51
|
Suomalainen O, Pilv J, Loimaala A, Mätzke S, Heliö T, Uusitalo V. Prognostic significance of incidental suspected transthyretin amyloidosis on routine bone scintigraphy. J Nucl Cardiol 2022; 29:1021-1029. [PMID: 33094472 PMCID: PMC9163012 DOI: 10.1007/s12350-020-02396-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 09/14/2020] [Indexed: 01/15/2023]
Abstract
BACKGROUND Transthyretin amyloidosis (ATTR) is an occasional incidental finding on bone scintigraphy. We studied its prognostic impact in elderly patients. METHODS The study population consisted of 2000 patients aged over 70 years who underwent bone scintigraphies with clinical indications in three nuclear medicine departments (Kymenlaakso, Jorvi and Meilahti hospitals) in Finland. All studies were performed using 99mTechnetium labeled hydroxymethylene diphosphonate (HMDP). ATTR was suspected in patients with ≥grade 2 Perugini grade uptake (grade 0-3). Heart-to-contralateral ratio (H/CL) of ≥ 1.30 was considered positive for ATTR. The overall and cardiovascular mortality were obtained from the Finnish National Statistical Service. RESULTS There were a total of 1014 deaths (51%) and 177 cardiovascular deaths (9%) during median follow-up of 4 ± 2 years. ATTR was suspected in 69 patients (3.6%) of which 54 (2.7%) had grade 2 and 15 (.8%) had grade 3 uptake and in 47 patients (2.4%) by H/CL ratio. In multivariate analyses age, bone metastasis, H/CL ratio and grade 3 uptake were independent predictors of overall and cardiovascular mortality. Grade 2 uptake was a predictor of cardiovascular mortality. CONCLUSIONS A suspected ATTR as an incidental finding on bone scintigraphy predicts elevated overall and cardiovascular mortality in elderly patients.
Collapse
Affiliation(s)
- Olli Suomalainen
- Cardiology Department, Kymenlaakso Central Hospital, Kotkantie 41, 48210 Kotka, Finland
| | - Jaagup Pilv
- Clinical Physiology and Nuclear Medicine, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Antti Loimaala
- Clinical Physiology and Nuclear Medicine, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Sorjo Mätzke
- Clinical Physiology and Nuclear Medicine, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Tiina Heliö
- Heart and Lung Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Valtteri Uusitalo
- Clinical Physiology and Nuclear Medicine, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| |
Collapse
|
52
|
Tyebally S, Ghose A, Chen DH, Abiodun AT, Ghosh AK. Chest Pain in the Cancer Patient. Eur Cardiol 2022; 17:e15. [PMID: 35702571 PMCID: PMC9185574 DOI: 10.15420/ecr.2021.45] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 01/10/2022] [Indexed: 11/30/2022] Open
Abstract
Chest pain is one of the most common presenting symptoms in patients seeking care from a physician. Risk assessment tools and scores have facilitated prompt diagnosis and optimal management in these patients; however, it is unclear as to whether a standardised approach can adequately triage chest pain in cancer patients and survivors. This is of concern because cancer patients are often at an increased risk of cardiovascular mortality and morbidity given the shared risk factors between cancer and cardiovascular disease, compounded by the fact that certain anti-cancer therapies are associated with an increased risk of cardiovascular events that can persist for weeks and even years after treatment. This article describes the underlying mechanisms of the most common causes of chest pain in cancer patients with an emphasis on how their management may differ to that of non-cancer patients with chest pain. It will also highlight the role of the cardio-oncology team, who can aid in identifying cancer therapy-related cardiovascular side-effects and provide optimal multidisciplinary care for these patients.
Collapse
Affiliation(s)
- Sara Tyebally
- Cardio-Oncology Service, Barts Heart Centre, St Bartholomew’s Hospital, London, UK
| | - Aruni Ghose
- Oncology Department, St Bartholomew’s Hospital, London, UK
| | - Daniel H Chen
- Cardio-Oncology Service, Barts Heart Centre, St Bartholomew’s Hospital, London, UK; Hatter Cardiovascular Institute, UCL Institute of Cardiovascular Science, University College London Hospital, London, UK
| | - Aderonke T Abiodun
- Cardio-Oncology Service, Barts Heart Centre, St Bartholomew’s Hospital, London, UK
| | - Arjun K Ghosh
- Cardio-Oncology Service, Barts Heart Centre, St Bartholomew’s Hospital, London, UK; Hatter Cardiovascular Institute, UCL Institute of Cardiovascular Science, University College London Hospital, London, UK
| |
Collapse
|
53
|
Harada Y, Shimada K, Kubota Y, Yamashita M. Effectiveness of Iodine-123 β-methyl-P-iodophenyl-pentadecanoic acid (BMIPP) Myocardial Scintigraphy for Cancer Therapeutics-Related Cardiac Dysfunction (CTRCD) in Breast Cancer Patients. Cureus 2022; 14:e25524. [PMID: 35800804 PMCID: PMC9245443 DOI: 10.7759/cureus.25524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2022] [Indexed: 11/05/2022] Open
|
54
|
Eberhardt RT, Bonaca MP, Abu Daya H, Garcia LA, Gupta K, Mena-Hurtado C, Rogers RK, Sethi SS, Young MN, Piazza G. Call for Formalized Pathways in Vascular Medicine Training: JACC Review Topic of the Week. J Am Coll Cardiol 2022; 79:2129-2139. [PMID: 35618351 DOI: 10.1016/j.jacc.2022.03.365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/10/2022] [Accepted: 03/21/2022] [Indexed: 11/28/2022]
Abstract
The burden of vascular diseases and complexity of their management have been growing. Vascular medicine specialists may help to bridge gaps in care, especially as part of multidisciplinary teams. However, there is a limited number of vascular medicine specialists because of constraints in training. Despite established pathways for training in vascular medicine, there are obstacles that restrict completion of training in dedicated programs. A key factor is lack of funding as a result of inadequate recognition by key national accrediting and credentialing organizations. A concerted effort is required to overcome the obstacles to expand vascular medicine training programs and ultimately the pool of vascular medicine specialists. Well-trained vascular medicine specialists will be well positioned to ease the burden of vascular disease and optimize patient outcomes.
Collapse
Affiliation(s)
- Robert T Eberhardt
- Division of Cardiovascular Medicine, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts, USA.
| | - Marc P Bonaca
- Department of Medicine-Cardiology, University of Colorado School of Medicine, Aurora, Colorado, USA. https://twitter.com/MarcBonaca
| | - Hussein Abu Daya
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, USA. https://twitter.com/Dr_AbuDaya
| | - Lawrence A Garcia
- Division of Cardiovascular Medicine, Department of Medicine, Tufts University School of Medicine, St Elizabeth's Medical Center, Boston, Massachusetts, USA
| | - Kamal Gupta
- Department of Cardiovascular Medicine, University of Kansas School of Medicine, Kansas City, Kansas, USA
| | - Carlos Mena-Hurtado
- Vascular Medicine Outcomes Program, Division of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - R Kevin Rogers
- Department of Medicine-Cardiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Sanjum S Sethi
- Columbia Interventional Cardiovascular Care, Division of Cardiology, Columbia University Irving Medical School/New York-Presbyterian Hospital, New York, New York, USA. https://twitter.com/sanjum
| | - Michael N Young
- Cardiology Division, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA. https://twitter.com/mnyoung1
| | - Gregory Piazza
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
55
|
Szczepaniak P, Siedlinski M, Hodorowicz-Zaniewska D, Nosalski R, Mikolajczyk TP, Dobosz AM, Dikalova A, Dikalov S, Streb J, Gara K, Basta P, Krolczyk J, Sulicka-Grodzicka J, Jozefczuk E, Dziewulska A, Saju B, Laksa I, Chen W, Dormer J, Tomaszewski M, Maffia P, Czesnikiewicz-Guzik M, Crea F, Dobrzyn A, Moslehi J, Grodzicki T, Harrison DG, Guzik TJ. Breast cancer chemotherapy induces vascular dysfunction and hypertension through NOX4 dependent mechanism. J Clin Invest 2022; 132:149117. [PMID: 35617030 PMCID: PMC9246378 DOI: 10.1172/jci149117] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/19/2022] [Indexed: 11/17/2022] Open
Abstract
Cardiovascular disease is the major cause of morbidity and mortality in breast cancer survivors. Chemotherapy contributes to this risk. We aimed to define the mechanisms of long-term vascular dysfunction caused by neoadjuvant chemotherapy (NACT) and identify novel therapeutic targets.We studied arteries from postmenopausal women who had undergone breast cancer treatment using docetaxel, doxorubicin and cyclophosphamide (NACT), and women with no history of such treatment matched for key clinical parameters. Mechanisms were explored in wild-type and Nox4-/- mice and human microvascular endothelial cells.Endothelium-dependent vasodilatation is severely impaired in patients after NACT, while endothelium-independent responses remain normal. This was mimicked by 24-hour exposure of arteries to NACT agents ex-vivo. When applied individually, only docetaxel impaired endothelial function in human vessels. Mechanistic studies showed that NACT increased inhibitory eNOS phosphorylation of threonine 495 in a ROCK-dependent manner and augmented vascular superoxide and hydrogen peroxide production and NADPH oxidase activity. Docetaxel increased expression of NADPH oxidase NOX4 in endothelial and smooth muscle cells and NOX2 in the endothelium. NOX4 increase in human arteries may be mediated epigenetically by diminished DNA methylation of the NOX4 promoter. Docetaxel induced endothelial dysfunction and hypertension in mice. These were prevented in Nox4-/- and by pharmacological inhibition of Nox4 or Rock.Commonly used chemotherapeutic agents, and in particular, docetaxel, alter vascular function by promoting inhibitory phosphorylation of eNOS and enhancing ROS production by NADPH oxidases.
