1
|
Wright JC, Sharma S, Potter AS. Arrhythmia Challenges in Cardio-Oncology: High-Risk Therapies, Management, and Anticoagulation. Cardiol Clin 2025; 43:43-56. [PMID: 39551561 DOI: 10.1016/j.ccl.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Cardiovascular disease and cancer are the leading cause of mortality in the United States. In 2021, there were 695,547 and 605,213 deaths due to heart disease and cancer, respectively. With novel oncologic and cardiac therapies, survival has improved leading to increased life-expectancy albeit with chronic illness burden. Arrhythmia management in patients with cancer, whether active or in remission, can be quite challenging. In this review, we will discuss high-risk oncological therapies, prevention, and management of Atrial fibrillation, Ventricular Arrhythmias, and Bradyarrhythmias.
Collapse
Affiliation(s)
- Jonathan C Wright
- Cardio-oncology Program, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Sneha Sharma
- Cardio-oncology Program, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Division of Cardiovascular Medicine, The Ohio State University, Columbus, OH, USA
| | - Adam S Potter
- Cardio-oncology Program, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Internal Medicine, The Ohio State University, Columbus, OH, USA; Division of Cardiovascular Medicine, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
2
|
Chen XD, Xiao KH, Zhou CB. Drug-induced retinal vein occlusion: a disproportionality analysis from the FDA adverse event reporting system (2004-2023). Front Pharmacol 2024; 15:1480269. [PMID: 39734405 PMCID: PMC11671269 DOI: 10.3389/fphar.2024.1480269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/19/2024] [Indexed: 12/31/2024] Open
Abstract
Introduction Retinal vein occlusion (RVO) often causes irreversible visual impairment, making early prevention crucial. This study aims to identify associations between different medications and RVO and provide information for clinical practice. Method This study included reports of RVO from the FDA Adverse Event Reporting System (FAERS) database from the first quarter (Q1) of 2004 to the fourth quarter (Q4) of 2023. The reported drugs were analyzed for adverse drug reaction (ADR) signals using four disproportionality algorithms. Kaplan-Meier curves and median time to onset were used to evaluate the drugs. Results From 2004 to 2023, the FAERS database recorded 6,151 reports associated with RVO. Disproportionality analyses identified 25 drugs significantly associated with RVO. Mirabegron showed the highest risk signal, followed by Raloxifene, Tadalafil, Fingolimod, and Bimatoprost. These high-risk drugs are distributed across different therapeutic areas, including urogenital system and sex hormones, ophthalmic drugs, nervous system drugs, musculoskeletal system drugs, anti-tumor and immune-modulating drugs, and anti-parasitic drugs. Specific drug targets such as adrenergic receptor agonists, hormone regulators, and PDE5 inhibitors were identified as high risk. Ophthalmic drugs exhibited the longest median time to adverse ocular reactions at 532.01 days, followed by anti-parasitic drugs, nervous system drugs, urogenital system and sex hormone drugs, anti-tumor and immune-modulating drugs, and musculoskeletal system drugs. Conclusion This study provides an overview of drug-induced RVO, identifying potential culprit drugs and their distribution characteristics. These findings enhance understanding of medication safety and help optimize clinical practice.
Collapse
Affiliation(s)
- Xiao-Dong Chen
- Department of Ophthalmology, Hui’an County Hospital, Quanzhou, Fujian, China
- Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
- Department of Ophthalmology and Optometry, Fujian Medical University, Fuzhou, China
| | - Kun-Hong Xiao
- Department of Ophthalmology and Optometry, Fujian Medical University, Fuzhou, China
| | - Chao-Bing Zhou
- Department of Ophthalmology, Hui’an County Hospital, Quanzhou, Fujian, China
- Department of Ophthalmology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| |
Collapse
|
3
|
Zibrova D, Ernst T, Hochhaus A, Heller R. The BCR::ABL1 tyrosine kinase inhibitors ponatinib and nilotinib differentially affect endothelial angiogenesis and signalling. Mol Cell Biochem 2024:10.1007/s11010-024-05070-5. [PMID: 39009935 DOI: 10.1007/s11010-024-05070-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 07/09/2024] [Indexed: 07/17/2024]
Abstract
BCR::ABL1 inhibitors, the treatment of choice for the majority of patients with chronic myeloid leukaemia (CML), can cause vascular side effects that vary between agents. The exact underlying mechanisms are still poorly understood, but the vascular endothelium has been proposed as a site of origin. The present study investigates the effects of three BCR::ABL1 inhibitors, ponatinib, nilotinib and imatinib, on angiogenesis and signalling in human endothelial cells in response to vascular endothelial growth factor (VEGF). The experiments were performed in endothelial cells isolated from human umbilical veins. After exposure to imatinib, ponatinib and nilotinib, the angiogenic capacity of endothelial cells was assessed in spheroid assays. VEGF-induced signalling pathways were examined in Western blotting experiments using different specific antibodies. RNAi technology was used to downregulate proteins of interest. Intracellular cGMP levels were measured by ELISA. Imatinib had no effect on endothelial function. Ponatinib inhibited VEGF-induced sprouting, while nilotinib increased spontaneous and VEGF-stimulated angiogenesis. These effects did not involve wild-type ABL1 or ABL2, as siRNA-mediated knockdown of these kinases did not affect angiogenesis and VEGF signalling. Consistent with their effects on sprouting, ponatinib and nilotinib affected angiogenic pathways in opposite directions. While ponatinib inhibited VEGF-induced signalling and cGMP formation, nilotinib activated angiogenic signalling, in particular phosphorylation of extracellular signal-regulated kinase 1/2 (Erk1/2). The latter occurred in an epidermal growth factor receptor (EGFR)-dependent manner possibly via suppressing Fyn-related kinase (FRK), a negative regulator of EGFR signalling. Both, pharmacological inhibition of Erk1/2 or EGFR suppressed nilotinib-induced angiogenic sprouting. These results support the notion that the vascular endothelium is a site of action of BCR::ABL1 inhibitors from which side effects may arise, and that the different vascular toxicity profiles of BCR::ABL1 inhibitors may be due to their different actions at the molecular level. In addition, the as yet unknown pro-angiogenic effect of nilotinib should be considered in the treatment of patients with comorbidities associated with pathological angiogenesis, such as ocular disease, arthritis or obesity.
Collapse
Affiliation(s)
- Darya Zibrova
- Center for Molecular Biomedicine, Institute of Molecular Cell Biology, Jena University Hospital, Hans-Knöll-Straße 2, 07745, Jena, Germany
| | - Thomas Ernst
- Department of Hematology and Oncology, Jena University Hospital, Jena, Germany
| | - Andreas Hochhaus
- Department of Hematology and Oncology, Jena University Hospital, Jena, Germany
| | - Regine Heller
- Center for Molecular Biomedicine, Institute of Molecular Cell Biology, Jena University Hospital, Hans-Knöll-Straße 2, 07745, Jena, Germany.
| |
Collapse
|
4
|
Mozneb M, Jenkins A, Sances S, Pohlman S, Workman MJ, West D, Ondatje B, El-Ghazawi K, Woodbury A, Garcia VJ, Patel S, Arzt M, Dezem F, Laperle AH, Moser VA, Ho R, Yucer N, Plummer J, Barrett RJ, Svendsen CN, Sharma A. Multi-lineage heart-chip models drug cardiotoxicity and enhances maturation of human stem cell-derived cardiovascular cells. LAB ON A CHIP 2024; 24:869-881. [PMID: 38252454 DOI: 10.1039/d3lc00745f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
Cardiovascular toxicity causes adverse drug reactions and may lead to drug removal from the pharmaceutical market. Cancer therapies can induce life-threatening cardiovascular side effects such as arrhythmias, muscle cell death, or vascular dysfunction. New technologies have enabled cardiotoxic compounds to be identified earlier in drug development. Human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CMs) and vascular endothelial cells (ECs) can screen for drug-induced alterations in cardiovascular cell function and survival. However, most existing hiPSC models for cardiovascular drug toxicity utilize two-dimensional, immature cells grown in static culture. Improved in vitro models to mechanistically interrogate cardiotoxicity would utilize more adult-like, mature hiPSC-derived cells in an integrated system whereby toxic drugs and protective agents can flow between hiPSC-ECs that represent systemic vasculature and hiPSC-CMs that represent heart muscle (myocardium). Such models would be useful for testing the multi-lineage cardiotoxicities of chemotherapeutic drugs such as VEGFR2/PDGFR-inhibiting tyrosine kinase inhibitors (VPTKIs). Here, we develop a multi-lineage, fully-integrated, cardiovascular organ-chip that can enhance hiPSC-EC and hiPSC-CM functional and genetic maturity, model endothelial barrier permeability, and demonstrate long-term functional stability. This microfluidic organ-chip harbors hiPSC-CMs and hiPSC-ECs on separate channels that can be subjected to active fluid flow and rhythmic biomechanical stretch. We demonstrate the utility of this cardiovascular organ-chip as a predictive platform for evaluating multi-lineage VPTKI toxicity. This study may lead to the development of new modalities for the evaluation and prevention of cancer therapy-induced cardiotoxicity.
