establish a CO program: 1. Clinical program: integrating staff and resources from the Heart and Vascular, and Cancer Centers; 2. Education Program: establishing a platform to educate/advocate with respect to CO; 3. Engagement with professional societies: active engagement allowed for the successful establishment of the proposed CO program; and 4. Research program: establishing data collection modalities/cooperation with other institutions. RESULTS 474 consecutive patients were treated by our CO program during the first 25 months of operation. Clinical data, information about cancer treatment, cardiovascular co morbidities, cardiac testing and impact of CO management are reported. CONCLUSIONS A successful CO program can be established utilizing existing resources without the need for significant additional assets. Integration with professional societies, advocacy, education and research, provide a platform for learning and growth. This model improves access to care and can be reproduced in a variety of settings.
Collapse
Affiliation(s)
- Diego Sadler
- Cleveland Clinic Florida, Heart and Vascular Center, 2950 Cleveland Clinic Blvd, Weston, FL 33331 USA
| | - Chakra Chaulagain
- Cleveland Clinic Florida, Maroone Cancer Center, 2950 Cleveland Clinic Blvd, Weston, FL 33331 USA
| | - Beatrice Alvarado
- Cleveland Clinic Florida, Heart and Vascular Center, 2950 Cleveland Clinic Blvd, Weston, FL 33331 USA
| | - Robert Cubeddu
- Cleveland Clinic Florida, Heart and Vascular Center, 2950 Cleveland Clinic Blvd, Weston, FL 33331 USA
| | - Elizabeth Stone
- Cleveland Clinic Florida, Maroone Cancer Center, 2950 Cleveland Clinic Blvd, Weston, FL 33331 USA
| | - Thomas Samuel
- Cleveland Clinic Florida, Maroone Cancer Center, 2950 Cleveland Clinic Blvd, Weston, FL 33331 USA
| | - Bruno Bastos
- Cleveland Clinic Florida, Maroone Cancer Center, 2950 Cleveland Clinic Blvd, Weston, FL 33331 USA
| | - David Grossman
- Cleveland Clinic Florida, Maroone Cancer Center, 2950 Cleveland Clinic Blvd, Weston, FL 33331 USA
| | - Chieh-Lin Fu
- Cleveland Clinic Florida, Maroone Cancer Center, 2950 Cleveland Clinic Blvd, Weston, FL 33331 USA
| | - Evan Alley
- Cleveland Clinic Florida, Maroone Cancer Center, 2950 Cleveland Clinic Blvd, Weston, FL 33331 USA
| | - Arun Nagarajan
- Cleveland Clinic Florida, Maroone Cancer Center, 2950 Cleveland Clinic Blvd, Weston, FL 33331 USA
| | - Timmy Nguyen
- Cleveland Clinic Florida, Maroone Cancer Center, 2950 Cleveland Clinic Blvd, Weston, FL 33331 USA
| | - Wesam Ahmed
- Cleveland Clinic Florida, Maroone Cancer Center, 2950 Cleveland Clinic Blvd, Weston, FL 33331 USA
| | - Leah Elson
- Cleveland Clinic Florida, Maroone Cancer Center, 2950 Cleveland Clinic Blvd, Weston, FL 33331 USA
| | - Zeina Nahleh
- Cleveland Clinic Florida, Maroone Cancer Center, 2950 Cleveland Clinic Blvd, Weston, FL 33331 USA
| |
Collapse
|
66
|
Jin Y, Xu Z, Yan H, He Q, Yang X, Luo P. A Comprehensive Review of Clinical Cardiotoxicity Incidence of FDA-Approved Small-Molecule Kinase Inhibitors. Front Pharmacol 2020; 11:891. [PMID: 32595510 PMCID: PMC7303342 DOI: 10.3389/fphar.2020.00891] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 05/29/2020] [Indexed: 11/24/2022] Open
Abstract
Numerous protein kinases encoded in the genome have become attractive targets for the treatment of different types of cancer. As of January 2020, a total of 52 small-molecule kinase inhibitors (SMKIs) have been approved by the FDA. With the numerous clinical trials and a heavy focus on drug safety, SMKI-induced cardiotoxicity, which is a life-threatening risk, has greatly attracted the attention of researchers. In this review, the SMKIs with cardiotoxicity incidence were described exhaustively. The data were collected from 42 clinical trials, 25 FDA-published documents, seven meta-analysis/systematic reviews, three case reports and more than 50 other types of articles. To date, 73% (38 of 52) of SMKIs have reported treatment-related cardiotoxicity. Among the 38 SMKIs with known cardiotoxicity, the rates of incidence of cardiac adverse events were QT prolongation: 47% (18 of 38), hypertension: 40% (15 of 38), left ventricular dysfunction: 34% (13 of 38), arrhythmia: 34% (13 of 38), heart failure: 26% (10 of 38) and ischemia or myocardial infarction: 29% (11 of 38). In the development process of novel SMKIs, more attention should be paid to balancing the treatment efficacy and the risk of cardiotoxicity. In preclinical drug studies, producing an accurate and reliable cardiotoxicity evaluation model is of key importance. To avoid the clinical potential cardiotoxicity risk and discontinuation of a highly effective drug, patients treated with SMKIs should be proactively monitored on the basis of a global standard. Moreover, the underlying mechanisms of SMKI-induced cardiotoxicity need to be further studied to develop new therapies for SMKI-induced cardiotoxicity.
Collapse
Affiliation(s)
| | | | | | | | - Xiaochun Yang
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Peihua Luo
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
67
|
Moslehi J, Fujiwara K, Guzik T. Cardio-oncology: a novel platform for basic and translational cardiovascular investigation driven by clinical need. Cardiovasc Res 2020; 115:819-823. [PMID: 30888396 DOI: 10.1093/cvr/cvz048] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Javid Moslehi
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.,Division of Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.,Cardio-Oncology Program, Department of Medicine, Vanderbilt University Medical Center, 2220 Pierce Avenue, Nashville, TN, USA
| | - Keigi Fujiwara
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tomasz Guzik
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK.,Department of Internal and Agricultural Medicine, Jagiellonian University Collegium Medicum, Krakow, Poland
| |
Collapse
|
68
|
Alvarez-Cardona J, Mitchell J, Lenihan D. Vascular Toxicity in Patients with Cancer: Is There a Recipe to Clarify Treatment? CME. Methodist Debakey Cardiovasc J 2020; 15:289-299. [PMID: 31988690 DOI: 10.14797/mdcj-15-4-289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The acknowledgement of cardiovascular disease as one of the leading causes of mortality and morbidity among cancer survivors is the cornerstone of the growing field of cardio-oncology. Although standardizing treatment for any given disease is often considered ideal, it is important to recognize the value of pursuing a practical and personalized approach when caring for an oncology patient to minimize the risk of treatment-related cardiotoxicity. We hereby discuss a series of cases that illustrate the ways vascular toxicity can manifest in patients with cancer and, when appropriate, provide scientific evidence that supports clinical decision making. We also raise questions about the complex management of these patients while shedding light on future research in this growing field.
Collapse
|
69
|
Zito C, Manganaro R, Carerj S, Antonini-Canterin F, Benedetto F. Peripheral Artery Disease and Stroke. J Cardiovasc Echogr 2020; 30:S17-S25. [PMID: 32566462 PMCID: PMC7293872 DOI: 10.4103/jcecho.jcecho_4_19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/23/2019] [Accepted: 10/03/2019] [Indexed: 12/21/2022] Open
Abstract
Peripheral artery disease (PAD) and stroke can occur as vascular complication of anticancer treatment. Although the mechanisms, monitoring, and management of cardiotoxicities have received broad attention, vascular toxicities remain often underrecognized. In addition, the development of new chemotherapeutic drugs bears the risk of vasotoxicities that are yet to be identified and may not be realized with short-term follow-up periods. The propensity to develop PAD and/or stroke reflects the complex interplay between patient's baseline risk and preexisting vascular disease, particularly hypertension and diabetes, while evidence for genetic predisposition is increasing. Chemotherapeutic agents with a prominent vascular side effect profile have been identified. Interruption of vascular endothelial growth factor (VEGF) inhibitors (VEGFIs) signaling (i.e., bevacizumab) is associated with vascular toxicity and clinical sequelae such as hypertension, stroke, and thromboembolism beyond acute coronary syndromes. Cisplatin and 5-fluorouracil are the main drugs involved in the stroke risk. In addition, circulating concentrations of VEGF are reduced by cyclophosphamide administered at continuous low doses, which might underpin some of the observed vascular toxicity, such as stroke, as seen in patients treated with VEGF inhibitors. The risk of stroke is also increased after treatment with anthracyclines that can induce endothelial dysfunction and increase arterial stiffness. Proteasome inhibitors ( bortezomib and carfilzomib) and immunomodulatory agents (thalidomide, lenalidomide, and pomalidomide), approved for use in multiple myeloma, carry a black box warning for an increased risk of stroke. Finally, head-and-neck radiotherapy is associated with a doubled risk of cerebrovascular ischemic event, especially if exposure occurs in childhood. The mechanisms involved in radiation vasculopathy are represented by endothelial dysfunction, medial necrosis, fibrosis, and accelerated atherosclerosis. However, BCR-ABL tyrosine kinase inhibitor (TKI), used for the treatment of chronic myeloid leukemia (CML), is the main antineoplastic drugs involved in the development of PAD. In particular, second- and third-generation TKIs, such as nilotinib and ponatinib, while emerging as a potent arm in contrasting CML, are associated with a higher risk of PAD development rather than traditional imatinib. Factors favoring vascular complication are the presence of traditional cardiovascular risk factors (CVRF) and predisposing genetic factors, high doses of BCR-ABL TKIs, longer time of drug exposure, and sequential use of potent TKIs. Therefore, accurate cardiovascular risk stratification is strongly recommended in patient candidate to anticancer treatment associated with higher risk of vascular complication, in order to reduce the incidence of PAD and stroke through CVRF correction and selection of appropriate tailored patient strategy of treatment. Then, a clinical follow-up, eventually associated with instrumental evaluation through vascular ultrasound, should be performed.