Collapse
Affiliation(s)
- Piotr Szczepaniak
- Department of Medicine, Collegium Medicum, Jagiellonian University, Krakow, Poland
| | - Mateusz Siedlinski
- Department of Medicine, Collegium Medicum, Jagiellonian University, Krakow, Poland
| | | | - Ryszard Nosalski
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Tomasz P Mikolajczyk
- Department of Medicine, Collegium Medicum, Jagiellonian University, Krakow, Poland
| | - Aneta M Dobosz
- Laboratory of Cell Signaling and Metabolic Disorders, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Anna Dikalova
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University, Nashville, United States of America
| | - Sergey Dikalov
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University, Nashville, United States of America
| | - Joanna Streb
- Department of Oncology, Collegium Medicum, Jagiellonian University, Krakow, Poland
| | - Katarzyna Gara
- Department of Surgery, Collegium Medicum, Jagiellonian University, Krakow, Poland
| | - Pawel Basta
- Department of Gynecology and Gynecological Oncology, Collegium Medicum, Jagiellonian University, Krakow, Poland
| | - Jaroslaw Krolczyk
- Department of Internal Medicine and Gerontology, Collegium Medicum, Jagiellonian University, Krakow, Poland
| | | | - Ewelina Jozefczuk
- Department of Medicine, Collegium Medicum, Jagiellonian University, Krakow, Poland
| | - Anna Dziewulska
- Laboratory of Cell Signaling and Metabolic Disorders, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Blessy Saju
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Iwona Laksa
- Department of Oncology, Collegium Medicum, Jagiellonian University, Krakow, Poland
| | - Wei Chen
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University, Nashville, United States of America
| | - John Dormer
- Department of Cellular Pathology, University Hospitals of Leicester, Leicester, United Kingdom
| | - Maciej Tomaszewski
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| | - Pasquale Maffia
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Marta Czesnikiewicz-Guzik
- Department of Periodontology and Oral Sciences Research Group, University of Glasgow, Glasgow, United Kingdom
| | - Filippo Crea
- Department of Cardiovascular and Thoracic Sciences, University of the Sacred Heart, Rome, Italy
| | - Agnieszka Dobrzyn
- Laboratory of Cell Signaling and Metabolic Disorders, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Javid Moslehi
- University of California San Fransisco, San Francisco, United States of America
| | - Tomasz Grodzicki
- Department of Internal Medicine and Gerontology, Collegium Medicum, Jagiellonian University, Krakow, Poland
| | - David G Harrison
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University, Nashville, United States of America
| | - Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
56
|
Quintanilha JCF, Etheridge AS, Graynor BJ, Larson NB, Crona DJ, Mitchell BD, Innocenti F. Polygenic Risk Scores for Blood Pressure to Assess the Risk of Severe Bevacizumab-Induced Hypertension in Cancer Patients (Alliance). Clin Pharmacol Ther 2022; 112:364-371. [PMID: 35527502 PMCID: PMC9296545 DOI: 10.1002/cpt.2635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 05/01/2022] [Indexed: 11/10/2022]
Abstract
Hypertension is a common bevacizumab-induced toxicity. No markers are available to predict patients at risk of developing hypertension. We hypothesized that genetic risk of essential hypertension, as measured by a blood pressure polygenic risk score (PRS), would be associated with risk of severe bevacizumab-induced hypertension. PRSs were calculated for 1,027 bevacizumab-treated patients of European descent with cancer from four clinical trials (Alliance for Clinical Trials in Oncology (Alliance) / Cancer and Leukemia Group B (CALGB) 80303, 40503, 90401, 40502) using summary systolic blood pressure (SBP) and diastolic blood pressure (DBP) genome-wide association results obtained from 757,601 individuals of European descent. The association between PRS and grade 3 bevacizumab-induced hypertension (Common Toxicity Criteria for Adverse Events version 3) in each trial was performed by multivariable logistic regression. Fixed-effect meta-analyses odds ratios (ORs) per standard deviation (SD) of the association of PRS (quantitative) and hypertension across trials were estimated by inverse-variance weighting. PRSs were additionally stratified into quintiles, with the bottom quintile as the referent group. The OR of the association between hypertension and each quintile vs. the referent group was determined by logistic regression. The most significant PRS (quantitative)-hypertension association included up to 67 single-nucleotide variants (SNPs) associated with SBP (P = 0.0077, OR per SD = 1.31, 95% confidence interval (CI), 1.07-1.60), and up to 53 SNPs associated with DBP (P = 0.0209, OR per SD = 1.27, 95% CI, 1.04-1.56). Patients in the top quintile had a higher risk of developing bevacizumab-induced hypertension compared with patients in the bottom quintile using SNPs associated with SBP (P = 4.75 × 10-4 , OR = 3.72, 95% CI, 1.84-8.16) and DBP (P = 0.076, OR = 1.83, 95% CI, 0.95-3.64). Genetic variants associated with essential hypertension, mainly SBP, increase the risk of severe bevacizumab-induced hypertension.
Collapse
Affiliation(s)
- Julia C F Quintanilha
- University of North Carolina Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Amy S Etheridge
- University of North Carolina Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Brady J Graynor
- School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Nicholas B Larson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Daniel J Crona
- University of North Carolina Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | | | | |
Collapse
|
57
|
Cardio-onco-metabolism: metabolic remodelling in cardiovascular disease and cancer. Nat Rev Cardiol 2022; 19:414-425. [PMID: 35440740 PMCID: PMC10112835 DOI: 10.1038/s41569-022-00698-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/17/2022] [Indexed: 02/07/2023]
Abstract
Cardiovascular disease and cancer are the two leading causes of morbidity and mortality in the world. The emerging field of cardio-oncology has revealed that these seemingly disparate disease processes are intertwined, owing to the cardiovascular sequelae of anticancer therapies, shared risk factors that predispose individuals to both cardiovascular disease and cancer, as well the possible potentiation of cancer growth by cardiac dysfunction. As a result, interest has increased in understanding the fundamental biological mechanisms that are central to the relationship between cardiovascular disease and cancer. Metabolism, appropriate regulation of energy, energy substrate utilization, and macromolecular synthesis and breakdown are fundamental processes for cellular and organismal survival. In this Review, we explore the emerging data identifying metabolic dysregulation as an important theme in cardio-oncology. We discuss the growing recognition of metabolic reprogramming in cardiovascular disease and cancer and view the novel area of cardio-oncology through the lens of metabolism.
Collapse
|
58
|
Jung MH, Yi SW, An SJ, Yi JJ, Ihm SH, Han S, Ryu KH, Jung HO, Youn HJ. Associations between the triglyceride-glucose index and cardiovascular disease in over 150,000 cancer survivors: a population-based cohort study. Cardiovasc Diabetol 2022; 21:52. [PMID: 35429972 PMCID: PMC9013459 DOI: 10.1186/s12933-022-01490-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 04/04/2022] [Indexed: 01/04/2023] Open
Abstract
Background The prevention of subsequent cardiovascular disease (CVD) is an essential part of cancer survivorship care. We conducted the present study to investigate the association between the TyG index (a surrogate marker of insulin resistance) and the risk of cardiovascular disease (CVD) events in cancer survivors. Methods Adult cancer patients, who underwent routine health examinations during 2009–2010 and were survived for more than 5 years as of January 1, 2011, were followed for hospitalization of CVD (either ischemic heart disease, stroke, or heart failure) until December 2020. Cox model was used to calculate hazard ratios associated with baseline TyG index (loge [fasting triglyceride (mg) × fasting glucose (mg)/2]) for the CVD hospitalization. Results A total of 155,167 cancer survivors (mean age 59.9 ± 12.0 years, female 59.1%) were included in this study. A graded positive association was observed between TyG and CVD hospitalization. An 8% elevated risk for CVD hospitalization was observed for a TyG index of 8-8.4 (aHR 1.08 [95% CI 1.01–1.14]); 10% elevated risk for a TyG index of 8.5–8.9 (aHR 1.10 [95% CI 1.03–1.17]); 23% elevated risk for a TyG index of 9.0-9.4 (aHR 1.23 [95% CI 1.15–1.31]); 34% elevated risk for a TyG index of 9.5–9.9 (aHR 1.34 [95% CI 1.23–1.47]); and 55% elevated risk for a TyG index ≥ 10 compared to the reference group (TyG index < 8). Per 1-unit increase in the TyG index, a 16% increase in CVD hospitalization and a 45% increase in acute myocardial infarction hospitalization were demonstrated. Graded positive associations were evident for atherosclerotic CVD subtypes, such as ischemic heart disease, acute myocardial infarction, and ischemic stroke, but not for hemorrhagic stroke or heart failure. Conclusions The TyG index may serve as a simple surrogate marker for the risk stratification of future CVD events, particularly atherosclerotic subtypes, in cancer survivors. Supplementary Information The online version contains supplementary material available at 10.1186/s12933-022-01490-z.
Collapse
|
59
|
Nakayama T, Oshima Y, Shintani Y, Yamamoto J, Yokoi M, Ito T, Wakami K, Kitada S, Goto T, Hashimoto H, Kusumoto S, Sugiura T, Iida S, Seo Y. Ventricular Sigmoid Septum as a Risk Factor for Anthracycline-Induced Cancer Therapeutics-Related Cardiac Dysfunction in Patients With Malignant Lymphoma. Circ Rep 2022; 4:173-182. [PMID: 35434414 PMCID: PMC8977195 DOI: 10.1253/circrep.cr-21-0145] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 02/26/2022] [Accepted: 03/06/2022] [Indexed: 11/28/2022] Open
Abstract
Background: Identifying risk factors for cancer therapeutics-related cardiac dysfunction (CTRCD) is essential for the early detection and prompt initiation of medial therapy for CTRCD. No study has investigated whether the sigmoid septum is a risk factor for anthracycline-induced CTRCD. Methods and Results: We enrolled 167 patients with malignant lymphoma who received a CHOP-like regimen from January 2008 to December 2017 and underwent both baseline and follow-up echocardiography. Patients with left ventricular ejection fraction (LVEF) ≤50% were excluded. CTRCD was defined as a ≥10% decline in LVEF and LVEF <50% after chemotherapy. The angle between the anterior wall of the aorta and the ventricular septal surface (ASA) was measured to quantify the sigmoid septum. CTRCD was observed in 36 patients (22%). Mean LVEF and global longitudinal strain (GLS) were lower, left ventricular mass index was higher, and ASA was smaller in patients with CTRCD. In a multivariable Cox proportional hazard analysis, GLS (hazard ratio [HR] per 1% decrease 1.20; 95% confidence interval [CI] 1.07-1.35) and ASA (HR per 1° increase 0.97; 95% CI 0.95-0.99) were identified as independent determinants of CTRCD. An integrated discrimination improvement evaluation confirmed the significant incremental value of ASA for developing CTRCD. Conclusions: Smaller ASA was an independent risk factor and had significant incremental value for CTRCD in patients with malignant lymphoma who received the CHOP-like regimen.