Collapse
Affiliation(s)
- Maedeh Mozneb
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Amelia Jenkins
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Samuel Sances
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Stephany Pohlman
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Michael J Workman
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Dylan West
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Briana Ondatje
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Kareem El-Ghazawi
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Amanda Woodbury
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Veronica J Garcia
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Shachi Patel
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Madelyn Arzt
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Felipe Dezem
- Center for Bioinformatics and Functional Genomics, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Alex H Laperle
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - V Alexandra Moser
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Ritchie Ho
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Nur Yucer
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Jasmine Plummer
- Center for Bioinformatics and Functional Genomics, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Robert J Barrett
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Clive N Svendsen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Arun Sharma
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
5
|
Barachini S, Ghelardoni S, Varga ZV, Mehanna RA, Montt-Guevara MM, Ferdinandy P, Madonna R. Antineoplastic drugs inducing cardiac and vascular toxicity - An update. Vascul Pharmacol 2023; 153:107223. [PMID: 37678516 DOI: 10.1016/j.vph.2023.107223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/14/2023] [Accepted: 09/04/2023] [Indexed: 09/09/2023]
Abstract
With the improvement in cancer prognosis due to advances in antitumor therapeutic protocols and new targeted and immunotherapies, we are witnessing a growing increase in survival, however, at the same timeincrease in morbidity among cancer survivors as a consequences of the increased cardiovascular adverse effects of antineoplastic drugs. Common cardiovascular complications of antineoplastic therapies may include cardiac complications such as arrhythmias, myocardial ischemia, left ventricular dysfunction culminating in heart failure as well as vascular complications including arterial hypertension, thromboembolic events, and accelerated atherosclerosis. The toxicity results from the fact that these drugs not only target cancer cells but also affect normal cells within the cardiovascular system. In this article, we review the clinical features and main mechanisms implicated in antineoplastic drug-induced cardiovascular toxicity, including oxidative stress, inflammation, immunothrombosis and growth factors-induced signaling pathways.
Collapse
Affiliation(s)
- Serena Barachini
- Department of Clinical and Experimental Medicine, Laboratory for Cell Therapy, University of Pisa, Pisa, Italy
| | - Sandra Ghelardoni
- Department of Pathology, Laboratory of Biochemistry, University of Pisa, Pisa, Italy
| | - Zoltán V Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary; MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Radwa A Mehanna
- Medical Physiology Department, Center of Excellence for Research in Regenerative Medicine and Applications (CERRMA), Faculty of Medicine, Alexandria University, Egypt
| | | | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Rosalinda Madonna
- Department of Pathology, Cardiology Division, University of Pisa, Pisa, Italy.
| |
Collapse
|
6
|
Larsen CM, Garcia Arango M, Dasari H, Arciniegas Calle M, Adjei E, Rico Mesa J, Scott CG, Thompson CA, Cerhan JR, Haddad TC, Goetz MP, Herrmann J, Villarraga HR. Association of Anthracycline With Heart Failure in Patients Treated for Breast Cancer or Lymphoma, 1985-2010. JAMA Netw Open 2023; 6:e2254669. [PMID: 36735254 PMCID: PMC9898820 DOI: 10.1001/jamanetworkopen.2022.54669] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
IMPORTANCE Anthracyclines increase the risk for congestive heart failure (CHF); however, long-term cumulative incidence and risk factors for CHF after anthracycline therapy are not well defined in population-based studies. OBJECTIVE To compare the long-term cumulative incidence of CHF in patients with breast cancer or lymphoma treated with anthracycline therapy compared with healthy controls from the same community. DESIGN, SETTING, AND PARTICIPANTS This retrospective population-based case-control study included data from the Rochester Epidemiology Project. Participants included residents of Olmsted County, Minnesota, diagnosed with breast cancer or lymphoma from January 1985 through December 2010 matched for age, sex, and comorbidities with healthy controls, with a final ratio of 1 case to 1.5 controls. Statistical analysis was performed between July 2017 and February 2022. EXPOSURES Cancer treatment and CHF risk factors. MAIN OUTCOMES AND MEASURES The main outcome was new-onset CHF, as defined by the modified Framingham criteria. Cox proportional hazards regression was used to estimate hazard ratios (HRs) to compare the risk of CHF in participants with cancer vs controls, adjusted for age, sex, diabetes, hypertension, hyperlipidemia, coronary artery disease, obesity, and smoking history. RESULTS A total of 2196 individuals were included, with 812 patients with cancer and 1384 participants without cancer. The mean (SD) age was 52.62 (14.56) years and 1704 participants (78%) were female. Median (IQR) follow-up was 8.6 (5.2-13.4) years in the case group vs 12.5 (8.7-17.5) years in the control group. Overall, patients with cancer had higher risk of CHF compared with the control cohort even after adjusting for age, sex, diabetes, hypertension, coronary artery disease, hyperlipidemia, obesity, and smoking status (HR, 2.86 [95% CI, 1.90-4.32]; P < .001). After adjusting for the same variables, CHF risk was greater for patients with cancer receiving anthracycline (HR, 3.25 [95% CI, 2.11-5.00]; P < .001) and was attenuated and lost statistical significance for patients with cancer not receiving anthracyclines (HR, 1.78 [95% CI, 0.83-3.81]; P = .14). Higher cumulative incidence for patients treated with anthracyclines vs comparator cohort was observed at 1 year (1.81% vs 0.09%), 5 years (2.91% vs 0.79%), 10 years (5.36% vs 1.74%), 15 years (7.42% vs 3.18%), and 20 years (10.75% vs 4.98%) (P < .001). There were no significant differences in risk of CHF for patients receiving anthracycline at a dose of less than 180 mg/m2 compared with those at a dose of 180 to 250 mg/m2 (HR, 0.54 [95% CI, 0.19-1.51]) or at a dose of more than 250 mg/m2 (HR, 1.23 [95% CI, 0.52-2.91]). At diagnosis, age was an independent risk factor associated with CHF (HR per 10 years, 2.77 [95% CI, 1.99-3.86]; P < .001). CONCLUSIONS AND RELEVANCE In this retrospective population-based case-control study, anthracyclines were associated with an increased risk of CHF early during follow-up, and the increased risk persisted over time. The cumulative incidence of CHF in patients with breast cancer or lymphoma treated with anthracyclines at 15 years was more than 2-fold that of the control group.