Collapse
Affiliation(s)
- Concetta Zito
- Department of Clinical and Experimental Medicine, Cardiology Unit, University of Messina, Azienda Ospedaliera Universitaria "Policlinico G. Martino", Messina, Italy
| | - Roberta Manganaro
- Department of Clinical and Experimental Medicine, Cardiology Unit, University of Messina, Azienda Ospedaliera Universitaria "Policlinico G. Martino", Messina, Italy
| | - Scipione Carerj
- Department of Clinical and Experimental Medicine, Cardiology Unit, University of Messina, Azienda Ospedaliera Universitaria "Policlinico G. Martino", Messina, Italy
| | | | - Frank Benedetto
- Cardiology Department, Hospital 'Bianchi Melacrino Morelli' Reggio Calabria, Italy
| |
Collapse
|
70
|
Tanriverdi O, Ates S, Sandal KK, Uylas S, Bosna IC, Alkan A. Left ventricular dysfunction associated with axitinib and nivolumab experience in an advanced renal cell carcinoma. J Oncol Pharm Pract 2020; 26:1765-1768. [DOI: 10.1177/1078155220909422] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Introduction Tyrosine kinase inhibitors and immune checkpoint inhibitors are widely used in advanced renal cell carcinoma. Here we reported a left ventricular dysfunction associated with axitinib and nivolumab experience in this patient with heart failure. Case report A 70-year-old male patient with advanced renal cell carcinoma was treated with interferon alpha-2b 10 million U thrice weekly. After progression, sunitinib provided 18 months of stable disease. In third line, the patient was treated with axitinib 10 mg daily. Under axitinib, the patient presented with dyspnea and palpitations. The diagnostic work-up showed a left ventricular dysfunction with an ejection fraction (EF) of 35% in echocardiography. He was treated with diuretics, acetylsalicylic acid 100 mg and low molecular weight heparin. Management and outcome: After excluding cardiac ischemic and pulmonary pathologies, we concluded a possible adverse event diagnosis of axitinib-related cardiotoxicity. After close follow up for cardiac dysfunction, the patient was treated with nivolumab 3 mg/kg every two weeks. The initial EF was 32%. After three months therapy, the patient was asymptomatic for cardiac dysfunction and EF was 50%. CT scan showed partial response in pulmonary lesions. Discussion We have limited no data about cardiotoxicity associated axitinib and limited data about ICIPs. Our case is unique by providing data about how to manage a metastatic RCC patient with left ventricular dysfunction under axitinib and how to follow-up the cardiac functions while under nivolumab therapy.
Collapse
Affiliation(s)
- Ozgur Tanriverdi
- Division of Medical Oncology, Department of Internal Medicine, School of Medicine, Mugla Sitki Kocman University, Mugla, Turkey
| | - Sedef Ates
- School of Medicine, Mugla Sitki Kocman University, Mugla, Turkey
| | - Kerim K Sandal
- School of Medicine, Mugla Sitki Kocman University, Mugla, Turkey
| | - Selcuk Uylas
- School of Medicine, Mugla Sitki Kocman University, Mugla, Turkey
| | - Islam C Bosna
- Department of Internal Medicine, School of Medicine, Mugla Sitki Kocman University, Muğla, Turkey
| | - Ali Alkan
- Division of Medical Oncology, Department of Internal Medicine, School of Medicine, Mugla Sitki Kocman University, Mugla, Turkey
| |
Collapse
|
71
|
Hayek SS, Ganatra S, Lenneman C, Scherrer-Crosbie M, Leja M, Lenihan DJ, Yang E, Ryan TD, Liu J, Carver J, Mousavi N, O'Quinn R, Arnold A, Banchs J, Barac A, Ky B. Preparing the Cardiovascular Workforce to Care for Oncology Patients: JACC Review Topic of the Week. J Am Coll Cardiol 2020; 73:2226-2235. [PMID: 31047011 DOI: 10.1016/j.jacc.2019.02.041] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 02/04/2019] [Indexed: 10/26/2022]
Abstract
Cardiovascular disease and cancer are the 2 main causes of death in the United States. They intersect on multiple levels, sharing common causal mechanisms and epidemiological risk factors. The growing prevalence and complexity of cardiovascular disease and cancer have resulted in the development of the discipline of cardio-oncology. Preparing the cardiovascular workforce for the care of a growing population of cancer patients is necessary to enhance the delivery of high-quality cardiovascular care for patients with cancer. The goal of this review is to present the dedicated efforts of the cardio-oncology community to meet the growing need for education and training of cardiovascular practitioners providing care to cancer patients and survivors. Integration in general cardiology training programs and the efforts of the stakeholder organizations serve as an example of how a multidimensional, innovative approach can address provider education and training needs in a relatively new discipline.
Collapse
Affiliation(s)
- Salim S Hayek
- Division of Cardiology, University of Michigan, Ann Arbor, Michigan
| | - Sarju Ganatra
- Lahey Hospital and Medical Center, Burlington, Massachusetts
| | - Carrie Lenneman
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama
| | | | - Monika Leja
- Division of Cardiology, University of Michigan, Ann Arbor, Michigan
| | - Daniel J Lenihan
- Division of Cardiology, Cardio-Oncology Center of Excellence, Washington University in St. Louis, St. Louis, Missouri
| | - Eric Yang
- Division of Cardiology, University of California at Los Angeles, Los Angeles, California
| | - Thomas D Ryan
- Department of Pediatrics, University College of Medicine and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Jennifer Liu
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical Center, New York, New York
| | - Joseph Carver
- Division of Cardiology, Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Negareh Mousavi
- Division of Cardiology, McGill University, Montreal, Quebec, Canada
| | - Rupal O'Quinn
- Division of Cardiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Anita Arnold
- Division of Cardiology, Florida State University, Tallahassee, Florida
| | - Jose Banchs
- Division of Cardiology, MD Anderson Cancer Center, Houston, Texas
| | - Ana Barac
- MedStar Heart and Vascular Institute, Washington, DC.
| | - Bonnie Ky
- Department of Epidemiology, Biostatistics and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
72
|
Mechanisms of Cardiovascular Toxicity of BCR-ABL1 Tyrosine Kinase Inhibitors in Chronic Myelogenous Leukemia. Curr Hematol Malig Rep 2020; 15:20-30. [DOI: 10.1007/s11899-020-00560-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
73
|
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: 543] [Impact Index Per Article: 135.8] [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
|
74
|
Campia U, Moslehi JJ, Amiri-Kordestani L, Barac A, Beckman JA, Chism DD, Cohen P, Groarke JD, Herrmann J, Reilly CM, Weintraub NL. Cardio-Oncology: Vascular and Metabolic Perspectives: A Scientific Statement From the American Heart Association. Circulation 2019; 139:e579-e602. [PMID: 30786722 DOI: 10.1161/cir.0000000000000641] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cardio-oncology has organically developed as a new discipline within cardiovascular medicine as a result of the cardiac and vascular adverse sequelae of the major advances in cancer treatment. Patients with cancer and cancer survivors are at increased risk of vascular disease for a number of reasons. First, many new cancer therapies, including several targeted therapies, are associated with vascular and metabolic complications. Second, cancer itself serves as a risk factor for vascular disease, especially by increasing the risk for thromboembolic events. Finally, recent data suggest that common modifiable and genetic risk factors predispose to both malignancies and cardiovascular disease. Vascular complications in patients with cancer represent a new challenge for the clinician and a new frontier for research and investigation. Indeed, vascular sequelae of novel targeted therapies may provide insights into vascular signaling in humans. Clinically, emerging challenges are best addressed by a multidisciplinary approach in which cardiovascular medicine specialists and vascular biologists work closely with oncologists in the care of patients with cancer and cancer survivors. This novel approach realizes the goal of providing superior care through the creation of cardio-oncology consultative services and the training of a new generation of cardiovascular specialists with a broad understanding of cancer treatments.
Collapse
|
75
|
Lamore SD, Kohnken RA, Peters MF, Kolaja KL. Cardiovascular Toxicity Induced by Kinase Inhibitors: Mechanisms and Preclinical Approaches. Chem Res Toxicol 2019; 33:125-136. [DOI: 10.1021/acs.chemrestox.9b00387] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Sarah D. Lamore
- Preclinical Development, Wave Life Sciences, Lexington, Massachusetts 02421, United States
| | - Rebecca A. Kohnken
- Preclinical Safety, Abbvie, North Chicago, Illinois 60064, United States
| | - Matthew F. Peters
- Oncology Safety, Clinical Pharmacology and Safety Sciences, AstraZeneca Pharmaceuticals, Waltham, Massachusetts 02451, United States
| | - Kyle L. Kolaja
- Investigative Toxicology and Cell Therapy Safety, Nonclinical Development, Celgene Corporation, Summit, New Jersey 07901, United States
| |
Collapse
|
76
|
Babaev VR, Ding L, Zhang Y, May JM, Ramsey SA, Vickers KC, Linton MF. Loss of 2 Akt (Protein Kinase B) Isoforms in Hematopoietic Cells Diminished Monocyte and Macrophage Survival and Reduces Atherosclerosis in Ldl Receptor-Null Mice. Arterioscler Thromb Vasc Biol 2019; 39:156-169. [PMID: 30567482 DOI: 10.1161/atvbaha.118.312206] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Objective- Macrophages express 3 Akt (protein kinase B) isoforms, Akt1, Akt2, and Akt3, which display isoform-specific functions but may be redundant in terms of Akt survival signaling. We hypothesize that loss of 2 Akt isoforms in macrophages will suppress their ability to survive and modulate the development of atherosclerosis. Approach and Results- To test this hypothesis, we reconstituted male Ldlr-/- mice with double Akt2/Akt3 knockout hematopoietic cells expressing only the Akt1 isoform (Akt1only). There were no differences in body weight and plasma lipid levels between the groups after 8 weeks of the Western diet; however, Akt1only→ Ldlr-/- mice developed smaller (57.6% reduction) atherosclerotic lesions with more apoptotic macrophages than control mice transplanted with WT (wild type) cells. Next, male and female Ldlr-/- mice were reconstituted with double Akt1/Akt2 knockout hematopoietic cells expressing the Akt3 isoform (Akt3only). Female and male Akt3only→ Ldlr-/- recipients had significantly smaller (61% and 41%, respectively) lesions than the control WT→ Ldlr-/- mice. Loss of 2 Akt isoforms in hematopoietic cells resulted in markedly diminished levels of white blood cells, B cells, and monocytes and compromised viability of monocytes and peritoneal macrophages compared with WT cells. In response to lipopolysaccharides, macrophages with a single Akt isoform expressed low levels of inflammatory cytokines; however, Akt1only macrophages were distinct in expressing high levels of antiapoptotic Il10 compared with WT and Akt3only cells. Conclusions- Loss of 2 Akt isoforms in hematopoietic cells, preserving only a single Akt1 or Akt3 isoform, markedly compromises monocyte and macrophage viability and diminishes early atherosclerosis in Ldlr-/- mice.