Collapse
Affiliation(s)
- Takafumi Nakayama
- Department of Cardiology, Nagoya City University Graduate School of Medical Sciences Nagoya Japan
| | - Yoshiko Oshima
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences Nagoya Japan
| | - Yasuhiro Shintani
- Department of Cardiology, Nagoya City University Graduate School of Medical Sciences Nagoya Japan
| | - Junki Yamamoto
- Department of Cardiology, Nagoya City University Graduate School of Medical Sciences Nagoya Japan
| | - Masashi Yokoi
- Department of Cardiology, Nagoya City University Graduate School of Medical Sciences Nagoya Japan
| | - Tsuyoshi Ito
- Department of Cardiology, Nagoya City University Graduate School of Medical Sciences Nagoya Japan
| | - Kazuaki Wakami
- Department of Cardiology, Nagoya City University Graduate School of Medical Sciences Nagoya Japan
| | - Shuichi Kitada
- Department of Cardiology, Nagoya City University Graduate School of Medical Sciences Nagoya Japan
| | - Toshihiko Goto
- Department of Cardiology, Nagoya City University Graduate School of Medical Sciences Nagoya Japan
| | - Hiroya Hashimoto
- Clinical Research Management Center, Nagoya City University Hospital Nagoya Japan
| | - Shigeru Kusumoto
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences Nagoya Japan
| | - Tomonori Sugiura
- Department of Cardiology, Nagoya City University Graduate School of Medical Sciences Nagoya Japan
| | - Shinsuke Iida
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences Nagoya Japan
| | - Yoshihiro Seo
- Department of Cardiology, Nagoya City University Graduate School of Medical Sciences Nagoya Japan
| |
Collapse
|
60
|
Di Lisi D, Madaudo C, Alagna G, Santoro M, Rossetto L, Siragusa S, Novo G. The new HFA/ICOS risk assessment tool to identify patients with chronic myeloid leukaemia at high risk of cardiotoxicity. ESC Heart Fail 2022; 9:1914-1919. [PMID: 35355425 PMCID: PMC9065844 DOI: 10.1002/ehf2.13897] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 02/06/2022] [Accepted: 03/02/2022] [Indexed: 11/10/2022] Open
Abstract
AIMS Tyrosine kinase inhibitors (TKIs) used to treat chronic myeloid leukaemia (CML) can cause cardiovascular adverse events. So far, the Systematic Coronary Risk Evaluation (SCORE) charts of the European Society of Cardiology (ESC) have been used to identify cancer patients at increased cardiovascular risk. The primary aim of our study was to evaluate the usefulness of the new cardiovascular risk assessment model proposed by the Cardio-Oncology Study Group of the Heart Failure Association (HFA) of the ESC in collaboration with the International Cardio-Oncology Society (ICOS) to stratify the cardiovascular risk in CML patients, compared with SCORE risk charts. The secondary aim was to establish the incidence of adverse arterial events (AEs) in patients with CML treated with TKIs and the influence of preventive treatment with aspirin. METHODS AND RESULTS A retrospective single-centre observational study was carried out on 58 patients (32 men and 26 women; mean age ± SD: 59 ± 15 years) with CML treated with TKIs for a median period of 43 ± 31 months. Cardiological evaluation was performed and cardiovascular risk was estimated with SCORE risk charts and with the new risk assessment tool proposed by HFA/ICOS. AEs were recorded. According to SCORE charts and the new HFA/ICOS risk stratification tool, respectively, 46% (Group A1) and 60% (Group A2) of patients were at high-very high risk, and 54% (Group B1) and 40% (Group B2) at low-moderate risk. AEs were significantly more frequent in Group A1 than Group B1 (P value < 0.01) when considered overall; they were significantly more frequent in Group A2 than Group B2 either overall or considered individually. HFA/ICOS risk stratification tool was significantly more sensitive than SCORE (P < 0.01) in identifying patients at higher risk of cardiovascular toxicity. In addition, we did not find AEs in patients pretreated with aspirin. CONCLUSIONS The new HFA/ICOS risk stratification model allows a more tailored cardiovascular risk stratification in patients with CML and it is more sensitive than SCORE charts.
Collapse
Affiliation(s)
- Daniela Di Lisi
- Cardiology Unit, University Hospital Paolo Giaccone, Palermo, Italy.,Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Cristina Madaudo
- Cardiology Unit, University Hospital Paolo Giaccone, Palermo, Italy.,Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Giulia Alagna
- Cardiology Unit, University Hospital Paolo Giaccone, Palermo, Italy.,Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Marco Santoro
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy.,Haematology Unit, University Hospital Paolo Giaccone, Palermo, Italy
| | - Ludovico Rossetto
- Cardiology Unit, University Hospital Paolo Giaccone, Palermo, Italy.,Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Sergio Siragusa
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy.,Haematology Unit, University Hospital Paolo Giaccone, Palermo, Italy
| | - Giuseppina Novo
- Cardiology Unit, University Hospital Paolo Giaccone, Palermo, Italy.,Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| |
Collapse
|
61
|
Kersting D, Settelmeier S, Mavroeidi IA, Herrmann K, Seifert R, Rischpler C. Shining Damaged Hearts: Immunotherapy-Related Cardiotoxicity in the Spotlight of Nuclear Cardiology. Int J Mol Sci 2022; 23:3802. [PMID: 35409161 PMCID: PMC8998973 DOI: 10.3390/ijms23073802] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/20/2022] [Accepted: 03/25/2022] [Indexed: 11/30/2022] Open
Abstract
The emerging use of immunotherapies in cancer treatment increases the risk of immunotherapy-related cardiotoxicity. In contrast to conventional chemotherapy, these novel therapies have expanded the forms and presentations of cardiovascular damage to a broad spectrum from asymptomatic changes to fulminant short- and long-term complications in terms of cardiomyopathy, arrythmia, and vascular disease. In cancer patients and, particularly, cancer patients undergoing (immune-)therapy, cardio-oncological monitoring is a complex interplay between pretherapeutic risk assessment, identification of impending cardiotoxicity, and post-therapeutic surveillance. For these purposes, the cardio-oncologist can revert to a broad spectrum of nuclear cardiological diagnostic workup. The most promising commonly used nuclear medicine imaging techniques in relation to immunotherapy will be discussed in this review article with a special focus on the continuous development of highly specific molecular markers and steadily improving methods of image generation. The review closes with an outlook on possible new developments of molecular imaging and advanced image evaluation techniques in this exciting and increasingly growing field of immunotherapy-related cardiotoxicity.
Collapse
Affiliation(s)
- David Kersting
- Department of Nuclear Medicine, University Hospital Essen, West German Cancer Center (WTZ), University of Duisburg-Essen, 45147 Essen, Germany; (K.H.); (R.S.); (C.R.)
- German Cancer Consortium (DKTK, Partner Site Essen/Düsseldorf), 45147 Essen, Germany;
| | - Stephan Settelmeier
- Department of Cardiology and Vascular Medicine, University Hospital Essen, West German Heart and Vascular Center, University of Duisburg-Essen, 45147 Essen, Germany;
| | - Ilektra-Antonia Mavroeidi
- German Cancer Consortium (DKTK, Partner Site Essen/Düsseldorf), 45147 Essen, Germany;
- Clinic for Internal Medicine (Tumor Research), University Hospital Essen, West German Cancer Center (WTZ), University of Duisburg-Essen, 45147 Essen, Germany
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Essen, West German Cancer Center (WTZ), University of Duisburg-Essen, 45147 Essen, Germany; (K.H.); (R.S.); (C.R.)
- German Cancer Consortium (DKTK, Partner Site Essen/Düsseldorf), 45147 Essen, Germany;
| | - Robert Seifert
- Department of Nuclear Medicine, University Hospital Essen, West German Cancer Center (WTZ), University of Duisburg-Essen, 45147 Essen, Germany; (K.H.); (R.S.); (C.R.)
- German Cancer Consortium (DKTK, Partner Site Essen/Düsseldorf), 45147 Essen, Germany;
| | - Christoph Rischpler
- Department of Nuclear Medicine, University Hospital Essen, West German Cancer Center (WTZ), University of Duisburg-Essen, 45147 Essen, Germany; (K.H.); (R.S.); (C.R.)
- German Cancer Consortium (DKTK, Partner Site Essen/Düsseldorf), 45147 Essen, Germany;
| |
Collapse
|
62
|
Meilhac A, Cautela J, Thuny F. Cancer Therapies and Vascular Toxicities. Curr Treat Options Oncol 2022; 23:333-347. [PMID: 35244888 DOI: 10.1007/s11864-022-00964-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2022] [Indexed: 12/24/2022]
Abstract
OPINION STATEMENT Vascular events have become an important issue in the overall management of cancer patients. They usually result from a combination of (i) direct or indirect toxicity of anticancer treatments, (ii) a higher prevalence of cardiovascular risk factors in cancer patients, and (iii) prolonged exposure to treatments due to an increasing patient survival rate. In addition to conventional chemotherapies and radiotherapy, targeted therapies and immunotherapies have been developed which improve the prognosis of cancer patients but sometimes at the cost of vascular toxicity, which can lead to systemic or pulmonary hypertension and arterial/venous thromboembolic events. Endothelial dysfunction, a procoagulant state and metabolic disorders are the three main pathophysiological patterns leading to cancer treatment-related vascular toxicity. This issue is challenging because serious vascular adverse events can necessitate cancer treatment being put on hold or stopped, which could compromise patient survival. In addition to increasing the risk of thrombotic adverse events, cancer therapies may lead to an increased risk of bleeding, especially in treatments with vascular endothelial growth factor inhibitors. Therefore, we can define vasculo-oncology as a part of the cardio-oncology specialty; its aims are to predict, prevent, screen, and treat vascular toxicity related to cancer treatments. While the level of evidence is low regarding the management of vascular toxicity during cancer therapy, cardiologists and specialists in vascular diseases should closely collaborate with oncologists and hematologists to determine the optimal strategy for each patient.