Collapse
Affiliation(s)
- Carolyn M. Larsen
- Department of Cardiovascular Medicine, Mayo Clinic, Scottsdale, Arizona
| | | | - Harika Dasari
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota
| | | | - Effie Adjei
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Juan Rico Mesa
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | | | | | - James R. Cerhan
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Tufia C. Haddad
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Matthew P. Goetz
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Joerg Herrmann
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | | |
Collapse
|
7
|
Leiva O, Bohart I, Ahuja T, Park D. Off-Target Effects of Cancer Therapy on Development of Therapy-Induced Arrhythmia: A Review. Cardiology 2023; 148:324-334. [PMID: 36702116 PMCID: PMC10614257 DOI: 10.1159/000529260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 01/17/2023] [Indexed: 01/27/2023]
Abstract
BACKGROUND Advances in cancer therapeutics have improved overall survival and prognosis in this patient population; however, this has come at the expense of cardiotoxicity including arrhythmia. SUMMARY Cancer and its therapies are associated with cardiotoxicity via several mechanisms including inflammation, cardiomyopathy, and off-target effects. Among cancer therapies, anthracyclines and tyrosine kinase inhibitors (TKIs) are particularly known for their pro-arrhythmia effects. In addition to cardiomyopathy, anthracyclines may be pro-arrhythmogenic via reactive oxygen species (ROS) generation and altered calcium handling. TKIs may mediate their cardiotoxicity via inhibition of off-target tyrosine kinases. Ibrutinib-mediated inhibition of CSK may be responsible for the increased prevalence of atrial fibrillation. Further investigation is warranted to further elucidate the mechanisms behind arrhythmias in cancer therapies. KEY MESSAGES Arrhythmias are a common cardiotoxicity of cancer therapies. Cancer therapies may induce arrhythmias via off-target effects. Understanding the mechanisms underlying arrhythmogenesis associated with cancer therapies may help design cancer therapies that can avoid these toxicities.
Collapse
Affiliation(s)
- Orly Leiva
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York City, New York, USA
| | - Isaac Bohart
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York City, New York, USA
| | - Tania Ahuja
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York City, New York, USA
| | - David Park
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York City, New York, USA
| |
Collapse
|
8
|
Carracedo M, Pawelzik SC, Artiach G, Pouwer MG, Plunde O, Saliba-Gustafsson P, Ehrenborg E, Eriksson P, Pieterman E, Stenke L, Princen HMG, Franco-Cereceda A, Bäck M. The tyrosine kinase inhibitor nilotinib targets discoidin domain receptor 2 in calcific aortic valve stenosis. Br J Pharmacol 2022; 179:4709-4721. [PMID: 35751904 PMCID: PMC9544120 DOI: 10.1111/bph.15911] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/16/2022] [Accepted: 04/14/2022] [Indexed: 11/27/2022] Open
Abstract
Background and Purpose Tyrosine kinase inhibitors (TKI) used to treat chronic myeloid leukaemia (CML) have been associated with cardiovascular side effects, including reports of calcific aortic valve stenosis. The aim of this study was to establish the effects of first and second generation TKIs in aortic valve stenosis and to determine the associated molecular mechanisms. Experimental Approach Hyperlipidemic APOE*3Leiden.CETP transgenic mice were treated with nilotinib, imatinib or vehicle. Human valvular interstitial cells (VICs) were isolated and studied in vitro. Gene expression analysis was perfromed in aortic valves from 64 patients undergoing aortic valve replacement surgery. Key Results Nilotinib increased murine aortic valve thickness. Nilotinib, but not imatinib, promoted calcification and osteogenic activation and decreased autophagy in human VICs. Differential tyrosine kinase expression was detected between healthy and calcified valve tissue. Transcriptomic target identification revealed that the discoidin domain receptor DDR2, which is preferentially inhibited by nilotinib, was predominantly expressed in human aortic valves but markedly downregulated in calcified valve tissue. Nilotinib and selective DDR2 targeting in VICs induced a similar osteogenic activation, which was blunted by increasing the DDR2 ligand, collagen. Conclusions and Implications These findings suggest that inhibition of DDR2 by nilotinib promoted aortic valve thickening and VIC calcification, with possible translational implications for cardiovascular surveillance and possible personalized medicine in CML patients.
Collapse
Affiliation(s)
| | - Sven-Christian Pawelzik
- Department of Medicine, Karolinska Institutet.,Theme Heart and Vessels, Division of Valvular and Coronary Disease, Karolinska University Hospital, Stockholm, Sweden
| | | | - Marianne G Pouwer
- Metabolic Health Research, Gaubius Laboratory, The Netherlands Organization of Applied Scientific Research (TNO), Leiden, the Netherlands
| | | | | | | | | | - Elsbet Pieterman
- Metabolic Health Research, Gaubius Laboratory, The Netherlands Organization of Applied Scientific Research (TNO), Leiden, the Netherlands
| | - Leif Stenke
- Department of Medicine, Karolinska Institutet.,Theme Cancer, Division of Hematology, Karolinska University Hospital
| | - Hans M G Princen
- Metabolic Health Research, Gaubius Laboratory, The Netherlands Organization of Applied Scientific Research (TNO), Leiden, the Netherlands
| | - Anders Franco-Cereceda
- Department of Molecular Medicine and Surgery, Karolinska Institutet.,Theme Heart and Vessels, Division of Valvular and Coronary Disease, Karolinska University Hospital, Stockholm, Sweden
| | - Magnus Bäck
- Department of Medicine, Karolinska Institutet.,Theme Heart and Vessels, Division of Valvular and Coronary Disease, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
9
|
Herrmann J, McCullough KB, Habermann TM. How I treat cardiovascular complications in patients with lymphoid malignancies. Blood 2022; 139:1501-1516. [PMID: 34752600 PMCID: PMC8914183 DOI: 10.1182/blood.2019003893] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/27/2021] [Indexed: 11/20/2022] Open
Abstract
The prognosis of several lymphoid malignancies has improved through development of novel therapies, combination with traditional chemotherapies, and delineation of appropriate therapeutic sequencing. Toxicities that are arising because of prolonged or multiple sequential therapeutic interventions are becoming increasingly impactful. Among the broad spectrum of complications that patients with lymphoid malignancies may experience, cardiovascular toxicities are significant in terms of morbidity and mortality. The entire cardiovascular system can be affected, but cardiomyopathy, heart failure, and arrhythmias remain of greatest concerns with the use of anthracyclines, hematopoietic stem cell transplantation, and radiation therapy in patients with lymphoid malignancies. These aspects will be covered in this article within the framework of case-based discussions. Key to the management of cardiovascular complications in patients with lymphoid malignancies is awareness and preparedness across the cancer continuum. Baseline risk stratification helps to direct surveillance and early intervention efforts before, during, and after cancer therapy, which are paramount for the best possible outcomes. Along these lines, the overall goal is to enable the best possible therapies for lymphoid malignancies without the complications of clinically significant cardiovascular events.
Collapse
Affiliation(s)
| | | | - Thomas M Habermann
- 3Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| |
Collapse
|
10
|
Chhabra N, Kennedy J. A Review of Cancer Immunotherapy Toxicity II: Adoptive Cellular Therapies, Kinase Inhibitors, Monoclonal Antibodies, and Oncolytic Viruses. J Med Toxicol 2022; 18:43-55. [PMID: 33821435 PMCID: PMC8021214 DOI: 10.1007/s13181-021-00835-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/29/2021] [Accepted: 02/18/2021] [Indexed: 12/14/2022] Open
Abstract
Immunotherapy for cancer has undergone a rapid expansion in classes, agents, and indications. By utilizing aspects of the body's innate immune system, immunotherapy has improved life expectancy and quality of life for patients with several types of cancer. Adoptive cellular therapies, including chimeric antigen receptor T (CAR T) cell therapy, involve the genetic engineering of patient T cells to allow for targeting of neoplastic cells. Monitoring of patients during the lymphodepletion prior to therapy and following CAR T cell infusion is necessary to detect toxicity of therapy. Specific toxicities include cytokine release syndrome and neurologic toxicity, both of which may be life-threatening. Tocilizumab and/or corticosteroids should be considered for moderate to severe toxicity. Kinase inhibitor toxicity can occur as "on target" effects or "off target" effects to multiple organ systems due to shared protein epitopes. Treatments are organ-specific. Infusion reactions are common during treatment with monoclonal antibodies and treatment is largely supportive. Clinical experience with oncolytic viruses is limited, but local reactions including cellulitis as well as systemic influenza-like syndromes have been seen but are typically mild. Although clinical experience with adverse effects due to newer immunotherapy agents is growing, an up-to-date understanding of their mechanisms and potential toxicities is critical.