Collapse
Affiliation(s)
- Vladimir R Babaev
- From the Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Department of Medicine (V.R.B., L.D., Y.Z., J.M.M., K.C.V., M.F.L.), Vanderbilt University School of Medicine, Nashville, TN
| | - Lei Ding
- From the Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Department of Medicine (V.R.B., L.D., Y.Z., J.M.M., K.C.V., M.F.L.), Vanderbilt University School of Medicine, Nashville, TN
| | - Youmin Zhang
- From the Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Department of Medicine (V.R.B., L.D., Y.Z., J.M.M., K.C.V., M.F.L.), Vanderbilt University School of Medicine, Nashville, TN
| | - James M May
- From the Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Department of Medicine (V.R.B., L.D., Y.Z., J.M.M., K.C.V., M.F.L.), Vanderbilt University School of Medicine, Nashville, TN.,Department of Molecular Physiology and Biophysics (J.M.M., K.C.V.), Vanderbilt University School of Medicine, Nashville, TN
| | - Stephen A Ramsey
- Department of Biomedical Sciences, Oregon State University, School of Electrical Engineering and Computer Science, Corvallis (S.A.R.)
| | - Kasey C Vickers
- From the Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Department of Medicine (V.R.B., L.D., Y.Z., J.M.M., K.C.V., M.F.L.), Vanderbilt University School of Medicine, Nashville, TN.,Department of Molecular Physiology and Biophysics (J.M.M., K.C.V.), Vanderbilt University School of Medicine, Nashville, TN
| | - MacRae F Linton
- From the Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Department of Medicine (V.R.B., L.D., Y.Z., J.M.M., K.C.V., M.F.L.), Vanderbilt University School of Medicine, Nashville, TN.,Department of Pharmacology (M.F.L.), Vanderbilt University School of Medicine, Nashville, TN
| |
Collapse
|
77
|
Cohen JB, Geara AS, Hogan JJ, Townsend RR. Hypertension in Cancer Patients and Survivors: Epidemiology, Diagnosis, and Management. JACC CardioOncol 2019; 1:238-251. [PMID: 32206762 PMCID: PMC7089580 DOI: 10.1016/j.jaccao.2019.11.009] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 11/01/2019] [Accepted: 11/04/2019] [Indexed: 12/28/2022] Open
Abstract
Cancer patients and survivors of cancer have a greater burden of cardiovascular disease compared to the general population. Much of the elevated cardiovascular risk in these individuals is likely attributable to hypertension, as individuals with cancer have a particularly high incidence of hypertension following cancer diagnosis. Treatment with chemotherapy is an independent risk factor for hypertension due to direct effects of many agents on endothelial function, sympathetic activity, and renin-angiotensin system activity as well as nephrotoxicity. Diagnosis and management of hypertension in cancer patients requires accurate blood pressure measurement and consideration of potential confounding factors, such as adjuvant treatments and acute pain, that can temporarily elevate blood pressure readings. Home blood pressure monitoring can be a useful tool to facilitate longitudinal blood pressure monitoring for titration of antihypertensive medications. Selection of antihypertensive agents in cancer patients should account for treatment-specific morbidities and target organ injury.
Collapse
Affiliation(s)
- Jordana B. Cohen
- Renal-Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Abdallah S. Geara
- Renal-Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jonathan J. Hogan
- Renal-Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Raymond R. Townsend
- Renal-Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
78
|
Ala CK, Klein AL, Moslehi JJ. Cancer Treatment-Associated Pericardial Disease: Epidemiology, Clinical Presentation, Diagnosis, and Management. Curr Cardiol Rep 2019; 21:156. [PMID: 31768769 DOI: 10.1007/s11886-019-1225-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Cancer therapeutics have seen tremendous growth in the last decade and have been effective in the treatment of several cancer types. However, with advanced therapies like kinase inhibitors and immunotherapies, there have been unintended consequences of cardiotoxicities. While traditional chemotherapy and radiation-induced cardiotoxicity have been well studied, further research is needed to understand the adverse effects of newer regimens. RECENT FINDINGS Both immune-mediated and non-immune-medicated cytotoxicity have been noted with targeted therapies such as tyrosine kinase inhibitors and immune checkpoint inhibitors. In this manuscript, we describe the pericardial syndromes associated with cancer therapies and propose management strategies. Pericardial effusion and pericarditis are common presentations in cancer patients and often difficult to diagnose. Concomitant myocarditis may also present with pericardial toxicity, especially with immunotherapies. In addition to proper history and physical, additional testing such as cardiovascular imaging and tissue histology need to be obtained as appropriate. Holding the offending oncology drug, and institution of anti-inflammatory medications, and immunosuppressants such as steroids are indicated. A high index of suspicion, use of standardized definitions, and comprehensive evaluation are needed for early identification, appropriate treatment, and better outcomes for patients with cancer treatment-associated pericardial disease. Further research is needed to understand the pathophysiology and to evaluate how the management of pericardial conditions in these patients differ from traditional management and also evaluate new therapies.
Collapse
Affiliation(s)
- Chandra K Ala
- Department of Cardiology, Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH, USA.,Department of Cardiology, Detroit Medical Center/Wayne State University, Detroit, MI, USA
| | - Allan L Klein
- Center for the Diagnosis and Treatment of Pericardial Diseases, Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Javid J Moslehi
- Division of Cardiovascular Medicine, Cardio-Oncology Program, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
79
|
Yeh ETH, Ewer MS, Moslehi J, Dlugosz-Danecka M, Banchs J, Chang HM, Minotti G. Mechanisms and clinical course of cardiovascular toxicity of cancer treatment I. Oncology. Semin Oncol 2019; 46:397-402. [PMID: 31753424 DOI: 10.1053/j.seminoncol.2019.10.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 10/09/2019] [Indexed: 12/13/2022]
Abstract
The opening session of Second International Colloquium on Cardio-Oncology addressed two areas of vital interest. The first reviewed new thoughts related to established agents. While anthracycline cardiotoxicity has been studied and reviewed extensively, ongoing research attempting to understand why it appears the mechanism(s) of toxicity differs from that of oncologic efficacy continue to evoke comment and intriguing speculation. Better understanding of the role of β-topoisomerase II in toxicity has advanced our understanding of the cascade of events that lead to heart failure. Additionally, the cardioprotective role of dexrazoxane fits well with our new understanding of how β-topoisomerase II works. Beyond the anthracyclines, new insight is providing us insight to better understand the impact on cardiac function seen with other agents including those targeting HER2 and several tyrosine-kinase inhibitors. Unlike the anthracyclines, these agents affect cardiac function in ways that are less direct, and therefore have different characteristics and should be thought of in alternate ways. This new knowledge regarding established agents furthers our understanding of the spectrum of cardiotoxicity and cardiac dysfunction in the cancer patient. The session also addressed cardiovascular toxicities of newer and established agents beyond myocardial dysfunction including effects on the vasculature. These agents cause changes that may be temporary or permanent, and that range from subclinical to life-threatening. The session ended with a discussion of the cardiac effects of immune checkpoint inhibitors. These agents can cause rare and sometimes fatal cardiac inflammation, for which long-term follow up may be required.
Collapse
Affiliation(s)
- Edward T H Yeh
- Center for Precision Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO
| | - Michael S Ewer
- The University of Texas, MD Anderson Cancer Center, Houston, TX
| | - Javid Moslehi
- Cardio-Oncology Program, Vanderbilt School of Medicine, Nashville, TN
| | | | - Jose Banchs
- The University of Texas, MD Anderson Cancer Center, Houston, TX
| | - Hui-Ming Chang
- Center for Precision Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO
| | - Giorgio Minotti
- Department of Medicine, Campus Bio-Medico University, Rome, Italy.
| |
Collapse
|
80
|
Denlinger CS, Sanft T, Baker KS, Broderick G, Demark-Wahnefried W, Friedman DL, Goldman M, Hudson M, Khakpour N, King A, Koura D, Lally RM, Langbaum TS, McDonough AL, Melisko M, Montoya JG, Mooney K, Moslehi JJ, O'Connor T, Overholser L, Paskett ED, Peppercorn J, Pirl W, Rodriguez MA, Ruddy KJ, Silverman P, Smith S, Syrjala KL, Tevaarwerk A, Urba SG, Wakabayashi MT, Zee P, McMillian NR, Freedman-Cass DA. Survivorship, Version 2.2018, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2019; 16:1216-1247. [PMID: 30323092 DOI: 10.6004/jnccn.2018.0078] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The NCCN Guidelines for Survivorship provide screening, evaluation, and treatment recommendations for common physical and psychosocial consequences of cancer and cancer treatment to help healthcare professionals who work with survivors of adult-onset cancer in the posttreatment period. This portion of the guidelines describes recommendations regarding the management of anthracycline-induced cardiotoxicity and lymphedema. In addition, recommendations regarding immunizations and the prevention of infections in cancer survivors are included.