Collapse
Affiliation(s)
- Alexandra Meilhac
- Department of Cardiology, North Hospital, Assistance Publique - Hôpitaux de Marseille, Centre for CardioVascular and Nutrition Research (C2VN), University Mediterranean Center of Cardio-Oncology, Unit of Heart Failure and Valvular Heart Diseases, Inserm 1263, Inrae 1260, Aix-Marseille University, Chemin des Bourrely, 13015, Marseille, France
| | - Jennifer Cautela
- Department of Cardiology, North Hospital, Assistance Publique - Hôpitaux de Marseille, Centre for CardioVascular and Nutrition Research (C2VN), University Mediterranean Center of Cardio-Oncology, Unit of Heart Failure and Valvular Heart Diseases, Inserm 1263, Inrae 1260, Aix-Marseille University, Chemin des Bourrely, 13015, Marseille, France
| | - Franck Thuny
- Department of Cardiology, North Hospital, Assistance Publique - Hôpitaux de Marseille, Centre for CardioVascular and Nutrition Research (C2VN), University Mediterranean Center of Cardio-Oncology, Unit of Heart Failure and Valvular Heart Diseases, Inserm 1263, Inrae 1260, Aix-Marseille University, Chemin des Bourrely, 13015, Marseille, France.
| |
Collapse
|
63
|
Martinez DSL, Noseworthy PA, Akbilgic O, Herrmann J, Ruddy KJ, Hamid A, Maddula R, Singh A, Davis R, Gunturkun F, Jefferies JL, Brown SA. Artificial intelligence opportunities in cardio-oncology: Overview with spotlight on electrocardiography. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2022; 15:100129. [PMID: 35721662 PMCID: PMC9202996 DOI: 10.1016/j.ahjo.2022.100129] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/20/2022] [Accepted: 03/21/2022] [Indexed: 01/21/2023]
Abstract
Cardiovascular disease is a leading cause of death among cancer survivors, second only to cancer recurrence or development of new tumors. Cardio-oncology has therefore emerged as a relatively new specialty focused on prevention and management of cardiovascular consequences of cancer therapies. Yet challenges remain regarding precision and accuracy with predicting individuals at highest risk for cardiotoxicity. Barriers such as access to care also limit screening and early diagnosis to improve prognosis. Thus, developing innovative approaches for prediction and early detection of cardiovascular illness in this population is critical. In this review, we provide an overview of the present state of machine learning applications in cardio-oncology. We begin by outlining some factors that should be considered while utilizing machine learning algorithms. We then examine research in which machine learning has been applied to improve prediction of cardiac dysfunction in cancer survivors. We also highlight the use of artificial intelligence (AI) in conjunction with electrocardiogram (ECG) to predict cardiac malfunction and also atrial fibrillation (AF), and we discuss the potential role of wearables. Additionally, the article summarizes future prospects and critical takeaways for the application of machine learning in cardio-oncology. This study is the first in a series on artificial intelligence in cardio-oncology, and complements our manuscript on echocardiography and other forms of imaging relevant to cancer survivors cared for in cardiology clinical practice.
Collapse
Affiliation(s)
- Daniel Sierra-Lara Martinez
- Coronary Care Unit, National Institute of Cardiology/Instituto Nacional de Cardiologia, Ciudad de Mexico, Mexico
| | | | - Oguz Akbilgic
- Department of Health Informatics and Data Science, Parkinson School of Health Sciences and Public Health, Loyola University Chicago, Maywood, IL, USA
- Section of Cardiovascular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Wake Forest, NC, USA
| | - Joerg Herrmann
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Ashima Singh
- Institute of Health and Equity, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Robert Davis
- Center for Biomedical Informatics, University of Tennessee Health Sciences Center, USA
| | - Fatma Gunturkun
- Center for Biomedical Informatics, University of Tennessee Health Sciences Center, USA
| | - John L. Jefferies
- Division of Cardiovascular Diseases, University of Tennessee Health Sciences Center, USA
- Department of Epidemiology, St. Jude Children's Research Hospital, USA
| | - Sherry-Ann Brown
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
64
|
Paterson DI, Wiebe N, Cheung WY, Mackey JR, Pituskin E, Reiman A, Tonelli M. Incident Cardiovascular Disease Among Adults With Cancer: A Population-Based Cohort Study. JACC CardioOncol 2022; 4:85-94. [PMID: 35492824 PMCID: PMC9040097 DOI: 10.1016/j.jaccao.2022.01.100] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 12/13/2022] Open
Abstract
Background Patients with cancer and cancer survivors are at increased risk for incident heart failure, but there are conflicting data on the long-term risk for other cardiovascular events and how such risk may vary by cancer site. Objectives The aim of this study was to determine the impact of a new cancer diagnosis on the risk for fatal and nonfatal cardiovascular events. Methods Using administrative health care databases, a population-based retrospective cohort study was conducted among 4,519,243 adults residing in Alberta, Canada, from April 2007 to December 2018. Participants with new cancer diagnoses during the study period were compared with those without cancer with respect to risk for subsequent cardiovascular events (cardiovascular mortality, myocardial infarction, stroke, heart failure, and pulmonary embolism) using time-to-event survival models after adjusting for sociodemographic data and comorbidities. Results A total of 224,016 participants with new cancer diagnoses were identified, as well as 73,360 cardiovascular deaths and 470,481 nonfatal cardiovascular events during a median follow-up period of 11.8 years. After adjustment, participants with cancer had HRs of 1.33 (95% CI: 1.29-1.37) for cardiovascular mortality, 1.01 (95% CI: 0.97-1.05) for myocardial infarction, 1.44 (95% CI: 1.41-1.47) for stroke, 1.62 (95% CI: 1.59-1.65) for heart failure, and 3.43 (95% CI: 3.37-3.50) for pulmonary embolism, compared with participants without cancer. Cardiovascular risk was highest for patients with genitourinary, gastrointestinal, thoracic, nervous system and hematologic malignancies. Conclusions A new cancer diagnosis is independently associated with a significantly increased risk for cardiovascular death and nonfatal morbidity regardless of cancer site. These findings highlight the need for a collaborative approach to health care for patients with cancer and cancer survivors.
Collapse
Affiliation(s)
- D. Ian Paterson
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Natasha Wiebe
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Winson Y. Cheung
- Department of Oncology, University of Calgary, Calgary, Alberta, Canada
| | - John R. Mackey
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Edith Pituskin
- Faculty of Nursing, University of Alberta, Edmonton, Alberta, Canada
| | - Anthony Reiman
- Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Marcello Tonelli
- Department of Medicine, University of Calgary, Calgary, Alberta, Canada
- Address for correspondence: Dr Marcello Tonelli, University of Calgary, 7th Floor, TRW Building, 3280 Hospital Drive NW, Calgary, Alberta T2N 4Z6, Canada. @cellotonelli
| |
Collapse
|
65
|
Ba Z, Xiao Y, He M, Liu D, Wang H, Liang H, Yuan J. Risk Factors for the Comorbidity of Hypertension and Renal Cell Carcinoma in the Cardio-Oncologic Era and Treatment for Tumor-Induced Hypertension. Front Cardiovasc Med 2022; 9:810262. [PMID: 35252390 PMCID: PMC8892205 DOI: 10.3389/fcvm.2022.810262] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022] Open
Abstract
Advances in tumor diagnosis and treatment, especially the use of targeted therapies, have remarkably improved the survival rate of patients with renal cell carcinoma (RCC), accompanied by higher hypertension (HTN) incidence among patients with RCC, reflecting the coming of a cardio-oncologic era. Therefore, for patients with RCC and HTN simultaneously, finding risk factors for the comorbidity and giving better clinical treatment have been urgent problems. In this review, we thoroughly investigated risk factors for the comorbidity of HTN and RCC based on preclinical and clinical studies. Firstly, RCC and HTN may have common risk factors, such as obesity, smoking, and other modifiable lifestyles. Secondly, RCC and HTN may lead to each other directly or indirectly by their therapies. We then discussed measures of reducing the comorbidity and treatment of HTN in patients with RCC. We also discussed the deficiency of current studies and pointed out future directions. In conclusion, this review aims to deepen the understanding of cardio-oncology and bring benefit to the population who are at high risk of getting or have already got RCC and HTN simultaneously.
Collapse
Affiliation(s)
- Zhengqing Ba
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying Xiao
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Ming He
- Department of Infectious Diseases, Peking University First Hospital, Beijing, China
| | - Dong Liu
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hao Wang
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hanyang Liang
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiansong Yuan
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Pulmonary Vascular Medicine, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Jiansong Yuan
| |
Collapse
|
66
|
Preexisting Cardiovascular Risk Factors and Coronary Artery Atherosclerosis in Patients with and without Cancer. Cardiol Res Pract 2022; 2022:4570926. [PMID: 35154824 PMCID: PMC8826118 DOI: 10.1155/2022/4570926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 01/07/2022] [Indexed: 12/24/2022] Open
Abstract
Cancer survivors suffer a higher risk of coronary artery atherosclerosis (CAA). Whether cancer patients had increased baseline CAA burden prior to cardiotoxic therapy remains unclear. We conducted a case-control study, and 286 consecutive patients were finally included. Among these patients, 181 had newly diagnosed cancer and 105 had nonmalignant diseases. Cancer was confirmed by biopsy. The severity of CAA was determined by coronary angiography and evaluated using the percentage of stenosis or Gensini scoring (GS). Patients with cancer versus cancer-free controls were older (OR = 1.052, 95% CI: 1.021–1.084, p < 0.001), more commonly male (OR = 0.048, 95% CI: 1.004–2.676, p=0.048), and more severely exposed to smoking (OR = 1.020, 95% CI: 1.007–1.033, p=0.003). Cancer patients were significantly more commonly complicated by ≥90% coronary stenosis than the gender- and age-matched cancer-free controls (9/93 versus 1/93, OR = 4.875, 95% CI: 1.024–23.213, p=0.047). After adjustment for age, gender, hypertension, diabetes, smoking history, blood glucose, and total cholesterol, cancer was significantly associated with high GS (adjusted OR = 2.208, 95% CI: 1.077–4.524, p=0.031). Our study demonstrated that cancer patients had increased CAA burden prior to cardiotoxic therapy. Further study is necessary to investigate the link between CAA and cancer.