Collapse
Affiliation(s)
- Neeraj Chhabra
- Department of Emergency Medicine, Division of Medical Toxicology, Cook County Health, 1950 W Polk Street, 7th Floor, Chicago, IL, 60612, USA.
- Toxikon Consortium, Chicago, IL, USA.
| | - Joseph Kennedy
- Department of Emergency Medicine, Division of Medical Toxicology, Cook County Health, 1950 W Polk Street, 7th Floor, Chicago, IL, 60612, USA
- Toxikon Consortium, Chicago, IL, USA
| |
Collapse
|
11
|
De Leo A, Di Simone E, Spano A, Puliani G, Petrone F. Nursing Management and Adverse Events in Thyroid Cancer Treatments with Tyrosine Kinase Inhibitors. A Narrative Review. Cancers (Basel) 2021; 13:cancers13235961. [PMID: 34885070 PMCID: PMC8656717 DOI: 10.3390/cancers13235961] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/19/2021] [Accepted: 11/24/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Tyrosine kinase inhibitors are an effective and promising therapy in the treatment of advanced differentiated medullary thyroid cancers. The prevention and management of new adverse events of these drugs are important to keep patients on their treatment course, avoiding drug discontinuation or interruption, and are associated with a faster recovery of the disease. The contribution of a multidisciplinary team of healthcare professionals optimizes the management of adverse events, maximizing the benefits and reducing the risks of treatment, consequently improving the quality of life of patients. Abstract Background: The advent of multikinase inhibitors has changed the treatment of advanced, metastatic, unresectable thyroid cancers, refractory to available treatments. These drugs cause new adverse events that should be prevented and treated for long periods, and sometimes beyond their discontinuation. The purpose of this narrative review was the description, prevention, and nursing management of the most frequent adverse events of locally advanced or metastatic differentiated thyroid cancer with sorafenib and lenvatinib, and medullary Thyroid cancer with vandetanib and cabozantinib treatment. Methods: A narrative literature review. Results: Studies included in this narrative review suggest that over 90% of patients treated with tyrosine kinase inhibitors experience at least 1 adverse event of any grade affecting their quality of life. Patients treated with tyrosine kinase inhibitors experienced at least one adverse event at any grade in ≥90% of cases, with a higher incidence in the first 6–8 weeks of treatment. The most frequent adverse events that can affect a patients’ quality of life are dermatological, gastrointestinal, cardiovascular, and metabolic. Conclusions: Early assessment of risk factors and identification of adverse events can help nurses support these patients throughout their clinical-therapeutic pathway, increasing the benefits of treatment and reducing reduction/discontinuation.
Collapse
Affiliation(s)
- Aurora De Leo
- Nursing: Technical, Rehabilitation, Assistance and Research Department-IRCCS Istituti Fisioterapici Ospitalieri and Regina Elena National Cancer Institute, 00144 Rome, Italy; (A.D.L.); (E.D.S.); (F.P.)
- Biomedicine and Prevention Department, Tor Vergata University, 00133 Rome, Italy
| | - Emanuele Di Simone
- Nursing: Technical, Rehabilitation, Assistance and Research Department-IRCCS Istituti Fisioterapici Ospitalieri and Regina Elena National Cancer Institute, 00144 Rome, Italy; (A.D.L.); (E.D.S.); (F.P.)
| | - Alessandro Spano
- Nursing: Technical, Rehabilitation, Assistance and Research Department-IRCCS Istituti Fisioterapici Ospitalieri and Regina Elena National Cancer Institute, 00144 Rome, Italy; (A.D.L.); (E.D.S.); (F.P.)
- Correspondence: ; Tel.: +39-0652-666-881
| | - Giulia Puliani
- Oncological Endocrinology Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Fabrizio Petrone
- Nursing: Technical, Rehabilitation, Assistance and Research Department-IRCCS Istituti Fisioterapici Ospitalieri and Regina Elena National Cancer Institute, 00144 Rome, Italy; (A.D.L.); (E.D.S.); (F.P.)
| |
Collapse
|
12
|
Unravelling the tangled web of hypertension and cancer. Clin Sci (Lond) 2021; 135:1609-1625. [PMID: 34240734 DOI: 10.1042/cs20200307] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 01/11/2023]
Abstract
Cardiovascular disease remains the primary cause of mortality globally, being responsible for an estimated 17 million deaths every year. Cancer is the second leading cause of death on a global level with roughly 9 million deaths per year being attributed to neoplasms. The two share multiple common risk factors such as obesity, poor physical exercise, older age, smoking and there exists rare monogenic hypertension syndromes. Hypertension is the most important risk factor for cardiovascular disease and affects more than a billion people worldwide and may also be a risk factor for the development of certain types of cancer (e.g. renal cell carcinoma (RCC)). The interaction space of the two conditions becomes more complicated when the well-described hypertensive effect of certain antineoplastic drugs is considered along with the extensive amount of literature on the association of different classes of antihypertensive drugs with cancer risk/prevention. The cardiovascular risks associated with antineoplastic treatment calls for efficient management of relative adverse events and the development of practical strategies for efficient decision-making in the clinic. Pharmacogenetic interactions between cancer treatment and hypertension-related genes is not to be ruled out, but the evidence is not still ample to be incorporated in clinical practice. Precision Medicine has the potential to bridge the gap of knowledge regarding the full spectrum of interactions between cancer and hypertension (and cardiovascular disease) and provide novel solutions through the emerging field of cardio-oncology. In this review, we aimed to examine the bidirectional associations between cancer and hypertension including pharmacotherapy.
Collapse
|
13
|
Paik DT, Chandy M, Wu JC. Patient and Disease-Specific Induced Pluripotent Stem Cells for Discovery of Personalized Cardiovascular Drugs and Therapeutics. Pharmacol Rev 2020; 72:320-342. [PMID: 31871214 PMCID: PMC6934989 DOI: 10.1124/pr.116.013003] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Human induced pluripotent stem cells (iPSCs) have emerged as an effective platform for regenerative therapy, disease modeling, and drug discovery. iPSCs allow for the production of limitless supply of patient-specific somatic cells that enable advancement in cardiovascular precision medicine. Over the past decade, researchers have developed protocols to differentiate iPSCs to multiple cardiovascular lineages, as well as to enhance the maturity and functionality of these cells. Despite significant advances, drug therapy and discovery for cardiovascular disease have lagged behind other fields such as oncology. We speculate that this paucity of drug discovery is due to a previous lack of efficient, reproducible, and translational model systems. Notably, existing drug discovery and testing platforms rely on animal studies and clinical trials, but investigations in animal models have inherent limitations due to interspecies differences. Moreover, clinical trials are inherently flawed by assuming that all individuals with a disease will respond identically to a therapy, ignoring the genetic and epigenomic variations that define our individuality. With ever-improving differentiation and phenotyping methods, patient-specific iPSC-derived cardiovascular cells allow unprecedented opportunities to discover new drug targets and screen compounds for cardiovascular disease. Imbued with the genetic information of an individual, iPSCs will vastly improve our ability to test drugs efficiently, as well as tailor and titrate drug therapy for each patient.