Collapse
|
81
|
Waliany S, Sainani KL, Park LS, Zhang CA, Srinivas S, Witteles RM. Increase in Blood Pressure Associated With Tyrosine Kinase Inhibitors Targeting Vascular Endothelial Growth Factor. JACC: CARDIOONCOLOGY 2019; 1:24-36. [PMID: 34396159 PMCID: PMC8352203 DOI: 10.1016/j.jaccao.2019.08.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/30/2019] [Accepted: 08/15/2019] [Indexed: 02/07/2023]
Abstract
Objectives This study quantified the change in blood pressure (BP) during antivascular endothelial growth factor (VEGF) tyrosine kinase inhibitor (TKI) therapy, compared BPs between TKIs, and analyzed change in BP during antihypertensive therapy. Background TKIs targeting VEGF are associated with hypertension. The absolute change in BP during anti-VEGF TKI treatment is not well characterized outside clinical trials. Methods A retrospective single-center study included patients with metastatic renal cell carcinoma who received anti-VEGF TKIs between 2007 and 2018. Mixed models analyzed 3,088 BPs measured at oncology clinics. Results In 228 patients (baseline systolic blood pressure [SBP] 130.2 ± 16.3 mm Hg, diastolic blood pressure [DBP] 76.8 ± 9.3 mm Hg), anti-VEGF TKIs were associated with mean increases in SBP of 8.5 mm Hg (p < 0.0001) and DBP of 6.7 mm Hg (p <0.0001). Of the anti-VEGF TKIs evaluated, axitinib was associated with the greatest BP increase, with an increase in SBP of 12.6 mm Hg (p < 0.0001) and in DBP of 10.3 mm Hg (p < 0.0001) relative to baseline. In pairwise comparisons between agents, axitinib was associated with greater SBPs than cabozantinib by 8.4 mm Hg (p = 0.004) and pazopanib by 5.1 mm Hg (p = 0.01). Subsequent anti-VEGF TKI courses were associated with small increases in DBP, but not SBP, relative to the first course. During anti-VEGF TKI therapy, calcium-channel blockers and potassium-sparing diuretic agents were associated with the largest BP reductions, with decreases in SBP of 5.6 mm Hg (p < 0.0001) and 9.9 mm Hg (p = 0.007), respectively. Conclusions Anti-VEGF TKIs are associated with increased BP; greatest increases are observed with axitinib. Calcium-channel blockers and potassium-sparing diuretic agents were associated with the largest reductions in BP.
Collapse
Key Words
- ACE, angiotensin-converting enzyme
- ARB, angiotensin II receptor blocker
- BP, blood pressure
- CCB, calcium-channel blocker
- CTCAE, Common Terminology Criteria for Adverse Events
- DBP, diastolic blood pressure
- SBP, systolic blood pressure
- TKI, tyrosine kinase inhibitor
- VEGF, vascular endothelial growth factor
- antiangiogenic therapy
- antihypertensive agents
- blood pressure
- calcium-channel blockers
- diuretics
- eGFR, estimated glomerular filtration rate
- hypertension
- mRCC, metastatic renal cell carcinoma
- renal cell cancer
- treatment-related hypertension
- tyrosine kinase inhibitors
- vascular endothelial growth factor inhibitors
Collapse
Affiliation(s)
- Sarah Waliany
- Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Kristin L Sainani
- Department of Health Research and Policy, Stanford University School of Medicine, Stanford, California, USA
| | - Lesley S Park
- Center for Population Health Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Chiyuan Amy Zhang
- Department of Urology, Stanford University School of Medicine, Stanford, California, USA
| | - Sandy Srinivas
- Division of Medical Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Ronald M Witteles
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
82
|
Bottinor WJ, Shuey MM, Manouchehri A, Farber-Eger EH, Xu M, Nair D, Salem JE, Wang TJ, Brittain EL. Renin-Angiotensin-Aldosterone System Modulates Blood Pressure Response During Vascular Endothelial Growth Factor Receptor Inhibition. JACC: CARDIOONCOLOGY 2019; 1:14-23. [PMID: 32984850 PMCID: PMC7513950 DOI: 10.1016/j.jaccao.2019.07.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Objectives This study postulated that antihypertensive therapy with renin-angiotensin-aldosterone system (RAAS) inhibition may mitigate vascular endothelial growth factor inhibitor (VEGFi)–mediated increases in blood pressure more effectively than other antihypertensive medications in patients receiving VEGFi therapy. Background VEGFi therapy is commonly used in the treatment of cancer. One common side effect of VEGFi therapy is elevated blood pressure. Evidence suggests that the RAAS may be involved in VEGFi-mediated increases in blood pressure. Methods This retrospective cohort analysis was performed using a de-identified version of the electronic health record at Vanderbilt University Medical Center in Nashville, Tennessee. Subjects with cancer who were exposed to VEGFi therapy were identified, and blood pressure and medication data were extracted. Changes in mean systolic and diastolic blood pressure in response to VEGFi therapy in patients receiving RAAS inhibitor (RAASi) therapy before VEGFi initiation were compared with changes in mean systolic and diastolic blood pressure in patients not receiving RAASi therapy before VEGFi initiation. Results Mean systolic and diastolic blood pressure rose in both groups after VEGFi use; however, patients who had RAASi therapy before VEGFi initiation had a significantly lower increase in systolic blood pressure as compared with patients with no RAASi therapy (2.46 mm Hg [95% confidence interval: 0.7 to 4.2] compared with 4.56 mm Hg [95% confidence interval: 3.5 to 5.6], respectively; p = 0.034). Conclusions In a real-world clinical population, RAASi therapy before VEGFi initiation may ameliorate VEGFi-mediated increases in blood pressure. Randomized clinical trials are needed to further our understanding of the role of RAASi therapy in VEGFi-mediated increases in blood pressure.
Collapse
Affiliation(s)
- Wendy J Bottinor
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Megan M Shuey
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Ali Manouchehri
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Eric H Farber-Eger
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Meng Xu
- Department of Biostatistics, Vanderbilt University, Nashville, Tennessee
| | - Devika Nair
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Joe-Elie Salem
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee.,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee.,Department of Clinical Pharmacology, University of the Sorbonne, Assistance Publique Hôpitaux de Paris, Institut National de la Santé et de la Recherche Médicale CIC 14-21, Pitié-Salpêtrière Hospital, Paris, France
| | - Thomas J Wang
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Evan L Brittain
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
83
|
Seltzer JH, Gintant G, Amiri-Kordestani L, Singer J, Koplowitz LP, Moslehi JJ, Barac A, Yu AF. Assessing cardiac safety in oncology drug development. Am Heart J 2019; 214:125-133. [PMID: 31202099 PMCID: PMC7316329 DOI: 10.1016/j.ahj.2019.04.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 04/16/2019] [Indexed: 01/08/2023]
Affiliation(s)
| | | | | | - Jack Singer
- Washington State University, CTI Biopharma, Seattle WA
| | | | | | - Ana Barac
- Georgetown University/Medstar Health, Washington DC
| | | |
Collapse
|
84
|
Bonaca MP, Olenchock BA, Salem JE, Wiviott SD, Ederhy S, Cohen A, Stewart GC, Choueiri TK, Di Carli M, Allenbach Y, Kumbhani DJ, Heinzerling L, Amiri-Kordestani L, Lyon AR, Thavendiranathan P, Padera R, Lichtman A, Liu PP, Johnson DB, Moslehi J. Myocarditis in the Setting of Cancer Therapeutics: Proposed Case Definitions for Emerging Clinical Syndromes in Cardio-Oncology. Circulation 2019; 140:80-91. [PMID: 31390169 PMCID: PMC6779326 DOI: 10.1161/circulationaha.118.034497] [Citation(s) in RCA: 269] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Recent developments in cancer therapeutics have improved outcomes but have also been associated with cardiovascular complications. Therapies harnessing the immune system have been associated with an immune-mediated myocardial injury described as myocarditis. Immune checkpoint inhibitors are one such therapy with an increasing number of case and cohort reports describing a clinical syndrome of immune checkpoint inhibitor–associated myocarditis. Although the full spectrum of immune checkpoint inhibitor–associated cardiovascular disease still needs to be fully defined, described cases of myocarditis range from syndromes with mild signs and symptoms to fatal events. These observations in the clinical setting stand in contrast to outcomes from randomized clinical trials in which myocarditis is a rare event that is investigator reported and lacking in a specific case definition. The complexities associated with diagnosis, as well as the heterogeneous clinical presentation of immune checkpoint inhibitor–associated myocarditis, have made ascertainment and identification of myocarditis with high specificity challenging in clinical trials and other data sets, limiting the ability to better understand the incidence, outcomes, and predictors of these rare events. Therefore, establishing a uniform definition of myocarditis for application in clinical trials of cancer immunotherapies will enable greater understanding of these events. We propose an operational definition of cancer therapy-associated myocarditis that may facilitate case ascertainment and report and therefore may enhance the understanding of the incidence, outcomes, and risk factors of this novel clinical syndrome.