Collapse
|
67
|
Osman M, Benjamin MM, Balla S, Kheiri B, Bianco C, Sengupta PP, Daggubati R, Malla M, Liu SV, Mamas M, Patel B. Index Admission and Thirty-Day Readmission Outcomes of Patients With Cancer Presenting With STEMI. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2022; 35:121-128. [PMID: 33888417 PMCID: PMC8521583 DOI: 10.1016/j.carrev.2021.04.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/30/2021] [Accepted: 04/12/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND National-level data of cancer patients' readmissions after ST-segment elevation myocardial infarction (STEMI) are lacking. OBJECTIVES The primary aim of this study was to compare the rates and causes of 30-day readmissions in patients with and without cancer. METHODS Among patients admitted with STEMI in the United States National Readmission Database (NRD) from October 2015-December 2017, we identified patients with the diagnosis of active breast, colorectal, lung, or prostate cancer. The primary endpoint was the 30-day unplanned readmission rate. Secondary endpoints included in-hospital outcomes during the index admission and causes of readmissions. A propensity score model was used to compare the outcomes of patients with and without cancer. RESULTS A total of 385,522 patients were included in the analysis: 5956 with cancer and 379,566 without cancer. After propensity score matching, 23,880 patients were compared (Cancer = 5949, No Cancer = 17,931). Patients with cancer had higher 30-day readmission rates (19% vs. 14%, p < 0.01). The most common causes for readmission among patients with cancer were cardiac (31%), infectious (21%), oncologic (17%), respiratory (4%), stroke (4%), and renal (3%). During the first readmission, patients with cancer had higher adjusted rates of in-hospital mortality (15% vs. 7%; p < 0.01) and bleeding complications (31% vs. 21%; p < 0.01), compared to the non-cancer group. In addition, cancer (OR 1.5, 95% CI 1.2-1.6, p < 0.01) was an independent predictor for 30-day readmission. CONCLUSIONS About one in five cancer patients presenting with STEMI will be readmitted within 30 days. Cardiac causes predominated the reason for 30-day readmissions in patients with cancer.
Collapse
Affiliation(s)
- Mohammed Osman
- Division of Cardiology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Mina M. Benjamin
- Division of Cardiology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Sudarshan Balla
- Division of Cardiology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Babikir Kheiri
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
| | - Christopher Bianco
- Division of Cardiology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Partho P. Sengupta
- Division of Cardiology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Ramesh Daggubati
- Division of Cardiology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Midhun Malla
- Division of Oncology, West Virginia University, School of Medicine, Morgantown, WV, USA
| | - Stephen V. Liu
- Division of Oncology, Medstar Georgetown University Hospital, Washington, DC, USA
| | - Mamas Mamas
- Keele Cardiovascular Research Group, Keele University, Keele, United Kingdom,Royal Stoke University Hospital, Stoke-on-Trent, United Kingdom
| | - Brijesh Patel
- Division of Cardiology, West Virginia University School of Medicine, Morgantown, WV, USA,Corresponding author at: WVU Heart & Vascular Institute, West Virginia University, One Medical Center Drive, Box 8003, Morgantown, WV 26506., USA.
| |
Collapse
|
68
|
Chintamani, Tandon M, Ghosh J. Breast Cancer with Associated Problems. Breast Cancer 2022. [DOI: 10.1007/978-981-16-4546-4_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
69
|
Van Daele M, Cooper SL, Pannucci P, Wragg ES, March J, de Jong I, Woolard J. Monitoring haemodynamic changes in rodent models to better inform safety pharmacology: Novel insights from in vivo studies and waveform analysis. JRSM Cardiovasc Dis 2022; 11:20480040221092893. [PMID: 35646334 PMCID: PMC9133998 DOI: 10.1177/20480040221092893] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/21/2022] [Accepted: 03/22/2022] [Indexed: 01/13/2023] Open
Abstract
Animal models are essential for assessing cardiovascular responses to novel therapeutics. Cardiovascular safety liabilities represent a leading cause of drug attrition and better preclinical measurements are essential to predict drug-related toxicities. Presently, radiotelemetric approaches recording blood pressure are routinely used in preclinical in vivo haemodynamic assessments, providing valuable information on therapy-associated cardiovascular effects. Nonetheless, this technique is chiefly limited to the monitoring of blood pressure and heart rate alone. Alongside these measurements, Doppler flowmetry can provide additional information on the vasculature by simultaneously measuring changes in blood flow in multiple different regional vascular beds. However, due to the time-consuming and expensive nature of this approach, it is not widely used in the industry. Currently, analysis of waveform data obtained from telemetry and Doppler flowmetry typically examines averages or peak values of waveforms. Subtle changes in the morphology and variability of physiological waveforms have previously been shown to be early markers of toxicity and pathology. Therefore, a detailed analysis of pressure and flowmetry waveforms could enhance the understanding of toxicological mechanisms and the ability to translate these preclinical observations to clinical outcomes. In this review, we give an overview of the different approaches to monitor the effects of drugs on cardiovascular parameters (particularly regional blood flow, heart rate and blood pressure) and suggest that further development of waveform analysis could enhance our understanding of safety pharmacology, providing valuable information without increasing the number of in vivo studies needed.
Collapse
Affiliation(s)
- Marieke Van Daele
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Midlands, UK
| | - Samantha L Cooper
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Midlands, UK
| | - Patrizia Pannucci
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Midlands, UK
| | - Edward S Wragg
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Midlands, UK
| | - Julie March
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| | - Iwan de Jong
- Maastricht Instruments BV, Maastricht University, Maastricht, The Netherlands
| | - Jeanette Woolard
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Midlands, UK
| |
Collapse
|
70
|
Ording AG, Søgaard M, Skjøth F, Grove EL, Lip GYH, Larsen TB, Nielsen PB. Hematuria in anticoagulated patients with atrial fibrillation and urologic cancer. Res Pract Thromb Haemost 2022; 6:e12629. [PMID: 35059549 PMCID: PMC8756162 DOI: 10.1002/rth2.12629] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/29/2021] [Accepted: 10/09/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Based on their renal excretion, direct oral anticoagulants (DOACs) may increase the risk of hematuria in patients with atrial fibrillation (AF) and urologic cancer compared with vitamin K antagonists. OBJECTIVES To examine the risk of bleeding associated with DOAC versus warfarin in patients with AF and urologic cancer. METHODS We conducted a Danish nationwide cohort study with individually linked registry data on patients with AF and active or a history of urologic cancer. We calculated crude rates per 100 person-years of hospital episodes of major bleeding and hematuria. We then compared rates of hematuria during the year after initial oral anticoagulation filled prescription by treatment regimen using inverse probability of treatment weighting and Cox regression. RESULTS The study population included 2615 patients with AF and urologic cancer (6.1% women; median age, 76 years) initiating a DOAC or warfarin. One-year risk of hematuria was 4.8% in the DOAC group and 4.7% in the warfarin group with a corresponding weighted hazard ratio (HR) of 1.21 (95% confidence interval [CI], 0.81-1.81). HRs for hematuria were generally similar in analyses restricted to patients treated with standard-dose DOAC and patients with active cancer. For those with cancer of the kidney, renal pelvis, ureter, and bladder, the HR was 0.82 (95% CI, 0.44-1.54). Results were mirrored for other bleeding events, whereas the risk for intracranial bleeding was lower with DOACs. CONCLUSION In patients with AF and urologic cancer, there was a similar risk of hematuria associated with DOAC and warfarin treatment.
Collapse
Affiliation(s)
- Anne Gulbech Ording
- Unit for Thrombosis and Drug ResearchDepartment of CardiologyAalborg University HospitalAalborgDenmark
| | - Mette Søgaard
- Unit for Thrombosis and Drug ResearchDepartment of CardiologyAalborg University HospitalAalborgDenmark
- Aalborg Thrombosis Research UnitAalborg UniversityAalborgDenmark
| | - Flemming Skjøth
- Aalborg Thrombosis Research UnitAalborg UniversityAalborgDenmark
- Unit of Clinical BiostatisticsAalborg University HospitalAalborgDenmark
| | - Erik Lerkevang Grove
- Department of CardiologyAarhus University HospitalAarhus NDenmark
- Department of Clinical MedicineFaculty of HealthAarhus UniversityAarhus NDenmark
| | - Gregory Y. H. Lip
- Unit for Thrombosis and Drug ResearchDepartment of CardiologyAalborg University HospitalAalborgDenmark
- Liverpool Centre for Cardiovascular SciencesUniversity Liverpool and Liverpool Heart & Chest HospitalLiverpoolUK
| | - Torben Bjerregaard Larsen
- Unit for Thrombosis and Drug ResearchDepartment of CardiologyAalborg University HospitalAalborgDenmark
- Aalborg Thrombosis Research UnitAalborg UniversityAalborgDenmark
| | - Peter Brønnum Nielsen
- Unit for Thrombosis and Drug ResearchDepartment of CardiologyAalborg University HospitalAalborgDenmark
- Aalborg Thrombosis Research UnitAalborg UniversityAalborgDenmark
| |
Collapse
|
71
|
Sim JE, Park J, Bang OY. Non-thrombotic ischemic cerebrovascular events related to the use of tyrosine kinase inhibitors in patients with cancer: high-resolution magnetic resonance imaging findings. PRECISION AND FUTURE MEDICINE 2021. [DOI: 10.23838/pfm.2021.00072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The targeted cancer therapies that have been introduced in recent years are directed against cancer-specific molecules and signaling pathways, and thus, they have limited nonspecific toxicities. However, major vascular events, including stroke, are common in patients receiving tyrosine kinase inhibitors (TKIs). Inhibition of vascular endothelial growth factor receptor by some TKIs probably explains such events. Moreover, venous and arterial thromboses, atherosclerosis, and bleeding have been reported. Ischemic lesions can also occur due to impaired angiogenesis or endothelial dysfunction. However, the exact mechanisms of arterial stroke in patients with cancer receiving TKIs are unknown. Here, we have report two cases of non-thrombotic ischemic cerebrovascular events related to TKIs and described the high-resolution magnetic resonance imaging findings.