Collapse
Affiliation(s)
- David T Paik
- Stanford Cardiovascular Institute, Stanford University, Stanford, California
| | - Mark Chandy
- Stanford Cardiovascular Institute, Stanford University, Stanford, California
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University, Stanford, California
| |
Collapse
|
14
|
Abstract
Remarkable progress has been made in the development of new therapies for cancer, dramatically changing the landscape of treatment approaches for several malignancies and continuing to increase patient survival. Accordingly, adverse effects of cancer therapies that interfere with the continuation of best-possible care, induce life-threatening risks or lead to long-term morbidity are gaining increasing importance. Cardiovascular toxic effects of cancer therapeutics and radiation therapy are the epitome of such concerns, and proper knowledge, interpretation and management are needed and have to be placed within the context of the overall care of individual patients with cancer. Furthermore, the cardiotoxicity spectrum has broadened to include myocarditis with immune checkpoint inhibitors and cardiac dysfunction in the setting of cytokine release syndrome with chimeric antigen receptor T cell therapy. An increase in the incidence of arrhythmias related to inflammation such as atrial fibrillation can also be expected, in addition to the broadening set of cancer therapeutics that can induce prolongation of the corrected QT interval. Therefore, cardiologists of today have to be familiar not only with the cardiotoxicity associated with traditional cancer therapies, such as anthracycline, trastuzumab or radiation therapy, but even more so with an ever-increasing repertoire of therapeutics. This Review provides this information, summarizing the latest developments at the juncture of cardiology, oncology and haematology.
Collapse
Affiliation(s)
- Joerg Herrmann
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
15
|
Abstract
Cancer therapies can lead to a broad spectrum of cardiovascular complications. Among these, cardiotoxicities remain of prime concern, but vascular toxicities have emerged as the second most common group. The range of cancer therapies with a vascular toxicity profile and the clinical spectrum of vascular toxic effects are quite broad. Historically, venous thromboembolism has received the greatest attention but, over the past decade, the arterial toxic effects, which can present as acute vasospasm, acute thrombosis and accelerated atherosclerosis, of cancer therapies have gained greater recognition. This Review focuses on these types of cancer therapy-related arterial toxicity, including their mechanisms, and provides an update on venous thromboembolism and pulmonary hypertension associated with cancer therapies. Recommendations for the screening, treatment and prevention of vascular toxic effects of cancer therapies are outlined in the context of available evidence and society guidelines and consensus statements. The shift towards greater awareness of the vascular toxic effects of cancer therapies has further unveiled the urgent needs in this area in terms of defining best clinical practices. Well-designed and well-conducted clinical studies and registries are needed to more precisely define the incidence rates, risk factors, primary and secondary modes of prevention, and best treatment modalities for vascular toxicities related to cancer therapies. These efforts should be complemented by preclinical studies to outline the pathophysiological concepts that can be translated into the clinic and to identify drugs with vascular toxicity potential even before their widespread clinical use.
Collapse
Affiliation(s)
- Joerg Herrmann
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
16
|
Al-Jundi M, Thakur S, Gubbi S, Klubo-Gwiezdzinska J. Novel Targeted Therapies for Metastatic Thyroid Cancer-A Comprehensive Review. Cancers (Basel) 2020; 12:E2104. [PMID: 32751138 PMCID: PMC7463725 DOI: 10.3390/cancers12082104] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 07/22/2020] [Accepted: 07/25/2020] [Indexed: 12/18/2022] Open
Abstract
The knowledge on thyroid cancer biology has grown over the past decade. Thus, diagnostic and therapeutic strategies to manage thyroid cancer are rapidly evolving. With new insights into tumor biology and cancer genetics, several novel therapies have been approved for the treatment of thyroid cancer. Tyrosine kinase inhibitors (TKIs), such as lenvatinib and sorafenib, have been successfully utilized for the treatment of radioactive iodine (RAI)-refractory metastatic differentiated thyroid cancer (DTC). In addition, pretreatment with mitogen-activated protein kinase (MAPK) inhibitors (trametinib and selumetinib) has been shown to restore RAI avidity in previously RAI-refractory DTCs. Local therapies, such as external beam radiation and radiofrequency/ethanol ablation, have also been employed for treatment of DTC. Vandetanib and cabozantinib are the two TKIs currently approved by the Food and Drug Administration (FDA) for the treatment of medullary thyroid cancer (MTC). Other novel therapies, such as peptide receptor radionuclide therapy and carcinoembryonic antigen (CEA) vaccine, have also been utilized in treating MTC. Ongoing trials on selective rearranged-during-transfection (RET) protooncogene inhibitors, such as LOXO-292 and BLU-667, have demonstrated promising results in the treatment of metastatic MTC resistant to non-selective TKIs. The FDA-approved BRAF/MEK inhibitor combination of dabrafenib and trametinib has revolutionized treatment of BRAFV600E mutation positive anaplastic thyroid cancer. Several other emerging classes of medications, such as gene fusion inhibitors and immune checkpoint inhibitors, are being actively investigated in several clinical trials. In this review, we describe the molecular landscape of thyroid cancer and novel targeted therapies and treatment combinations available for the treatment of metastatic thyroid cancer.
Collapse
Affiliation(s)
| | | | | | - Joanna Klubo-Gwiezdzinska
- Thyroid Tumors and Functional Thyroid Disorders Section, Metabolic Disease Branch, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20814, USA; (M.A.-J.); (S.T.); (S.G.)
| |
Collapse
|
17
|
Graffagnino J, Kondapalli L, Arora G, Hawi R, Lenneman CG. Strategies to Prevent Cardiotoxicity. Curr Treat Options Oncol 2020; 21:32. [PMID: 32270293 DOI: 10.1007/s11864-020-0722-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OPINION STATEMENT Cardiovascular disease is a leading cause of death among cancer survivors. While the field of cardiology as a whole is driven by evidence generated through robust clinical trials, data in cardio-oncology is limited to a relatively small number of prospective clinical trials with heterogeneous groups of cancer patients. In addition, many pharmaceutical trials in oncology are flawed from a cardiovascular perspective because they exclude patients with significant cardiovascular (CV) history and have wide variation in the definitions of CV events and cardiotoxicity. Ultimately, oncology trials often underrepresent the possibility of cardiovascular events in a "real world" population. Thus, the signal for CV toxicity from a cancer treatment is often not manifested until phase IV studies; where we are often caught trying to mitigate the CV effects rather than preventing them. Most of the data about cardiotoxicity from cancer therapy and cardioprotective strategies has been developed from our experience in using anthracyclines for over 50 years with dramatic improvement in cancer survivorship. However, as we are in an era where cancer drug discovery is moving at lightning pace with increasing survival rates, it is imperative to move beyond anthracyclines and commit to research on the cardiovascular side effects of all aspects of cancer therapy with a focus on prevention. We emphasize the role of pre-cancer treatment CV assessment to anticipate cardiac issues and ultimately optimizing CV risk prior to cancer therapy as an opportunity to mitigate cardiovascular risk from cancer therapy.
Collapse
Affiliation(s)
- Jason Graffagnino
- Department of Medicine, University of Alabama at Birmingham, 321 Lyons Harrison Research Building, 1720 2nd Ave South, Birmingham, AL, 35294, USA
| | - Lavanya Kondapalli
- Division of Cardiology, Department of Medicine, University of Colorado, 12631 E. 17th Ave, Mail Stop B130, Aurora, CO, 80045, USA
| | - Garima Arora
- Department of Medicine, University of Alabama at Birmingham, 321 Lyons Harrison Research Building, 1720 2nd Ave South, Birmingham, AL, 35294, USA
- Division of Cardiology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Riem Hawi
- Department of Medicine, University of Alabama at Birmingham, 321 Lyons Harrison Research Building, 1720 2nd Ave South, Birmingham, AL, 35294, USA
- Division of Cardiology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Carrie G Lenneman
- Department of Medicine, University of Alabama at Birmingham, 321 Lyons Harrison Research Building, 1720 2nd Ave South, Birmingham, AL, 35294, USA.