Collapse
Affiliation(s)
- Marc P Bonaca
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Benjamin A Olenchock
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Joe-Elie Salem
- Division of Cardiovascular Medicine, Clinical Pharmacology, Cardio-Oncology Program, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN.,Division of Oncology, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN.,UNICO APHP 6 Cardio-Oncology Program,Sorbonne Universite, INSERM Clinical Investigation Center Paris-Est Assistance Publique - Hopitaux de Paris, Pitié-Salpêtrière Hospital, Department of Pharmacology, Paris, France
| | - Stephen D Wiviott
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Stephane Ederhy
- UNICO APHP 6 Cardio-Oncology Program,Service de cardiologie Hôpitaux Universitaires Est Parisien, Hôpital Saint Antoine, Assistance Publique–Hôpitaux de Paris, INSERM 856, Sorbonne-université, France
| | - Ariel Cohen
- UNICO APHP 6 Cardio-Oncology Program,Sorbonne-Université and INSERM 856, Hôpital Saint Antoine, Paris, France
| | - Garrick C Stewart
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Toni K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Marcelo Di Carli
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Yves Allenbach
- Sorbonne University, AP-PH, Pitié Salpêtrière Hospital, Department of Internal Medicine and Clinical Immunology, Paris, France
| | - Dharam J Kumbhani
- Division of Cardiology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX
| | | | - Laleh Amiri-Kordestani
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| | - Alexander R Lyon
- Cardio-Oncology Service, Royal Brompton Hospital, London, United Kingdom
| | - Paaladinesh Thavendiranathan
- Peter Munk Cardiac Centre, Ted Rogers Program in Cardiotoxicity Prevention and Department of Medical Imaging, University Health Network, University of Toronto, Ontario, Canada
| | - Robert Padera
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Andrew Lichtman
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Peter P Liu
- Departments of Medicine and Cellular & Molecular Medicine, University of Ottawa Heart Institute, Ontario, Canada
| | - Douglas B Johnson
- Division of Oncology, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Javid Moslehi
- Division of Cardiovascular Medicine, Clinical Pharmacology, Cardio-Oncology Program, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN.,Division of Oncology, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN
| |
Collapse
|
85
|
Stack JP, Moslehi J, Sayed N, Wu JC. Cancer therapy-induced cardiomyopathy: can human induced pluripotent stem cell modelling help prevent it? Eur Heart J 2019; 40:1764-1770. [PMID: 29377985 PMCID: PMC6554650 DOI: 10.1093/eurheartj/ehx811] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 10/12/2017] [Accepted: 12/22/2017] [Indexed: 11/14/2022] Open
Abstract
Cardiotoxic effects from cancer therapy are a major cause of morbidity during cancer treatment. Unexpected toxicity can occur during treatment and/or after completion of therapy, into the time of cancer survivorship. While older drugs such as anthracyclines have well-known cardiotoxic effects, newer drugs such as tyrosine kinase inhibitors, proteasome inhibitors, and immunotherapies also can cause diverse cardiovascular and metabolic complications. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are increasingly being used as instruments for disease modelling, drug discovery, and mechanistic toxicity studies. Promising results with hiPSC-CM chemotherapy studies are raising hopes for improving cancer therapies through personalized medicine and safer drug development. Here, we review the cardiotoxicity profiles of common chemotherapeutic agents as well as efforts to model them in vitro using hiPSC-CMs.
Collapse
Affiliation(s)
- Jonathan P Stack
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- The Institute for Stem Cell Biology and Regenerative Medicine, 265 Campus Drive, 3rd Floor, Stanford, CA, USA
- Division of Cardiology, Department of Medicine, Stanford University School of Medicine, 300 Pasteur Drive S102, Stanford, CA, USA
- Department of Comparative Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Edwards, Stanford, CA, USA
| | - Javid Moslehi
- Division of Cardiology, Department of Medicine, Vanderbilt School of Medicine, 2220 Pierce Avenue, 383 Preston Research Building, Nashville, TN USA
- Cardio-Oncology Program, Vanderbilt School of Medicine, 2220 Pierce Avenue, 383 Preston Research Building, Nashville, TN, USA
| | - Nazish Sayed
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- The Institute for Stem Cell Biology and Regenerative Medicine, 265 Campus Drive, 3rd Floor, Stanford, CA, USA
- Division of Cardiology, Department of Medicine, Stanford University School of Medicine, 300 Pasteur Drive S102, Stanford, CA, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- The Institute for Stem Cell Biology and Regenerative Medicine, 265 Campus Drive, 3rd Floor, Stanford, CA, USA
- Division of Cardiology, Department of Medicine, Stanford University School of Medicine, 300 Pasteur Drive S102, Stanford, CA, USA
| |
Collapse
|
86
|
Linton MF, Moslehi JJ, Babaev VR. Akt Signaling in Macrophage Polarization, Survival, and Atherosclerosis. Int J Mol Sci 2019; 20:ijms20112703. [PMID: 31159424 PMCID: PMC6600269 DOI: 10.3390/ijms20112703] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 05/28/2019] [Accepted: 05/29/2019] [Indexed: 12/15/2022] Open
Abstract
The PI3K/Akt pathway plays a crucial role in the survival, proliferation, and migration of macrophages, which may impact the development of atherosclerosis. Changes in Akt isoforms or modulation of the Akt activity levels in macrophages significantly affect their polarization phenotype and consequently atherosclerosis in mice. Moreover, the activity levels of Akt signaling determine the viability of monocytes/macrophages and their resistance to pro-apoptotic stimuli in atherosclerotic lesions. Therefore, elimination of pro-apoptotic factors as well as factors that antagonize or suppress Akt signaling in macrophages increases cell viability, protecting them from apoptosis, and this markedly accelerates atherosclerosis in mice. In contrast, inhibition of Akt signaling by the ablation of Rictor in myeloid cells, which disrupts mTORC2 assembly, significantly decreases the viability and proliferation of blood monocytes and macrophages with the suppression of atherosclerosis. In addition, monocytes and macrophages exhibit a threshold effect for Akt protein levels in their ability to survive. Ablation of two Akt isoforms, preserving only a single Akt isoform in myeloid cells, markedly compromises monocyte and macrophage viability, inducing monocytopenia and diminishing early atherosclerosis. These recent advances in our understanding of Akt signaling in macrophages in atherosclerosis may have significant relevance in the burgeoning field of cardio-oncology, where PI3K/Akt inhibitors being tested in cancer patients can have significant cardiovascular and metabolic ramifications.
Collapse
Affiliation(s)
- MacRae F Linton
- Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232-6300, USA.
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232-6300, USA.
| | - Javid J Moslehi
- Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232-6300, USA.
| | - Vladimir R Babaev
- Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232-6300, USA.
| |
Collapse
|
87
|
Bertero E, Ameri P, Maack C. Bidirectional Relationship Between Cancer and Heart Failure: Old and New Issues in Cardio-oncology. Card Fail Rev 2019; 5:106-111. [PMID: 31179021 PMCID: PMC6546001 DOI: 10.15420/cfr.2019.1.2] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 02/14/2019] [Indexed: 02/07/2023] Open
Abstract
The main focus of cardio-oncology has been the prevention and treatment of the cardiac toxicity of chemotherapy and radiotherapy. Furthermore, several targeted therapies have been associated with unexpected cardiotoxic side-effects. Recently, epidemiological studies reported a higher incidence of cancer in patients with heart failure (HF) compared with individuals without HF. On this basis, it has been proposed that HF might represent an oncogenic condition. This hypothesis is supported by preclinical studies demonstrating that hyperactivation of the sympathetic nervous system and renin-angiotensin-aldosterone system, which is a hallmark of HF, promotes cancer growth and dissemination. Another intriguing possibility is that the co-occurrence of HF and cancer is promoted by a common pathological milieu characterised by a state of chronic low-grade inflammation, which predisposes to both diseases. In this review, we provide an overview of the mechanisms underlying the bidirectional relationship between HF and cancer.
Collapse
Affiliation(s)
- Edoardo Bertero
- Comprehensive Heart Failure Center, University Clinic Würzburg Würzburg, Germany
| | - Pietro Ameri
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino - IRCCS Italian Cardiovascular Network Genova, Italy.,Department of Internal Medicine and Centre of Excellence for Biomedical Research, University of Genova Genova, Italy
| | - Christoph Maack
- Comprehensive Heart Failure Center, University Clinic Würzburg Würzburg, Germany
| |
Collapse
|
88
|
Lenihan DJ. The Horizon in Cardio-Oncology: "You Are Only as Good as Your Endothelium". J Am Coll Cardiol 2019; 70:163-164. [PMID: 28683963 DOI: 10.1016/j.jacc.2017.05.048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 05/26/2017] [Indexed: 11/26/2022]
Affiliation(s)
- Daniel J Lenihan
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
89
|
Nee A, Lipton JH. Update on the management of chronic myeloid leukemia: current best practice and future directions. Expert Opin Orphan Drugs 2019. [DOI: 10.1080/21678707.2019.1609939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Aisling Nee
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Jeffrey H. Lipton
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
90
|
Sayed N, Ameen M, Wu JC. Personalized medicine in cardio-oncology: the role of induced pluripotent stem cell. Cardiovasc Res 2019; 115:949-959. [PMID: 30768178 PMCID: PMC6933506 DOI: 10.1093/cvr/cvz024] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/14/2019] [Accepted: 02/06/2019] [Indexed: 12/19/2022] Open
Abstract
Treatment of cancer has evolved in the last decade with the introduction of new therapies. Despite these successes, the lingering cardiotoxic side-effects from chemotherapy remain a major cause of morbidity and mortality in cancer survivors. These effects can develop acutely during treatment, or even years later. Although many risk factors can be identified prior to beginning therapy, unexpected toxicity still occurs, often with lasting consequences. Specifically, cardiotoxicity results in cardiac cell death, eventually leading to cardiomyopathy and heart failure. Certain risk factors may predispose an individual to experiencing adverse cardiovascular effects, and when unexpected cardiotoxicity occurs, it is generally managed with supportive care. Animal models of chemotherapy-induced cardiotoxicity have provided some mechanistic insights, but the precise mechanisms by which these drugs affect the heart remains unknown. Moreover, the genetic rationale as to why some patients are more susceptible to developing cardiotoxicity has yet to be determined. Many genome-wide association studies have identified genomic variants that could be associated with chemotherapy-induced cardiotoxicity, but the lack of validation has made these studies more speculative rather than definitive. With the advent of human induced pluripotent stem cell (iPSC) technology, researchers not only have the opportunity to model human diseases, but also to screen drugs for their efficacy and toxicity using human cell models. Furthermore, it allows us to conduct validation studies to confirm the role of genomic variants in human diseases. In this review, we discuss the role of iPSCs in modelling chemotherapy-induced cardiotoxicity.