Collapse
|
72
|
Ohman RE, Yang EH, Abel ML. Inequity in Cardio-Oncology: Identifying Disparities in Cardiotoxicity and Links to Cardiac and Cancer Outcomes. J Am Heart Assoc 2021; 10:e023852. [PMID: 34913366 PMCID: PMC9075267 DOI: 10.1161/jaha.121.023852] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Minority and underresourced communities experience disproportionately high rates of fatal cancer and cardiovascular disease. The intersection of these disparities within the multidisciplinary field of cardio‐oncology is in critical need of examination, given the risk of perpetuating health inequities in the growing vulnerable population of patients with cancer and cardiovascular disease. This review identifies 13 cohort studies and 2 meta‐analyses investigating disparate outcomes in treatment‐associated cardiotoxicity and situates these data within the context of oncologic disparities, preexisting cardiovascular disparities, and potential system‐level inequities. Black survivors of breast cancer have elevated risks of cardiotoxicity morbidity and mortality compared with White counterparts. Adolescent and young adult survivors of cancer with lower socioeconomic status experience worsened cardiovascular outcomes compared with those of higher socioeconomic status. Female patients treated with anthracyclines or radiation have higher risks of cardiotoxicity compared with male patients. Given the paucity of data, our understanding of these racial and ethnic, socioeconomic, and sex and gender disparities remains limited and large‐scale studies are needed for elucidation. Prioritizing this research while addressing clinical trial inclusion and access to specialist care is paramount to reducing health inequity.
Collapse
Affiliation(s)
- Rachel E Ohman
- Department of Medicine University of California Los Angeles Los Angeles CA
| | - Eric H Yang
- UCLA Cardio-Oncology Program Division of Cardiology Department of Medicine University of California at Los Angeles CA
| | - Melissa L Abel
- Center for Cancer Research National Cancer Institute Bethesda MD
| |
Collapse
|
73
|
Martinez HR, Beasley GS, Goldberg JF, Absi M, Ryan KA, Guerrier K, Joshi VM, Johnson JN, Morin CE, Hurley C, Morrison RR, Rai P, Hankins JS, Bishop MW, Triplett BM, Ehrhardt MJ, Pui CH, Inaba H, Towbin JA. Pediatric Cardio-Oncology Medicine: A New Approach in Cardiovascular Care. CHILDREN (BASEL, SWITZERLAND) 2021; 8:children8121200. [PMID: 34943396 PMCID: PMC8699848 DOI: 10.3390/children8121200] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/08/2021] [Accepted: 12/10/2021] [Indexed: 12/11/2022]
Abstract
Survival for pediatric patients diagnosed with cancer has improved significantly. This achievement has been made possible due to new treatment modalities and the incorporation of a systematic multidisciplinary approach for supportive care. Understanding the distinctive cardiovascular characteristics of children undergoing cancer therapies has set the underpinnings to provide comprehensive care before, during, and after the management of cancer. Nonetheless, we acknowledge the challenge to understand the rapid expansion of oncology disciplines. The limited guidelines in pediatric cardio-oncology have motivated us to develop risk-stratification systems to institute surveillance and therapeutic support for this patient population. Here, we describe a collaborative approach to provide wide-ranging cardiovascular care to children and young adults with oncology diseases. Promoting collaboration in pediatric cardio-oncology medicine will ultimately provide excellent quality of care for future generations of patients.
Collapse
Affiliation(s)
- Hugo R. Martinez
- Division of Pediatric Cardiology, The Heart Institute at Le Bonheur Children’s Hospital, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (G.S.B.); (J.F.G.); (M.A.); (K.A.R.); (K.G.); (V.M.J.); (J.N.J.); (J.A.T.)
- Correspondence:
| | - Gary S. Beasley
- Division of Pediatric Cardiology, The Heart Institute at Le Bonheur Children’s Hospital, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (G.S.B.); (J.F.G.); (M.A.); (K.A.R.); (K.G.); (V.M.J.); (J.N.J.); (J.A.T.)
| | - Jason F. Goldberg
- Division of Pediatric Cardiology, The Heart Institute at Le Bonheur Children’s Hospital, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (G.S.B.); (J.F.G.); (M.A.); (K.A.R.); (K.G.); (V.M.J.); (J.N.J.); (J.A.T.)
| | - Mohammed Absi
- Division of Pediatric Cardiology, The Heart Institute at Le Bonheur Children’s Hospital, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (G.S.B.); (J.F.G.); (M.A.); (K.A.R.); (K.G.); (V.M.J.); (J.N.J.); (J.A.T.)
| | - Kaitlin A. Ryan
- Division of Pediatric Cardiology, The Heart Institute at Le Bonheur Children’s Hospital, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (G.S.B.); (J.F.G.); (M.A.); (K.A.R.); (K.G.); (V.M.J.); (J.N.J.); (J.A.T.)
| | - Karine Guerrier
- Division of Pediatric Cardiology, The Heart Institute at Le Bonheur Children’s Hospital, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (G.S.B.); (J.F.G.); (M.A.); (K.A.R.); (K.G.); (V.M.J.); (J.N.J.); (J.A.T.)
| | - Vijaya M. Joshi
- Division of Pediatric Cardiology, The Heart Institute at Le Bonheur Children’s Hospital, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (G.S.B.); (J.F.G.); (M.A.); (K.A.R.); (K.G.); (V.M.J.); (J.N.J.); (J.A.T.)
| | - Jason N. Johnson
- Division of Pediatric Cardiology, The Heart Institute at Le Bonheur Children’s Hospital, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (G.S.B.); (J.F.G.); (M.A.); (K.A.R.); (K.G.); (V.M.J.); (J.N.J.); (J.A.T.)
| | - Cara E. Morin
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA;
| | - Caitlin Hurley
- Division of Critical Care Medicine, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (C.H.); (R.R.M.)
- Department of Bone Marrow Transplantation & Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA;
| | - Ronald Ray Morrison
- Division of Critical Care Medicine, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (C.H.); (R.R.M.)
| | - Parul Rai
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (P.R.); (J.S.H.)
| | - Jane S. Hankins
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (P.R.); (J.S.H.)
| | - Michael W. Bishop
- Division of Solid Tumor, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA;
| | - Brandon M. Triplett
- Department of Bone Marrow Transplantation & Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA;
| | - Matthew J. Ehrhardt
- Division of Cancer Survivorship, Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA;
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Ching-Hon Pui
- Division of Leukemia/Lymphoma, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (C.-H.P.); (H.I.)
| | - Hiroto Inaba
- Division of Leukemia/Lymphoma, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (C.-H.P.); (H.I.)
| | - Jeffrey A. Towbin
- Division of Pediatric Cardiology, The Heart Institute at Le Bonheur Children’s Hospital, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (G.S.B.); (J.F.G.); (M.A.); (K.A.R.); (K.G.); (V.M.J.); (J.N.J.); (J.A.T.)
| |
Collapse
|
74
|
Kirichenko YY, Ilgisonis IS, Ivanova TV, Zolotukhina AS, Khabarova NV, Privalova EV, Belenkov YN. Cardiovascular toxicity of antitumor therapy: effect on myocardial and vascular remodeling. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2021. [DOI: 10.15829/1728-8800-2021-2923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Aim. To study the effect of multiagent chemotherapy on structural and functional vascular, electrophysiological parameters and cardiac hemodynamics in patients with stomach cancer.Material and methods. The study included 3 groups of 25 people: healthy volunteers, those with established cardiac disease (hypertension + coronary artery disease), gastric adenocarcinoma (fluoropyrimidine/platinum-based chemotherapy). Cancer patients before and after chemotherapy courses underwent non-invasive assessment of vascular wall and endothelial dysfunction (videocapillaroscopy, digital photoplethysmography), as well as electrocardiography and echocardiography. Healthy volunteers and cardiac patients were examined once.Results. In cancer patients, even before chemotherapy courses, endothelial dysfunction (ED) (occlusal index, 1,7 (1,4; 1,9), normal values >1,8) and structural vascular disorders (stiffness index, 8,9 m/s (7,7; 9,7), normal values <8 m/s; refractive index, 32,4% (27,5; 37,7), normal values <30%). All above-mentioned parameters significantly worsened after multiagent chemotherapy (progression of ED and vascular wall remodeling: occlusal index, 1,3 (1,2; 1,5) (p<0,0002); stiffness index, 10,3 m/s (9,5; 11,2) (p<0,0001); reflection index, 40,2% (35,5; 43,6) (p<0,001) Decrease in left ventricular ejection fraction and diastolic function was detected. The number of supraventricular and ventricular premature beats during chemotherapy increased 9 and 10 times, respectively (p<0,05).Conclusion. The study for the first time assessed the effect of multiagent chemotherapy on ED, vascular stiffness and cardiac hemodynamics in patients with gastric cancer. A significant aggravation of all endothelial function parameters after treatment has been proven, which requires further study in order to develop criteria for early cardiovascular toxicity.
Collapse
Affiliation(s)
| | | | | | | | | | - E. V. Privalova
- Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Yu. N. Belenkov
- Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| |
Collapse
|
75
|
Mohamed TMA, Moslehi J, Satin J. Editorial: Recent Advances in Cardiotoxicity Testing. Front Pharmacol 2021; 12:798189. [PMID: 34819868 PMCID: PMC8606750 DOI: 10.3389/fphar.2021.798189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 10/26/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Tamer M A Mohamed
- Department of Medicine, The Institute of Molecular Cardiology, University of Louisville, Louisville, KY, United States.,Department of Medicine, Diabetes and Obesity Center, Envirome Institute, University of Louisville, Louisville, KY, United States
| | - Javid Moslehi
- Department of Cardio-oncology, University of California, San Francisco, San Francisco, CA, United States
| | - Jonathan Satin
- Department of Physiology, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
76
|
Khouri C, Mahé J, Caquelin L, Locher C, Despas F. Pharmacology and pharmacovigilance of protein kinase inhibitors. Therapie 2021; 77:207-217. [PMID: 34895753 DOI: 10.1016/j.therap.2021.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 10/31/2021] [Indexed: 11/19/2022]
Abstract
Protein kinase inhibitors experienced their advent in the 2000s. Their market introduction made it possible to constitute a class of targeted therapies administered orally. This name was chosen to mark a break with conventional chemotherapy drugs, but it is important to stress that these are multi-target drugs with complex affinity profiles. Adverse effects can be explained by direct interactions with their targets of interest, chosen for their indications (on-target) but also interactions with other targets (off-target). The adverse effect profiles of these drugs are therefore varied and it is possible to identify common profiles related to inhibitions of common targets. Identification of these targets has improved the global understanding of the pathophysiological mechanisms underlying the onset of adverse drug reactions as well as of the related diseases, and makes it possible to predict the adverse effect profile of new protein kinase inhibitors based on their affinities. In this review, we describe the main adverse drug reactions associated with protein kinase inhibitors, their frequency and their plausible mechanisms of action.