- Division of Cardiology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
18
|
Carracedo M, Stenke L, Franco-Cereceda A, Bäck M. Aortic Stenosis and the Tyrosine Kinase Inhibitor Nilotinib in Chronic Myeloid Leukemia. JACC: CARDIOONCOLOGY 2020; 2:123-126. [PMID: 34396217 PMCID: PMC8352255 DOI: 10.1016/j.jaccao.2019.12.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 12/15/2019] [Accepted: 12/17/2019] [Indexed: 01/07/2023]
Affiliation(s)
- Miguel Carracedo
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Leif Stenke
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden.,Theme Cancer, Division of Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Anders Franco-Cereceda
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Theme Heart and Vessels, Division of Valvular and Coronary Disease, Karolinska University Hospital, Stockholm, Sweden
| | - Magnus Bäck
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden.,Theme Cancer, Division of Hematology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
19
|
Cabanillas ME, Ryder M, Jimenez C. Targeted Therapy for Advanced Thyroid Cancer: Kinase Inhibitors and Beyond. Endocr Rev 2019; 40:1573-1604. [PMID: 31322645 PMCID: PMC7341904 DOI: 10.1210/er.2019-00007] [Citation(s) in RCA: 202] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 07/11/2019] [Indexed: 02/07/2023]
Abstract
The treatment of advanced thyroid cancer has undergone rapid evolution in the last decade, with multiple kinase inhibitor drug approvals for each subtype of thyroid cancer and a number of other commercially available drugs that have been studied for this indication. Although most of the US Food and Drug Administration (FDA)-approved drugs are antiangiogenic multikinase inhibitors-vandetanib, cabozantinib, sorafenib, lenvatinib-there are two FDA indications that are mutation specific-dabrafenib/trametinib for BRAF-mutated anaplastic thyroid cancer and larotrectinib for NTRK-fusion thyroid cancer. Furthermore, other mutation-specific drugs, immunotherapies, and novel strategies for advanced thyroid cancer are under investigation. Understanding the molecular basis of thyroid cancer, the drugs of interest for treatment of advanced thyroid cancer, and how these drugs can be administered safely and in the appropriate clinical scenario are the topics of this review.
Collapse
Affiliation(s)
- Maria E Cabanillas
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mabel Ryder
- Department of Endocrinology and Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Camilo Jimenez
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
20
|
Abstract
The introduction of targeted agents into modern cancer therapy pursued the goal of molecularly more specific, and thereby more effective and safer, therapies. Paradoxically, however, several toxicities were brought to greater attention, among these not only cardiac but also vascular toxicities. The latter reach far beyond venous thromboembolism and include a broad spectrum of presentations based on the vascular territories and pathomechanisms involved, including abnormal vascular reactivity, acute thrombosis, or accelerated atherosclerosis. This article provides an overview of the most common presentations and their management strategies.
Collapse
Affiliation(s)
- Joerg Herrmann
- Department of Cardiovascular Diseases, Mayo Clinic, 200 First Street SW, Rochester, MN 55902, USA.
| |
Collapse
|
21
|
Synthesis and biological evaluation of phenyl-amino-pyrimidine and indole/oxindole conjugates as potential BCR-ABL inhibitors. Med Chem Res 2019. [DOI: 10.1007/s00044-019-02318-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
22
|
Targeting Tumor Microenvironment for Cancer Therapy. Int J Mol Sci 2019; 20:ijms20040840. [PMID: 30781344 PMCID: PMC6413095 DOI: 10.3390/ijms20040840] [Citation(s) in RCA: 771] [Impact Index Per Article: 128.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 02/12/2019] [Accepted: 02/13/2019] [Indexed: 02/07/2023] Open
Abstract
Cancer development is highly associated to the physiological state of the tumor microenvironment (TME). Despite the existing heterogeneity of tumors from the same or from different anatomical locations, common features can be found in the TME maturation of epithelial-derived tumors. Genetic alterations in tumor cells result in hyperplasia, uncontrolled growth, resistance to apoptosis, and metabolic shift towards anaerobic glycolysis (Warburg effect). These events create hypoxia, oxidative stress and acidosis within the TME triggering an adjustment of the extracellular matrix (ECM), a response from neighbor stromal cells (e.g., fibroblasts) and immune cells (lymphocytes and macrophages), inducing angiogenesis and, ultimately, resulting in metastasis. Exosomes secreted by TME cells are central players in all these events. The TME profile is preponderant on prognosis and impacts efficacy of anti-cancer therapies. Hence, a big effort has been made to develop new therapeutic strategies towards a more efficient targeting of TME. These efforts focus on: (i) therapeutic strategies targeting TME components, extending from conventional therapeutics, to combined therapies and nanomedicines; and (ii) the development of models that accurately resemble the TME for bench investigations, including tumor-tissue explants, “tumor on a chip” or multicellular tumor-spheroids.
Collapse
|
23
|
Park JY, Guo W, Al-Hijji M, El Sabbagh A, Begna KH, Habermann TM, Witzig TE, Lewis BR, Lerman A, Herrmann J. Acute coronary syndromes in patients with active hematologic malignancies - Incidence, management, and outcomes. Int J Cardiol 2019; 275:6-12. [PMID: 30318297 PMCID: PMC10853038 DOI: 10.1016/j.ijcard.2018.10.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 09/22/2018] [Accepted: 10/02/2018] [Indexed: 01/21/2023]
Abstract
BACKGROUND Cancer and cardiovascular diseases are the two leading causes of death in industrialized countries. Optimal management of life-threatening presentations of both of their diseases can pose significant challenges. The current study aimed to address the incidence, management, and outcome of acute coronary syndromes (ACS) in patients with active hematological malignancies. METHODS This retrospective registry-based cohort study included adults with active leukemia or lymphoma who were hospitalized at Mayo Clinic Rochester from 01/01/2004 to 12/31/2014. The diagnosis of ST-segment elevation MI (STEMI) or non-ST-segment elevation MI (NSTEMI) was made based on the 3rd Universal Definition for MI, or of unstable angina (UA) in the absence of cardiac troponin elevation. Main outcome measures included all-cause, cardiac, and non-cardiac death in-hospital and at one year. RESULTS Of 5300 adult patients with active hematological malignancies, 73 (1.4%) were diagnosed with an ACS (78.1% NSTEMI and 13.7% STEMI). 17.5% and 40% of NSTEMI and STEMI patients underwent coronary angiography, with percutaneous coronary intervention in 5.3% and 30%, respectively. While >80% of patients received β-blocker therapy, only half of all and <50% of patients managed "medically" received antiplatelet, anticoagulant, and/or statin therapy. The in-hospital and 1-year mortality was 21.9% and 58.9%, respectively, of which 25% and 15% were cardiac in etiology. Aspirin, beta-blocker, statins, and angiotensin-converting enzyme inhibitor/angiotensin-II receptor blocker were associated with better mortality outcomes. CONCLUSIONS In a large, contemporary study of adults with active hematologic malignancies, ACS was uncommon, but commonly managed not in keeping with societal guideline recommendations.
Collapse
Affiliation(s)
- Jae Yoon Park
- Division of Cardiovascular Diseases, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
| | - Wei Guo
- Division of Cardiovascular Diseases, Mayo Clinic and Mayo Foundation, Rochester, MN, USA; Peking University People's Hospital, Department of Emergency Medicine, Beijing, China
| | - Mohammed Al-Hijji
- Division of Cardiovascular Diseases, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
| | - Abdallah El Sabbagh
- Division of Cardiovascular Diseases, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
| | - Kebede H Begna
- Division of Hematology/Oncology, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
| | - Thomas M Habermann
- Division of Hematology/Oncology, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
| | - Thomas E Witzig
- Division of Hematology/Oncology, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
| | - Bradley R Lewis
- Department of Biostatistics, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
| | - Amir Lerman
- Division of Cardiovascular Diseases, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
| | - Joerg Herrmann
- Division of Cardiovascular Diseases, Mayo Clinic and Mayo Foundation, Rochester, MN, USA.
| |
Collapse
|
24
|
Oren O, Herrmann J. Arterial events in cancer patients-the case of acute coronary thrombosis. J Thorac Dis 2018; 10:S4367-S4385. [PMID: 30701104 PMCID: PMC6328398 DOI: 10.21037/jtd.2018.12.79] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 12/14/2018] [Indexed: 12/21/2022]
Abstract
Patients with cancer are at high risk for both venous and arterial thrombotic complications. A variety of factors account for the greater thrombotic risk, including the underlying malignancy and numerous cancer-directed therapies. The occurrence of an acute thrombotic event in patients with cancer is associated with substantial morbidity and mortality. Acute coronary syndrome (ACS) represents a particularly important cardiovascular complication in cancer patients. With cardio-vascular risk factors becoming more prevalent in an aging cancer population that is surviving longer, questions pertaining to the appropriate management of vascular toxicity are likely to assume even greater value in the coming years. In this article, we review the current understanding of ACS in patients with cancer. The predisposition to thrombosis in a malignant host and the cancer treatments most commonly associated with vascular toxicity are reviewed. Risk prediction and management strategies are discussed, and discrepancies in the clinical evidence are highlighted.