Collapse
Affiliation(s)
- Nazish Sayed
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Mohamed Ameen
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
91
|
Abstract
Subclinical left ventricular dysfunction is the most common cardiac complication after chemotherapy administration. Detection and early treatment are major issues for better cardiac outcomes in this cancer population. The most common definition of cardiotoxicity is a 10-percentage point decrease of left ventricular ejection fraction (LVEF) to a value <53%. The myocardial injury induced by chemotherapies is probably a continuum starting with cardiac biomarkers increase before the occurence of a structural myocardial deformation leading to a LVEF decline. An individualised risk profile (depending on age, cardiovascular risk factors, type of chemotherapy, baseline troponin, baseline global longitudinal strain and baseline LVEF) has to be determined before starting chemotherapy to consider cardioprotective treatment. To date, there is no proof of a systematic cardioprotective treatment (angiotensin-converting enzyme inhibitor and/or beta-blocker) in all cancer patients. However, early cardioprotective treatment in case of subclinical left ventricular dysfunction seems to be promising in the prevention of cardiac events.
Collapse
Affiliation(s)
- Martin Nicol
- Lariboisiere/Saint Louis Hospital, Cardiology Department Paris, France
| | - Mathilde Baudet
- Lariboisiere/Saint Louis Hospital, Cardiology Department Paris, France
| | - Alain Cohen-Solal
- Lariboisiere/Saint Louis Hospital, Cardiology Department Paris, France.,UMR-S942 (BioCANVAS), Paris Diderot University Paris, France
| |
Collapse
|
92
|
Totzeck M, Schuler M, Stuschke M, Heusch G, Rassaf T. Cardio-oncology - strategies for management of cancer-therapy related cardiovascular disease. Int J Cardiol 2019; 280:163-175. [PMID: 30661849 DOI: 10.1016/j.ijcard.2019.01.038] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 12/15/2018] [Accepted: 01/10/2019] [Indexed: 02/07/2023]
Abstract
Current therapy of advanced cancers is based on several modalities including radiotherapy, cytotoxic chemotherapy, molecularly targeted inhibitors and antibodies targeting immune checkpoints. All of those these modalities can negatively impact the cardiovascular system, and there is considerable experience in relation to radiotherapy and chemotherapy. In contrast, the knowledge base on cardiovascular toxicities of novel agents targeting signal transduction pathways and immune regulation is quite limited. In particular, potential late effects are of concern as cardiovascular pathology can negatively impact quality of life and prognosis in cancer survivors, particularly when additional cardiovascular risk factors are present. Treatment-associated adverse events include hypertension, venous thromboembolism, coronary artery disease, valvular heart disease, heart failure and arrhythmias. Early diagnosis of subclinical cardiotoxic effects of cancer therapies remains challenging. Integrated care, as provided by multidisciplinary cardio-oncology teams is the best option for prevention, diagnosis and treatment of cardiovascular diseases associated with cancer therapy. This review considers the cardiotoxic effects of specific cancer therapies and discusses novel diagnostic and therapeutic approaches as a reference for optimizing the care of cancer patients receiving novel cancer therapies.
Collapse
Affiliation(s)
- Matthias Totzeck
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University Hospital Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Martin Schuler
- Department of Medical Oncology, West German Cancer Center, Medical Faculty, University Hospital Essen, Hufelandstr. 55, 45147 Essen, Germany; German Cancer Consortium (DKTK), Partner site University Hospital Essen, Hufelandstrasse 55, 45147 Essen, Germany
| | - Martin Stuschke
- Department of Radiation Oncology, West German Cancer Center, Medical Faculty, University Hospital Essen, Hufelandstr. 55, 45147 Essen, Germany; German Cancer Consortium (DKTK), Partner site University Hospital Essen, Hufelandstrasse 55, 45147 Essen, Germany
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, Medical Faculty, University Hospital Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University Hospital Essen, Hufelandstr. 55, 45147 Essen, Germany.
| |
Collapse
|
93
|
Blaes AH, Thavendiranathan P, Moslehi J. Cardiac Toxicities in the Era of Precision Medicine: Underlying Risk Factors, Targeted Therapies, and Cardiac Biomarkers. Am Soc Clin Oncol Educ Book 2018; 38:764-774. [PMID: 30231407 DOI: 10.1200/edbk_208509] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cancer therapies can cause a variety of cardiac toxicities, including ischemia, cardiomyopathy, heart failure, myocarditis, arrhythmias, vascular disease, hypertension, and hyperlipidemia. Addressing cardiovascular risk at baseline, before initiating therapy, during cancer treatment, and in the survivorship period is imperative. It may be useful to risk stratify individuals with cardiovascular risk factors using biomarkers or imaging before they receive potentially cardiotoxic therapies. Additionally, new guidelines recommend cardiac imaging with echocardiography in the survivorship period 6 to 12 months after completing cancer therapy for these high-risk individuals. Close collaboration between cardiology and oncology in both clinical practice and future research is essential.
Collapse
Affiliation(s)
- Anne H Blaes
- From the Division of Hematology/Oncology, University of Minnesota, Minneapolis, MN; Peter Munk Cardiac Center, Toronto General Hospital, University Health Network, Division of Cardiology, University of Toronto, Toronto, ON, Canada; Division of Cardiology, Vanderbilt University, Nashville, TN
| | - Paaladinesh Thavendiranathan
- From the Division of Hematology/Oncology, University of Minnesota, Minneapolis, MN; Peter Munk Cardiac Center, Toronto General Hospital, University Health Network, Division of Cardiology, University of Toronto, Toronto, ON, Canada; Division of Cardiology, Vanderbilt University, Nashville, TN
| | - Javid Moslehi
- From the Division of Hematology/Oncology, University of Minnesota, Minneapolis, MN; Peter Munk Cardiac Center, Toronto General Hospital, University Health Network, Division of Cardiology, University of Toronto, Toronto, ON, Canada; Division of Cardiology, Vanderbilt University, Nashville, TN
| |
Collapse
|
94
|
Ye Y, Otahal P, Marwick TH, Wills KE, Neil AL, Venn AJ. Cardiovascular and other competing causes of death among patients with cancer from 2006 to 2015: An Australian population-based study. Cancer 2018; 125:442-452. [PMID: 30311655 DOI: 10.1002/cncr.31806] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 08/15/2018] [Accepted: 09/21/2018] [Indexed: 01/20/2023]
Abstract
BACKGROUND With improved cancer survivorship, cardiovascular disease (CVD) and other noncancer events compete with cancer as the underlying cause of death, but the risks of mortality in competing-risk settings have not been well characterized. METHODS The authors identified 21,637 individuals who had a first cancer registered between 2006 and 2013, with follow-up to 2015, in the Australian population-based Tasmanian Cancer Registry. The cumulative incidence of deaths from specific competing events was assessed in competing-risk analyses. Standardized mortality ratios (SMRs) and absolute excess risks (AERs) for deaths from noncancer causes were calculated for comparison with the general population. RESULTS Overall, 8844 deaths were observed, with 1946 (22%) from competing events. The cumulative incidence of deaths from CVD increased significantly with age at first cancer diagnosis and exceeded other competing events at age ≥65 years. The risk of death from CVD was more common than expected in the first year of follow-up (SMR, 1.44 [95% confidence interval, 1.26-1.64]; AER, 36.8 per 10,000 person-years). The SMR and AER for CVD deaths varied by first cancer site, indicating increased risks after a first diagnosis of lung cancer, hematologic malignancy, and urinary tract cancer. For other noncancer events, the SMRs increased significantly for deaths from infectious disease and respiratory disease and were highest in the first year of follow-up. CONCLUSIONS CVD was the leading cause of competing mortality among Tasmanian patients with cancer who were diagnosed from 2006 to 2013. The higher than expected occurrence of death from CVD and other noncancer events during the first year after a cancer diagnosis highlights the importance of early preventive interventions.
Collapse
Affiliation(s)
- Yuanzi Ye
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia.,Department of Pathology, Anhui Medical University, Hefei, Anhui Province, China
| | - Petr Otahal
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Thomas H Marwick
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia.,Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Karen E Wills
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Amanda L Neil
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Alison J Venn
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
95
|
Manolis AA, Manolis TA, Mikhailidis DP, Manolis AS. Cardiovascular safety of oncologic agents: a double-edged sword even in the era of targeted therapies - Part 2. Expert Opin Drug Saf 2018; 17:893-915. [PMID: 30126303 DOI: 10.1080/14740338.2018.1513489] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Patients with cancer are subject to the cardiotoxic effects of cancer therapy. Improved cancer treatments lead to more cancer-survivors, who though are exposed to various forms of cardiovascular (CV) disease (CVD) as they age. Aging patients are at increased risk of developing both malignancy and CVD or they may have survived some form of CVD as a result of effective CV treatments. Furthermore, patients with CVD may develop cancer and require treatment (and vice versa), all contributing to increased morbidity and mortality. The prevalence of both malignancy and CVD will increase due to the trend toward a longer lifespan. AREAS COVERED In part 2 of this review, the discussion of the CV effects of specific oncology drugs is completed with inclusion of additional immunological agents, current hormonal and other agents. Early detection and monitoring of cardiotoxicity, use of biomarkers and other imaging and diagnostic methods and prevention and treatment options are also discussed. EXPERT OPINION As outlined in part 1 of this review, oncologists need to be aware of the CV adverse-effects of their treatments and make careful and expectant clinical decisions, especially in patients with preexisting CVD or CV risk factors. Similarly, cardiologists should consider a detailed previous history of treatment for malignant disease, including prior chemotherapy exposure, dose(s) received, and/or combined modality therapy with chest radiotherapy. Both specialists should collaborate in order to minimize the impact of these two ubiquitous diseases (cancer and CVD) and mitigate the adverse effects of treatment modalities.