Collapse
Affiliation(s)
- Charles Khouri
- Pharmacovigilance Department, Grenoble Alpes University Hospital, 38000 Grenoble, France; Inserm UMR 1300-HP2 Laboratory, University Grenoble Alpes, 38000 Grenoble, France
| | - Julien Mahé
- Department of Pharmacology, Regional Pharmacovigilance Center, CHU de Nantes, 44093 Nantes, France
| | - Laura Caquelin
- Inserm, CIC 1414 (centre d'investigation clinique de Rennes), Université Rennes, CHU de Rennes, 35000 Rennes, France
| | - Clara Locher
- Inserm, CIC 1414 (centre d'investigation clinique de Rennes), Université Rennes, CHU de Rennes, 35000 Rennes, France
| | - Fabien Despas
- Inserm 1297, CIC 1436, Department of Medical and Clinical Pharmacology, Faculty of Medicine, CHU de Toulouse, University Paul-Sabatier, 31000 Toulouse, France.
| |
Collapse
|
77
|
Avagimyan AA, Mkrtchyan LH, Gevorkyan AA, Kononchuk NB, Kakturskiy LV, Djndoyan ZT. Relationship between Chemotherapy and Atrial Fibrillation: Clinical Case. RATIONAL PHARMACOTHERAPY IN CARDIOLOGY 2021. [DOI: 10.20996/1819-6446-2021-10-17] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The aim of this article is to represent the characterization of the clinical case of chemotherapy-related atrial fibrillation (AF) development in the young woman, elaborated as a result of multiple neoadjuvant and adjuvant modes of the intake of chemotherapy (both anthracycline based and non-anthracycline ones). In this case, the noted disturbances of heart rhythm should be recognized as a manifestation of cardiotoxicity. The latter implies the degree of detrimental effect of chemotherapeutical medication on the morphophynctional parameters of the cardiovascular system. Anthracycline drugs, being highly effective chemotherapytical agents, provide well-known toxic effects on the heart and vessels. Anthracycline mediated cardiotoxicity is a well- known veracity that dates back to the 60s of the last century, but up to now this medication sustains irreplaceable components of big volume of chemotherapy modes. Moreover, it should be noted that relatively newer drugs also posses certain cardiotoxicogenic potential.
Collapse
|
78
|
Abstract
Vasospastic disorders are prevalent in the general population and can affect individuals of any age. Primary (or idiopathic) vasospastic disorders often have a benign course; treatment focuses on the control of symptoms. Secondary vasospastic disorders occur owing to an underlying condition and have an increased risk of complications, including tissue loss and digital ulcerations; treatment should focus on the underlying condition. In this review, we discuss the pathophysiology, clinical presentation, diagnosis, and management of vasospastic disorders, including Raynaud syndrome, acrocyanosis, livedo reticularis, and pernio.
Collapse
Affiliation(s)
- Ana I Casanegra
- Vascular Medicine Division, Cardiovascular Department, Gonda Vascular Center, Mayo Clinic, 200 1st Street Southwest, Rochester, MN 55905, USA.
| | - Roger F Shepherd
- Vascular Medicine Division, Cardiovascular Department, Gonda Vascular Center, Mayo Clinic, 200 1st Street Southwest, Rochester, MN 55905, USA
| |
Collapse
|
79
|
Guo X, Qian X, Jin Y, Kong X, Qi Z, Cai T, Zhang L, Wu C, Li W. Hypertension Induced by Combination Therapy of Cancer: A Systematic Review and Meta-Analysis of Global Clinical Trials. Front Pharmacol 2021; 12:712995. [PMID: 34552487 PMCID: PMC8451955 DOI: 10.3389/fphar.2021.712995] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/20/2021] [Indexed: 12/26/2022] Open
Abstract
Background: Nowadays, due to the limitation of single therapy, combination therapy for cancer treatments has become important strategy. With the advancement of research on cardiotoxicities induced by anti-cancer treatment, among which cancer treatment-induced hypertension is the most frequent case. However, due to the small sample size and the absence of comparison (single-arm study alone), these studies have limitations to produce a feasible conclusion. Therefore, it is necessary to carry out a meta-analysis focusing on hypertension caused by cancer combination therapy. Methods: We systematically searched PubMed, Embase, Cochrane Library, Web of Science, and CNKI, from database inception to November 31, 2020, with randomized controlled trials (RCTs) associated with hypertension induced by cancer combination drugs. The main endpoint of which was to assess the difference in the incidence of hypertension in cancer patients with monotherapy or combination therapy. We calculated the corresponding 95% confidence interval (95% CIs) according to the random effect model and evaluated the heterogeneity between different groups. Results: According to the preset specific inclusion and exclusion criteria, a total of 23 eligible RCTs have been included in the present meta-analysis, including 6,241 patients (Among them, 2872 patients were the control group and 3369 patients were the experimental group). The results showed that cancer patients with combination therapy led to a higher risk of hypertension (All-grade: RR 2.85, 95% CI 2.52∼3.22; 1∼2 grade: RR 2.43, 95% CI 2.10∼2.81; 3∼4 grade: RR 4.37, 95% CI 3.33∼5.72). Furthermore, compared with the control group who received or did not receive a placebo, there was a higher risk of grade 3-4 hypertension caused by cancer combination treatment. Conclusion: The present meta-analysis carries out a comprehensive analysis on the risk of patients suffering from hypertension in the process of multiple cancer combination therapies. Findings in our study support that the risk of hypertension may increase significantly in cancer patients with multiple cancer combination therapies. The outcomes of this meta-analysis may provide a reference value for clinical practice and may supply insights in reducing the incidence of hypertension caused by cancer combined treatment.
Collapse
Affiliation(s)
- Xiaodan Guo
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Xiaoyu Qian
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Ying Jin
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xiangyi Kong
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhihong Qi
- Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Tie Cai
- State Key Laboratory of Coal Resources and Safe Mining, School of Chemical and Environmental Engineering, China University of Mining and Technology, Beijing, China
| | - Lin Zhang
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Melbourne School of Population and Global Health, University of Melbourne, Melbourne, VIC, Australia
| | - Caisheng Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Weihua Li
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
80
|
Sawami K, Tanaka A, Node K. Clinical roles of vascular function assessment in cancer care. Support Care Cancer 2021; 30:985-987. [PMID: 34378082 DOI: 10.1007/s00520-021-06465-6] [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: 01/22/2021] [Accepted: 07/24/2021] [Indexed: 11/24/2022]
Abstract
Atherosclerotic cardiovascular disease and cancer often share risk factors and influence each other's pathological features. In addition to the unfavorable effects of cancer on the cardiovascular system, the adverse effects of cancer therapies, such as chemotherapy and radiation, have also been determined. In particular, vascular toxic effects associated with cancer therapy (vasospasm, thrombosis formation, and promotion of arteriosclerosis), which are the second most common complications after myocardial adverse effects, are usually managed after the onset of vascular diseases, because screening and predictive methods are yet to be fully established. However, the onset of these vascular complications has a major influence on the implementation of cancer therapy, resulting in worsening of the quality of cancer care and prognosis of patients with cancer. It is therefore necessary to establish clinical strategies for detecting the vascular adverse effects of cancer therapy and evaluating vascular function during cancer care. In this article, we discuss the expected role of vascular function assessment using physiological testing tools for early detection of vascular adverse effects caused by cancer therapy and also preemptive assessment of vascular function prior to this treatment being initiated.
Collapse
Affiliation(s)
- Kosuke Sawami
- Department of Cardiovascular Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Atsushi Tanaka
- Department of Cardiovascular Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan.
| | - Koichi Node
- Department of Cardiovascular Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| |
Collapse
|
81
|
Chen X, Zhabyeyev P, Azad AK, Vanhaesebroeck B, Grueter CE, Murray AG, Kassiri Z, Oudit GY. Pharmacological and cell-specific genetic PI3Kα inhibition worsens cardiac remodeling after myocardial infarction. J Mol Cell Cardiol 2021; 157:17-30. [PMID: 33887328 DOI: 10.1016/j.yjmcc.2021.04.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND PI3Kα (Phosphoinositide 3-kinase α) regulates multiple downstream signaling pathways controlling cell survival, growth, and proliferation and is an attractive therapeutic target in cancer and obesity. The clinically-approved PI3Kα inhibitor, BYL719, is in further clinical trials for cancer and overgrowth syndrome. However, the potential impact of PI3Kα inhibition on the heart and following myocardial infarction (MI) is unclear. We aim to determine whether PI3Kα inhibition affects cardiac physiology and post-MI remodeling and to elucidate the underlying molecular mechanisms. METHODS AND RESULTS Wildtype (WT) 12-wk old male mice receiving BYL719 (daily, p.o.) for 10 days showed reduction in left ventricular longitudinal strain with normal ejection fraction, weight loss, mild cardiac atrophy, body composition alteration, and prolonged QTC interval. RNASeq analysis showed gene expression changes in multiple pathways including extracellular matrix remodeling and signaling complexes. After MI, both p110α and phospho-Akt protein levels were increased in human and mouse hearts. Pharmacological PI3Kα inhibition aggravated cardiac dysfunction and resulted in adverse post-MI remodeling, with increased apoptosis, elevated inflammation, suppressed hypertrophy, decreased coronary blood vessel density, and inhibited Akt/GSK3β/eNOS signaling. Selective genetic ablation of PI3Kα in endothelial cells was associated with worsened post-MI cardiac function and reduced coronary blood vessel density. In vitro, BYL719 suppressed Akt/eNOS activation, cell viability, proliferation, and angiogenic sprouting in coronary and human umbilical vein endothelial cells. Cardiomyocyte-specific genetic PI3Kα ablation resulted in mild cardiac systolic dysfunction at baseline. After MI, cardiac function markedly deteriorated with increased mortality concordant with greater apoptosis and reduced hypertrophy. In isolated adult mouse cardiomyocytes, BYL719 decreased hypoxia-associated activation of Akt/GSK3β signaling and cell survival. CONCLUSIONS PI3Kα is required for cell survival (endothelial cells and cardiomyocytes) hypertrophic response, and angiogenesis to maintain cardiac function after MI. Therefore, PI3Kα inhibition that is used as anti-cancer treatment, can be cardiotoxic, especially after MI.