Collapse
Affiliation(s)
- Ohad Oren
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | - Joerg Herrmann
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
25
|
Nhola LF, Abdelmoneim SS, Villarraga HR, Kohli M, Grothey A, Bordun KA, Cheung M, Best R, Cheung D, Huang R, Barros-Gomes S, Pitz M, Singal PK, Jassal DS, Mulvagh SL. Echocardiographic Assessment for the Detection of Cardiotoxicity Due to Vascular Endothelial Growth Factor Inhibitor Therapy in Metastatic Renal Cell and Colorectal Cancers. J Am Soc Echocardiogr 2018; 32:267-276. [PMID: 30459123 DOI: 10.1016/j.echo.2018.09.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Cardio-oncology is a recently established discipline that focuses on the management of patients with cancer who are at risk for developing cardiovascular complications as a result of their underlying oncologic treatment. In metastatic colorectal cancer (mCRC) and metastatic renal cell carcinoma (mRCC), vascular endothelial growth factor inhibitor (VEGF-i) therapy is commonly used to improve overall survival. Although these novel anticancer drugs may lead to the development of cardiotoxicity, whether early detection of cardiac dysfunction using serial echocardiography could potentially prevent the development of heart failure in this patient population requires further study. The aim of this study was to investigate the role of two-dimensional speckle-tracking echocardiography in the detection of cardiotoxicity due to VEGF-i therapy in patients with mCRC or mRCC. METHODS Patients with mRCC or mCRC were evaluated using serial echocardiography at baseline and 1, 3, and 6 months following VEGF-i treatment. RESULTS A total of 40 patients (34 men; mean age, 63 ± 9 years) receiving VEGF-i therapy were prospectively recruited at two academic centers: 26 (65%) were receiving sunitinib, eight (20%) pazopanib, and six (15%) bevacizumab. The following observations were made: (1) 8% of patients developed clinically asymptomatic cancer therapeutics-related cardiac dysfunction; (2) 30% of patients developed clinically significant decreases in global longitudinal strain, a marker for early subclinical cardiac dysfunction; (3) baseline abnormalities in global longitudinal strain may identify a subset of patients at higher risk for developing cancer therapeutics-related cardiac dysfunction; and (4) new or worsening hypertension was the most common adverse cardiovascular event, afflicting nearly one third of the study population. CONCLUSIONS Cardiac dysfunction defined by serial changes in myocardial strain assessed using two-dimensional speckle-tracking echocardiography occurs in patients undergoing treatment with VEGF-i for mCRC or mRCC, which may provide an opportunity for preventive interventions.
Collapse
Affiliation(s)
- Lara F Nhola
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Sahar S Abdelmoneim
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota; Division of Cardiology, Orman Heart Center, Assiut University, Assiut, Egypt
| | | | - Manish Kohli
- Department of Oncology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Axel Grothey
- Department of Oncology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Kimberly-Ann Bordun
- Institute of Cardiovascular Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Matthew Cheung
- Institute of Cardiovascular Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ryan Best
- Institute of Cardiovascular Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - David Cheung
- Institute of Cardiovascular Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Runqing Huang
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | | | - Marshall Pitz
- Section of Hematology/Oncology, Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Pawan K Singal
- Institute of Cardiovascular Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Davinder S Jassal
- Institute of Cardiovascular Sciences, University of Manitoba, Winnipeg, Manitoba, Canada; Section of Hematology/Oncology, Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada; Section of Cardiology, Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sharon L Mulvagh
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota; Division of Cardiology, Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
26
|
Tuzovic M, Herrmann J, Iliescu C, Marmagkiolis K, Ziaeian B, Yang EH. Arterial Thrombosis in Patients with Cancer. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2018; 20:40. [PMID: 29627870 PMCID: PMC7658957 DOI: 10.1007/s11936-018-0635-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Cancer is a common cause of morbidity and mortality in the USA. While the association between venous thrombosis and malignancy is well established, arterial thrombosis has more recently been recognized as a serious complication of cancer and certain chemotherapeutic agents. This review aims to summarize the most recent literature regarding the incidence and risk factors for cancer-related arterial thrombosis, understand the pathophysiologic mechanisms of thrombosis, and highlight the specific diagnostic and treatment considerations relevant to cancer patients. RECENT FINDINGS Based on a recent study looking at the Surveillance, Epidemiology, and End Results (SEER) database, the incidence of arterial thromboembolic events (ATEs) in patients with cancer at 6 months is 4.7%; the presence of an ATE is predictive of worse outcomes. Certain drugs such as platinum-based agents, vascular endothelial growth factor inhibitors, tyrosine kinase inhibitors, and taxanes have been associated with high rates of ATEs. Increased platelet reactivity appears crucial to development of arterial thrombosis in cancer patients. Cancer patients have an increased risk of arterial thrombosis that is likely due to both a cancer-associated procoagulant state as well as the adverse effects of certain chemotherapeutic agents. Treatment of arterial thromboembolism in cancer patients typically requires a multidisciplinary approach in part due to high rates of thrombocytopenia and stent thrombosis in the setting of percutaneous interventions. More studies are needed to investigate optimal prophylaxis, surveillance strategies, and treatments of cancer-related arterial thromboembolic disease.
Collapse
Affiliation(s)
- Mirela Tuzovic
- UCLA Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California at Los Angeles, 100 Medical Plaza, Suite 630, Los Angeles, CA, 90095, USA
| | - Joerg Herrmann
- Division of Cardiovascular Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Cezar Iliescu
- Division of Cardiology, Department of Medicine, MD Anderson Cancer Center, University of Texas at Houston, Houston, TX, USA
| | | | - Boback Ziaeian
- UCLA Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California at Los Angeles, 100 Medical Plaza, Suite 630, Los Angeles, CA, 90095, USA
| | - Eric H Yang
- UCLA Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California at Los Angeles, 100 Medical Plaza, Suite 630, Los Angeles, CA, 90095, USA.
| |
Collapse
|
27
|
Collins T, Gray K, Bista M, Skinner M, Hardy C, Wang H, Mettetal JT, Harmer AR. Quantifying the relationship between inhibition of VEGF receptor 2, drug-induced blood pressure elevation and hypertension. Br J Pharmacol 2018; 175:618-630. [PMID: 29161763 DOI: 10.1111/bph.14103] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 10/20/2017] [Accepted: 11/11/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE Several anti-angiogenic cancer drugs that inhibit VEGF receptor (VEGFR) signalling for efficacy are associated with a 15-60% incidence of hypertension. Tyrosine kinase inhibitors (TKIs) that have off-target activity at VEGFR-2 may also cause blood pressure elevation as an undesirable side effect. Therefore, the ability to translate VEGFR-2 off-target potency into blood pressure elevation would be useful in development of novel TKIs. Here, we have sought to quantify the relationship between VEGFR-2 inhibition and blood pressure elevation for a range of kinase inhibitors. EXPERIMENTAL APPROACH Porcine aortic endothelial cells overexpressing VEGFR-2 (PAE) were used to determine IC50 for VEGFR-2 phosphorylation. These IC50 values were compared with published reports of exposure attained during clinical use and the corresponding incidence of all-grade hypertension. Unbound average plasma concentration (Cav,u ) was selected to be the most appropriate pharmacokinetic parameter. The pharmacokinetic-pharmacodynamic (PKPD) relationship for blood pressure elevation was investigated for selected kinase inhibitors, using data derived either from clinical papers or from rat telemetry experiments. KEY RESULTS All-grade hypertension was predominantly observed when the Cav,u was >0.1-fold of the VEGFR-2 (PAE) IC50 . Furthermore, based on the PKPD analysis, an exposure-dependent blood pressure elevation >1 mmHg was observed only when the Cav,u was >0.1-fold of the VEGFR-2 (PAE) IC50 . CONCLUSIONS AND IMPLICATIONS Taken together, these data show that the risk of blood pressure elevation is proportional to the amount of VEGFR-2 inhibition, and a margin of >10-fold between VEGFR-2 IC50 and Cav,u appears to confer a minimal risk of hypertension.