Collapse
Affiliation(s)
| | | | - Dimitri P Mikhailidis
- c Department of Clinical Biochemistry , Royal Free Hospital Campus, University College London Medical School , London , UK
| | - Antonis S Manolis
- d Third Department of Cardiology , Athens University School of Medicine , Athens , Greece
| |
Collapse
|
96
|
Aghel N, Delgado DH, Lipton JH. Cardiovascular events in chronic myeloid leukemia clinical trials. Is it time to reassess and report the events according to cardiology guidelines? Leukemia 2018; 32:2095-2104. [PMID: 30201984 DOI: 10.1038/s41375-018-0247-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 07/29/2018] [Accepted: 08/03/2018] [Indexed: 11/09/2022]
Abstract
Tyrosine kinase inhibitors (TKIs) have revolutionized the treatment of chronic myeloid leukemia (CML). Although these treatments have changed the natural course of CML and many other cancers, they may cause cardiovascular and/or metabolic complications. In this review, we discuss how overlooking the main drivers of cardiovascular events (CVEs) and lack of standard definitions for cardiovascular adverse events might have affected these event rates in CML trials. Methodological limitations that affect the available data are discussed, with an emphasis on the future direction of cardiovascular safety research in trials of investigational drugs in cancer treatment.
Collapse
Affiliation(s)
- Nazanin Aghel
- Division of Cardiology, Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON, Canada.
| | - Diego Hernan Delgado
- Division of Cardiology, Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Jeffrey Howard Lipton
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
97
|
Manolis AA, Manolis TA, Mikhailidis DP, Manolis AS. Cardiovascular safety of oncologic agents: A double-edged sword even in the era of targeted therapies - part 1. Expert Opin Drug Saf 2018; 17:875-892. [PMID: 30126304 DOI: 10.1080/14740338.2018.1513488] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Patients with cancer are subject to the cardiotoxic effects of cancer therapy and as more patients survive cancer due to improved treatment they are exposed to various forms of cardiovascular (CV) disease as they age, and vice-versa. Such an interplay of age with both malignancy and CV disease may contribute to increased morbidity and mortality. AREAS COVERED This two-part review considers the effects of cancer drug treatment on the CV system. In Part I, the various types of CV and cardiometabolic toxicity of anti-cancer drugs and the possible mechanisms involved are discussed. Also, among the specific oncologic agents, the CV effects of the classical agents and of the large molecule immunological agents (monoclonal antibodies, including immune checkpoint inhibitors) are detailed. EXPERT OPINION Oncologic agents produce a variety of CV adverse effects, including cardiomyopathy and heart failure, peri-myocarditis, coronary artery disease, peripheral vascular disease, hypertension (HTN), cardiac arrhythmias, valvular heart disease, and pulmonary HTN. Both the oncologist and the cardiologist need to be aware of such adverse effects and of the specific agents that produce them. They need to join forces to prevent, anticipate, recognize, and manage such complications.
Collapse
Affiliation(s)
| | | | - Dimitri P Mikhailidis
- c Department of Clinical Biochemistry , University College London Medical School , London , UK
| | - Antonis S Manolis
- d Third Department of Cardiology , Athens University School of Medicine , Athens , Greece
| |
Collapse
|
98
|
Justice CN, Derbala MH, Baich TM, Kempton AN, Guo AS, Ho TH, Smith SA. The Impact of Pazopanib on the Cardiovascular System. J Cardiovasc Pharmacol Ther 2018; 23:387-398. [PMID: 29706106 PMCID: PMC6257996 DOI: 10.1177/1074248418769612] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Pazopanib is an approved treatment for renal cell carcinoma and a second-line treatment for nonadipocytic soft-tissue sarcoma. However, its clinical efficacy is limited by its cardiovascular side effects. Pazopanib and other vascular endothelial growth factor receptor tyrosine kinase inhibitors have been associated with the development of hypertension, QT interval prolongation, and other cardiovascular events; however, these mechanisms are largely unknown. Gaining a deeper understanding of these mechanisms is essential for the development of appropriate surveillance strategies and possible diagnostic biomarkers to allow us to monitor patients and modulate therapy prior to significant cardiac insult. This approach will be vital in keeping patients on these life-saving therapies and may be applicable to other tyrosine kinase inhibitors as well. In this review, we provide a comprehensive overview of the preclinical and clinical side effects of pazopanib with a focus on the mechanisms responsible for its toxicity to the cardiovascular system.
Collapse
Affiliation(s)
- Cody N. Justice
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Mohamed H. Derbala
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Tesla M. Baich
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Amber N. Kempton
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Aaron S. Guo
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Thai H. Ho
- Mayo Clinic Scottsdale, Arizona, Phoenix, AZ, USA
| | - Sakima A. Smith
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
- Division of Cardiology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| |
Collapse
|
99
|
Jordan JH, Todd RM, Vasu S, Hundley WG. Cardiovascular Magnetic Resonance in the Oncology Patient. JACC Cardiovasc Imaging 2018; 11:1150-1172. [PMID: 30092971 PMCID: PMC6242266 DOI: 10.1016/j.jcmg.2018.06.004] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 06/05/2018] [Accepted: 06/14/2018] [Indexed: 01/20/2023]
Abstract
Patients with or receiving potentially cardiotoxic treatment for cancer are susceptible to developing decrements in left ventricular mass, diastolic function, or systolic function. They may also experience valvular heart disease, pericardial disease, or intracardiac masses. Cardiovascular magnetic resonance may be used to assess cardiac anatomy, structure, and function and to characterize myocardial tissue. This combination of features facilitates the diagnosis and management of disease processes in patients with or those who have survived cancer. This report outlines and describes prior research involving cardiovascular magnetic resonance for assessing cardiovascular disease in patients with or previously having received treatment for cancer.
Collapse
Affiliation(s)
- Jennifer H Jordan
- Department of Internal Medicine, Section on Cardiovascular Medicine at the Wake Forest School of Medicine, Winston-Salem, North Carolina.
| | - Ryan M Todd
- Department of Internal Medicine, Section on Cardiovascular Medicine at the Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Sujethra Vasu
- Department of Internal Medicine, Section on Cardiovascular Medicine at the Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - W Gregory Hundley
- Department of Internal Medicine, Section on Cardiovascular Medicine at the Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
100
|
Armenian SH, Armstrong GT, Aune G, Chow EJ, Ehrhardt MJ, Ky B, Moslehi J, Mulrooney DA, Nathan PC, Ryan TD, van der Pal HJ, van Dalen EC, Kremer LC. Cardiovascular Disease in Survivors of Childhood Cancer: Insights Into Epidemiology, Pathophysiology, and Prevention. J Clin Oncol 2018; 36:2135-2144. [PMID: 29874141 PMCID: PMC6804893 DOI: 10.1200/jco.2017.76.3920] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular disease (CVD), which includes cardiomyopathy/heart failure, coronary artery disease, stroke, pericardial disease, arrhythmias, and valvular and vascular dysfunction, is a major concern for long-term survivors of childhood cancer. There is clear evidence of increased risk of CVD largely attributable to treatment exposures at a young age, most notably anthracycline chemotherapy and chest-directed radiation therapy, and compounded by traditional cardiovascular risk factors accrued during decades after treatment exposure. Preclinical studies are limited; thus, it is a high priority to understand the pathophysiology of CVD as a result of anticancer treatments, taking into consideration the growing and developing heart. Recently developed personalized risk prediction models can provide decision support before initiation of anticancer therapy or facilitate implementation of screening strategies in at-risk survivors of cancer. Although consensus-based screening guidelines exist for the application of blood and imaging biomarkers of CVD, the most appropriate timing and frequency of these measures in survivors of childhood cancer are not yet fully elucidated. Longitudinal studies are needed to characterize the prognostic importance of subclinical markers of cardiovascular injury on long-term CVD risk. A number of prevention trials across the survivorship spectrum are under way, which include primary prevention (before or during cancer treatment), secondary prevention (after completion of treatment), and integrated approaches to manage modifiable cardiovascular risk factors. Ongoing multidisciplinary collaborations between the oncology, cardiology, primary care, and other subspecialty communities are essential to reduce therapeutic exposures and improve surveillance, prevention, and treatment of CVD in this high-risk population.