Collapse
Affiliation(s)
- Teresa Collins
- AstraZeneca, Darwin Building, Cambridge Science Park, Milton Road, Cambridge, CB4, 0WG, UK
| | - Kelly Gray
- AstraZeneca, Darwin Building, Cambridge Science Park, Milton Road, Cambridge, CB4, 0WG, UK
| | - Michal Bista
- AstraZeneca, Darwin Building, Cambridge Science Park, Milton Road, Cambridge, CB4, 0WG, UK
| | - Matt Skinner
- AstraZeneca, Darwin Building, Cambridge Science Park, Milton Road, Cambridge, CB4, 0WG, UK
| | - Christopher Hardy
- AstraZeneca, Darwin Building, Cambridge Science Park, Milton Road, Cambridge, CB4, 0WG, UK
| | - Haiyun Wang
- AstraZeneca, Gatehouse Park, Waltham, MA, 02451, USA
| | | | - Alexander R Harmer
- AstraZeneca, Darwin Building, Cambridge Science Park, Milton Road, Cambridge, CB4, 0WG, UK
| |
Collapse
|
28
|
Tajiri K, Aonuma K, Sekine I. Cardiovascular toxic effects of targeted cancer therapy. Jpn J Clin Oncol 2017; 47:779-785. [PMID: 28531278 DOI: 10.1093/jjco/hyx071] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 05/07/2017] [Indexed: 01/02/2023] Open
Abstract
Over the past decade, there has been a major shift in chemotherapy from non-specific cytotoxic drugs to molecular targeted drug therapies. As more molecular targeted therapies are developed, new types of cardiovascular toxicities induced by targeted therapies are a growing problem. Cardiotoxicity induced by the human epidermal growth factor receptor-2 inhibitor trastuzumab manifests as decreased left ventricular ejection fraction. In contrast to anthracycline treatment, most cardiac events occur during trastuzumab treatment, but are reversed quickly when treatment is interrupted and cardiac intervention is established. Vascular endothelial growth factor pathway inhibitors decrease vascular tone, leading to hypertension. After drug initiation, the early detection and aggressive pharmacological management of hypertension are necessary to avoid severe complications. Cardiovascular safety is an emerging challenge in patients treated with newer generations of BCR-ABL inhibitors. Although rare, dasatinib-induced pulmonary hypertension is potentially fatal. Vascular events including cardiac and cerebral ischemic events and peripheral arterial occlusive disease have emerged as a new type of toxicity in patients treated with ponatinib and nilotinib. Thus, a wide variety of cardiovascular toxicities have been observed in patients treated with targeted drugs and have become a critically important topic of discussion for the practicing oncologist and cardiologists. Awareness of the potential side effects, recognition of signs and symptoms, and the establishment of therapeutic strategies are all crucial to providing quality patient care.
Collapse
Affiliation(s)
- Kazuko Tajiri
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba
| | - Kazutaka Aonuma
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba
| | - Ikuo Sekine
- Department of Medical Oncology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
29
|
Snipelisky D, Park JY, Lerman A, Mulvagh S, Lin G, Pereira N, Rodriguez-Porcel M, Villarraga HR, Herrmann J. How to Develop a Cardio-Oncology Clinic. Heart Fail Clin 2017; 13:347-359. [PMID: 28279420 DOI: 10.1016/j.hfc.2016.12.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cardiovascular demands to the care of cancer patients are common and important given the implications for morbidity and mortality. As a consequence, interactions with cardiovascular disease specialists have intensified to the point of the development of a new discipline termed cardio-oncology. As an additional consequence, so-called cardio-oncology clinics have emerged, in most cases staffed by cardiologists with an interest in the field. This article addresses this gap and summarizes key points in the development of a cardio-oncology clinic.
Collapse
Affiliation(s)
- David Snipelisky
- Department of Cardiovascular Diseases, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - Jae Yoon Park
- Department of Cardiovascular Diseases, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - Amir Lerman
- Department of Cardiovascular Diseases, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - Sharon Mulvagh
- Department of Cardiovascular Diseases, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - Grace Lin
- Department of Cardiovascular Diseases, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - Naveen Pereira
- Department of Cardiovascular Diseases, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - Martin Rodriguez-Porcel
- Department of Cardiovascular Diseases, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - Hector R Villarraga
- Department of Cardiovascular Diseases, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - Joerg Herrmann
- Department of Cardiovascular Diseases, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA.
| |
Collapse
|
30
|
Larsen CM, Mulvagh SL. Cardio-oncology: what you need to know now for clinical practice and echocardiography. Echo Res Pract 2017; 4:R33-R41. [PMID: 28254996 PMCID: PMC5435878 DOI: 10.1530/erp-17-0013] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 03/02/2017] [Indexed: 01/25/2023] Open
Abstract
Cardio-oncology is a rapidly growing field aimed at minimizing the effects of cardiovascular morbidity and mortality in cancer survivors. To meet this aim, patients are assessed at baseline to define their risk of cardiotoxicity and then followed closely during and after chemotherapy to assess for early signs or symptoms of cardiovascular disease. Cardiac imaging, and in particular, transthoracic echocardiography, plays an essential role in the baseline assessment and serial follow-up of cardio-oncology patients. The objectives of this paper are to review the mechanisms of cardiotoxicity of several common chemotherapeutic agents associated with an increased risk for left ventricular systolic dysfunction and to outline recommendations regarding the baseline assessment and serial follow-up of cardio-oncology patients with a focus on the role of echocardiography.
Collapse
Affiliation(s)
- Carolyn M Larsen
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Sharon L Mulvagh
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
31
|
Abstract
Acute kidney injury (AKI) is a growing problem with untoward economic and medical consequences. Anticancer drug toxicity remains an important and increasing cause of AKI. Importantly, drug-induced AKI affects all nephron segments—vasculature, glomerulus, tubules, and interstitium. Recent studies have increased insight into the subcellular mechanisms of drug-induced AKI that include direct cellular toxicity and immune-mediated effects. Identification of patients with high-risk cancer before drug exposure may allow prevention or at least a reduction in the development and severity of nephrotoxicity. Recognition of drug-induced AKI and rapid discontinuation (or dose reduction) of the offending agents, when appropriate, are critical to maximizing kidney function recovery. Preventive measures require understanding patient and drug-related risk factors coupled with correcting risk factors, assessing baseline kidney function before initiation of therapy, adjusting the drug dosage and avoiding use of nephrotoxic drug combinations.
Collapse
|
32
|
Brown SA, Nhola L, Herrmann J. Cardiovascular Toxicities of Small Molecule Tyrosine Kinase Inhibitors: An Opportunity for Systems-Based Approaches. Clin Pharmacol Ther 2016; 101:65-80. [DOI: 10.1002/cpt.552] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 10/31/2016] [Accepted: 10/31/2016] [Indexed: 12/12/2022]
Affiliation(s)
- S-A Brown
- Department of Cardiovascular Diseases; Mayo Clinic; Rochester Minnesota USA
| | - L Nhola
- Department of Cardiovascular Diseases; Mayo Clinic; Rochester Minnesota USA
| | - J Herrmann
- Department of Cardiovascular Diseases; Mayo Clinic; Rochester Minnesota USA
| |
Collapse
|