Collapse
Affiliation(s)
- Saro H. Armenian
- Saro H. Armenian, City of Hope, Duarte, CA; Gregory T. Armstrong, Matthew J. Ehrhardt, and Daniel A. Mulrooney, St Jude Children’s Research Hospital, Memphis; Javid Moslehi, Vanderbilt School of Medicine, Nashville, TN; Gregory Aune, Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX; Eric J. Chow, Fred Hutchinson Cancer Research Center, Seattle, WA; Bonnie Ky, University of Pennsylvania, Philadelphia, PA; Paul C. Nathan, The Hospital for Sick Children, Toronto, Ontario, Canada; Thomas D. Ryan, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; Helena J. van der Pal and Leontien C.M. Kremer, Princess Máxima Center for Pediatric Oncology, Utrecht; and Elvira C. van Dalen and Leontien C.M. Kremer, Emma Children’s Hospital/Academic Medical Center, Amsterdam, the Netherlands
| | - Gregory T. Armstrong
- Saro H. Armenian, City of Hope, Duarte, CA; Gregory T. Armstrong, Matthew J. Ehrhardt, and Daniel A. Mulrooney, St Jude Children’s Research Hospital, Memphis; Javid Moslehi, Vanderbilt School of Medicine, Nashville, TN; Gregory Aune, Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX; Eric J. Chow, Fred Hutchinson Cancer Research Center, Seattle, WA; Bonnie Ky, University of Pennsylvania, Philadelphia, PA; Paul C. Nathan, The Hospital for Sick Children, Toronto, Ontario, Canada; Thomas D. Ryan, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; Helena J. van der Pal and Leontien C.M. Kremer, Princess Máxima Center for Pediatric Oncology, Utrecht; and Elvira C. van Dalen and Leontien C.M. Kremer, Emma Children’s Hospital/Academic Medical Center, Amsterdam, the Netherlands
| | - Gregory Aune
- Saro H. Armenian, City of Hope, Duarte, CA; Gregory T. Armstrong, Matthew J. Ehrhardt, and Daniel A. Mulrooney, St Jude Children’s Research Hospital, Memphis; Javid Moslehi, Vanderbilt School of Medicine, Nashville, TN; Gregory Aune, Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX; Eric J. Chow, Fred Hutchinson Cancer Research Center, Seattle, WA; Bonnie Ky, University of Pennsylvania, Philadelphia, PA; Paul C. Nathan, The Hospital for Sick Children, Toronto, Ontario, Canada; Thomas D. Ryan, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; Helena J. van der Pal and Leontien C.M. Kremer, Princess Máxima Center for Pediatric Oncology, Utrecht; and Elvira C. van Dalen and Leontien C.M. Kremer, Emma Children’s Hospital/Academic Medical Center, Amsterdam, the Netherlands
| | - Eric J. Chow
- Saro H. Armenian, City of Hope, Duarte, CA; Gregory T. Armstrong, Matthew J. Ehrhardt, and Daniel A. Mulrooney, St Jude Children’s Research Hospital, Memphis; Javid Moslehi, Vanderbilt School of Medicine, Nashville, TN; Gregory Aune, Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX; Eric J. Chow, Fred Hutchinson Cancer Research Center, Seattle, WA; Bonnie Ky, University of Pennsylvania, Philadelphia, PA; Paul C. Nathan, The Hospital for Sick Children, Toronto, Ontario, Canada; Thomas D. Ryan, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; Helena J. van der Pal and Leontien C.M. Kremer, Princess Máxima Center for Pediatric Oncology, Utrecht; and Elvira C. van Dalen and Leontien C.M. Kremer, Emma Children’s Hospital/Academic Medical Center, Amsterdam, the Netherlands
| | - Matthew J. Ehrhardt
- Saro H. Armenian, City of Hope, Duarte, CA; Gregory T. Armstrong, Matthew J. Ehrhardt, and Daniel A. Mulrooney, St Jude Children’s Research Hospital, Memphis; Javid Moslehi, Vanderbilt School of Medicine, Nashville, TN; Gregory Aune, Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX; Eric J. Chow, Fred Hutchinson Cancer Research Center, Seattle, WA; Bonnie Ky, University of Pennsylvania, Philadelphia, PA; Paul C. Nathan, The Hospital for Sick Children, Toronto, Ontario, Canada; Thomas D. Ryan, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; Helena J. van der Pal and Leontien C.M. Kremer, Princess Máxima Center for Pediatric Oncology, Utrecht; and Elvira C. van Dalen and Leontien C.M. Kremer, Emma Children’s Hospital/Academic Medical Center, Amsterdam, the Netherlands
| | - Bonnie Ky
- Saro H. Armenian, City of Hope, Duarte, CA; Gregory T. Armstrong, Matthew J. Ehrhardt, and Daniel A. Mulrooney, St Jude Children’s Research Hospital, Memphis; Javid Moslehi, Vanderbilt School of Medicine, Nashville, TN; Gregory Aune, Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX; Eric J. Chow, Fred Hutchinson Cancer Research Center, Seattle, WA; Bonnie Ky, University of Pennsylvania, Philadelphia, PA; Paul C. Nathan, The Hospital for Sick Children, Toronto, Ontario, Canada; Thomas D. Ryan, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; Helena J. van der Pal and Leontien C.M. Kremer, Princess Máxima Center for Pediatric Oncology, Utrecht; and Elvira C. van Dalen and Leontien C.M. Kremer, Emma Children’s Hospital/Academic Medical Center, Amsterdam, the Netherlands
| | - Javid Moslehi
- Saro H. Armenian, City of Hope, Duarte, CA; Gregory T. Armstrong, Matthew J. Ehrhardt, and Daniel A. Mulrooney, St Jude Children’s Research Hospital, Memphis; Javid Moslehi, Vanderbilt School of Medicine, Nashville, TN; Gregory Aune, Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX; Eric J. Chow, Fred Hutchinson Cancer Research Center, Seattle, WA; Bonnie Ky, University of Pennsylvania, Philadelphia, PA; Paul C. Nathan, The Hospital for Sick Children, Toronto, Ontario, Canada; Thomas D. Ryan, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; Helena J. van der Pal and Leontien C.M. Kremer, Princess Máxima Center for Pediatric Oncology, Utrecht; and Elvira C. van Dalen and Leontien C.M. Kremer, Emma Children’s Hospital/Academic Medical Center, Amsterdam, the Netherlands
| | - Daniel A. Mulrooney
- Saro H. Armenian, City of Hope, Duarte, CA; Gregory T. Armstrong, Matthew J. Ehrhardt, and Daniel A. Mulrooney, St Jude Children’s Research Hospital, Memphis; Javid Moslehi, Vanderbilt School of Medicine, Nashville, TN; Gregory Aune, Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX; Eric J. Chow, Fred Hutchinson Cancer Research Center, Seattle, WA; Bonnie Ky, University of Pennsylvania, Philadelphia, PA; Paul C. Nathan, The Hospital for Sick Children, Toronto, Ontario, Canada; Thomas D. Ryan, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; Helena J. van der Pal and Leontien C.M. Kremer, Princess Máxima Center for Pediatric Oncology, Utrecht; and Elvira C. van Dalen and Leontien C.M. Kremer, Emma Children’s Hospital/Academic Medical Center, Amsterdam, the Netherlands
| | - Paul C. Nathan
- Saro H. Armenian, City of Hope, Duarte, CA; Gregory T. Armstrong, Matthew J. Ehrhardt, and Daniel A. Mulrooney, St Jude Children’s Research Hospital, Memphis; Javid Moslehi, Vanderbilt School of Medicine, Nashville, TN; Gregory Aune, Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX; Eric J. Chow, Fred Hutchinson Cancer Research Center, Seattle, WA; Bonnie Ky, University of Pennsylvania, Philadelphia, PA; Paul C. Nathan, The Hospital for Sick Children, Toronto, Ontario, Canada; Thomas D. Ryan, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; Helena J. van der Pal and Leontien C.M. Kremer, Princess Máxima Center for Pediatric Oncology, Utrecht; and Elvira C. van Dalen and Leontien C.M. Kremer, Emma Children’s Hospital/Academic Medical Center, Amsterdam, the Netherlands
| | - Thomas D. Ryan
- Saro H. Armenian, City of Hope, Duarte, CA; Gregory T. Armstrong, Matthew J. Ehrhardt, and Daniel A. Mulrooney, St Jude Children’s Research Hospital, Memphis; Javid Moslehi, Vanderbilt School of Medicine, Nashville, TN; Gregory Aune, Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX; Eric J. Chow, Fred Hutchinson Cancer Research Center, Seattle, WA; Bonnie Ky, University of Pennsylvania, Philadelphia, PA; Paul C. Nathan, The Hospital for Sick Children, Toronto, Ontario, Canada; Thomas D. Ryan, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; Helena J. van der Pal and Leontien C.M. Kremer, Princess Máxima Center for Pediatric Oncology, Utrecht; and Elvira C. van Dalen and Leontien C.M. Kremer, Emma Children’s Hospital/Academic Medical Center, Amsterdam, the Netherlands
| | - Helena J. van der Pal
- Saro H. Armenian, City of Hope, Duarte, CA; Gregory T. Armstrong, Matthew J. Ehrhardt, and Daniel A. Mulrooney, St Jude Children’s Research Hospital, Memphis; Javid Moslehi, Vanderbilt School of Medicine, Nashville, TN; Gregory Aune, Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX; Eric J. Chow, Fred Hutchinson Cancer Research Center, Seattle, WA; Bonnie Ky, University of Pennsylvania, Philadelphia, PA; Paul C. Nathan, The Hospital for Sick Children, Toronto, Ontario, Canada; Thomas D. Ryan, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; Helena J. van der Pal and Leontien C.M. Kremer, Princess Máxima Center for Pediatric Oncology, Utrecht; and Elvira C. van Dalen and Leontien C.M. Kremer, Emma Children’s Hospital/Academic Medical Center, Amsterdam, the Netherlands
| | - Elvira C. van Dalen
- Saro H. Armenian, City of Hope, Duarte, CA; Gregory T. Armstrong, Matthew J. Ehrhardt, and Daniel A. Mulrooney, St Jude Children’s Research Hospital, Memphis; Javid Moslehi, Vanderbilt School of Medicine, Nashville, TN; Gregory Aune, Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX; Eric J. Chow, Fred Hutchinson Cancer Research Center, Seattle, WA; Bonnie Ky, University of Pennsylvania, Philadelphia, PA; Paul C. Nathan, The Hospital for Sick Children, Toronto, Ontario, Canada; Thomas D. Ryan, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; Helena J. van der Pal and Leontien C.M. Kremer, Princess Máxima Center for Pediatric Oncology, Utrecht; and Elvira C. van Dalen and Leontien C.M. Kremer, Emma Children’s Hospital/Academic Medical Center, Amsterdam, the Netherlands
| | - Leontien C.M. Kremer
- Saro H. Armenian, City of Hope, Duarte, CA; Gregory T. Armstrong, Matthew J. Ehrhardt, and Daniel A. Mulrooney, St Jude Children’s Research Hospital, Memphis; Javid Moslehi, Vanderbilt School of Medicine, Nashville, TN; Gregory Aune, Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX; Eric J. Chow, Fred Hutchinson Cancer Research Center, Seattle, WA; Bonnie Ky, University of Pennsylvania, Philadelphia, PA; Paul C. Nathan, The Hospital for Sick Children, Toronto, Ontario, Canada; Thomas D. Ryan, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; Helena J. van der Pal and Leontien C.M. Kremer, Princess Máxima Center for Pediatric Oncology, Utrecht; and Elvira C. van Dalen and Leontien C.M. Kremer, Emma Children’s Hospital/Academic Medical Center, Amsterdam, the Netherlands
| |
Collapse
|