Collapse
Affiliation(s)
- Xueyi Chen
- Department of Medicine, University of Alberta, Edmonton, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Pavel Zhabyeyev
- Department of Medicine, University of Alberta, Edmonton, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Abul K Azad
- Department of Medicine, University of Alberta, Edmonton, Canada
| | | | - Chad E Grueter
- Division of Cardiovascular Medicine, Department of Internal Medicine, Francois M. Abboud Cardiovascular Research Center, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, USA
| | - Allan G Murray
- Department of Medicine, University of Alberta, Edmonton, Canada
| | - Zamaneh Kassiri
- Department of Physiology, University of Alberta, Edmonton, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Gavin Y Oudit
- Department of Medicine, University of Alberta, Edmonton, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada.
| |
Collapse
|
82
|
Cardiac risk stratification in cancer patients: A longitudinal patient-patient network analysis. PLoS Med 2021; 18:e1003736. [PMID: 34339408 PMCID: PMC8366997 DOI: 10.1371/journal.pmed.1003736] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 08/16/2021] [Accepted: 07/15/2021] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Cardiovascular disease is a leading cause of death in general population and the second leading cause of mortality and morbidity in cancer survivors after recurrent malignancy in the United States. The growing awareness of cancer therapy-related cardiac dysfunction (CTRCD) has led to an emerging field of cardio-oncology; yet, there is limited knowledge on how to predict which patients will experience adverse cardiac outcomes. We aimed to perform unbiased cardiac risk stratification for cancer patients using our large-scale, institutional electronic medical records. METHODS AND FINDINGS We built a large longitudinal (up to 22 years' follow-up from March 1997 to January 2019) cardio-oncology cohort having 4,632 cancer patients in Cleveland Clinic with 5 diagnosed cardiac outcomes: atrial fibrillation, coronary artery disease, heart failure, myocardial infarction, and stroke. The entire population includes 84% white Americans and 11% black Americans, and 59% females versus 41% males, with median age of 63 (interquartile range [IQR]: 54 to 71) years old. We utilized a topology-based K-means clustering approach for unbiased patient-patient network analyses of data from general demographics, echocardiogram (over 25,000), lab testing, and cardiac factors (cardiac). We performed hazard ratio (HR) and Kaplan-Meier analyses to identify clinically actionable variables. All confounding factors were adjusted by Cox regression models. We performed random-split and time-split training-test validation for our model. We identified 4 clinically relevant subgroups that are significantly correlated with incidence of cardiac outcomes and mortality. Among the 4 subgroups, subgroup I (n = 625) has the highest risk of de novo CTRCD (28%) with an HR of 3.05 (95% confidence interval (CI) 2.51 to 3.72). Patients in subgroup IV (n = 1,250) had the worst survival probability (HR 4.32, 95% CI 3.82 to 4.88). From longitudinal patient-patient network analyses, the patients in subgroup I had a higher percentage of de novo CTRCD and a worse mortality within 5 years after the initiation of cancer therapies compared to long-time exposure (6 to 20 years). Using clinical variable network analyses, we identified that serum levels of NT-proB-type Natriuretic Peptide (NT-proBNP) and Troponin T are significantly correlated with patient's mortality (NT-proBNP > 900 pg/mL versus NT-proBNP = 0 to 125 pg/mL, HR = 2.95, 95% CI 2.28 to 3.82, p < 0.001; Troponin T > 0.05 μg/L versus Troponin T ≤ 0.01 μg/L, HR = 2.08, 95% CI 1.83 to 2.34, p < 0.001). Study limitations include lack of independent cardio-oncology cohorts from different healthcare systems to evaluate the generalizability of the models. Meanwhile, the confounding factors, such as multiple medication usages, may influence the findings. CONCLUSIONS In this study, we demonstrated that the patient-patient network clustering methodology is clinically intuitive, and it allows more rapid identification of cancer survivors that are at greater risk of cardiac dysfunction. We believed that this study holds great promise for identifying novel cardiac risk subgroups and clinically actionable variables for the development of precision cardio-oncology.
Collapse
|
83
|
Sano S, Wang Y, Ogawa H, Horitani K, Sano M, Polizio AH, Kour A, Yura Y, Doviak H, Walsh K. TP53-mediated therapy-related clonal hematopoiesis contributes to doxorubicin-induced cardiomyopathy by augmenting a neutrophil-mediated cytotoxic response. JCI Insight 2021; 6:e146076. [PMID: 34236050 PMCID: PMC8410064 DOI: 10.1172/jci.insight.146076] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 05/13/2021] [Indexed: 12/27/2022] Open
Abstract
Therapy-related clonal hematopoiesis (t-CH) is often observed in cancer survivors. This form of clonal hematopoiesis typically involves somatic mutations in driver genes that encode components of the DNA damage response and confer hematopoietic stem and progenitor cells (HSPCs) with resistance to the genotoxic stress of the cancer therapy. Here, we established a model of TP53-mediated t-CH through the transfer of Trp53 mutant HSPCs to mice, followed by treatment with a course of the chemotherapeutic agent doxorubicin. These studies revealed that neutrophil infiltration in the heart significantly contributes to doxorubicin-induced cardiac toxicity and that this condition is amplified in the model of Trp53-mediated t-CH. These data suggest that t-CH could contribute to the elevated heart failure risk that occurs in cancer survivors who have been treated with genotoxic agents.
Collapse
Affiliation(s)
- Soichi Sano
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA.,Department of Cardiology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Ying Wang
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Hayato Ogawa
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Keita Horitani
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Miho Sano
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Ariel H Polizio
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Anupreet Kour
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Yoshimitsu Yura
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Heather Doviak
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Kenneth Walsh
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
84
|
Ording AG, Søgaard M, Skjøth F, Grove EL, Lip GYH, Larsen TB, Nielsen PB. Bleeding complications in patients with gastrointestinal cancer and atrial fibrillation treated with oral anticoagulants. Cancer Med 2021; 10:4405-4414. [PMID: 34114733 PMCID: PMC8267127 DOI: 10.1002/cam4.4012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 05/06/2021] [Accepted: 05/12/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Direct oral anticoagulants (DOACs) may increase the risk of gastrointestinal (GI) bleeding in patients with atrial fibrillation (AF) and GI cancer compared with vitamin K antagonists (VKA). METHODS We conducted a Danish nationwide cohort study comparing the bleeding risk associated with DOAC versus VKA in patients with AF and GI cancer. We calculated crude bleeding rates per 100 person-years (PYs) for GI and major bleeding. We then compared rates of bleeding at 1 year after initial oral anticoagulation filled prescription by treatment regimen using inverse probability of treatment weighting and Cox regression. RESULTS The unweighted study population included 1476 AF patients with GI cancer (41.6% women, median age 78 years) initiating a DOAC and 652 initiating a VKA. One-year risk of GI bleeding was 5.0% in the DOAC group and 4.7% in the VKA group with a corresponding weighted hazard ratio (HR) of 0.95 (95% confidence interval [CI]: 0.63, 1.45). For patients with active cancer, weighted GI bleeding rates were slightly higher in both the VKA and DOAC group, and the weighted HR was 1.00 (95% CI: 0.53, 1.88). The HR was 1.12 (95% CI: 0.71, 1.76) for all bleedings. Hazard ratios for GI bleeding were 0.61 (95% CI: 0.25, 1.52) for patients with upper GI cancer, and 0.92 (95% CI: 0.58, 1.46) in patients with colorectal cancer. CONCLUSION Evidence from this nationwide cohort study suggests a comparable 1-year risk of bleeding associated with DOAC compared with VKA among patients with AF and GI cancer.
Collapse
Affiliation(s)
- Anne Gulbech Ording
- Unit for Thrombosis and Drug ResearchDepartment of CardiologyAalborg University HospitalAalborgDenmark
| | - Mette Søgaard
- Unit for Thrombosis and Drug ResearchDepartment of CardiologyAalborg University HospitalAalborgDenmark
- Aalborg Thrombosis Research UnitAalborg UniversityAalborgDenmark
| | - Flemming Skjøth
- Aalborg Thrombosis Research UnitAalborg UniversityAalborgDenmark
- Unit of Clinical BiostatisticsAalborg University HospitalAalborgDenmark
| | - Erik Lerkevang Grove
- Department of CardiologyAarhus University HospitalAarhus NDenmark
- Department of Clinical MedicineFaculty of HealthAarhus UniversityAarhus CDenmark
| | - Gregory Y. H. Lip
- Unit for Thrombosis and Drug ResearchDepartment of CardiologyAalborg University HospitalAalborgDenmark
- Liverpool Centre for Cardiovascular SciencesUniversity Liverpool and Liverpool Heart & Chest HospitalLiverpoolUK
| | - Torben Bjerregaard Larsen
- Unit for Thrombosis and Drug ResearchDepartment of CardiologyAalborg University HospitalAalborgDenmark
- Aalborg Thrombosis Research UnitAalborg UniversityAalborgDenmark
| | - Peter Brønnum Nielsen
- Unit for Thrombosis and Drug ResearchDepartment of CardiologyAalborg University HospitalAalborgDenmark
- Aalborg Thrombosis Research UnitAalborg UniversityAalborgDenmark
| |
Collapse
|
85
|
Ording AG, Skjøth F, Søgaard M, Højen AA, Overvad TF, Noble S, Goldhaber SZ, Larsen TB. Increasing Incidence and Declining Mortality After Cancer-Associated Venous Thromboembolism: A Nationwide Cohort Study. Am J Med 2021; 134:868-876.e5. [PMID: 33631159 DOI: 10.1016/j.amjmed.2021.01.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 02/08/2023]
Abstract
PURPOSE The incidence of cancer-associated venous thromboembolism has increased, but whether short-term mortality after cancer-associated venous thromboembolism has changed remains uncertain. We investigated whether the increasing incidence of venous thromboembolism in cancer patients is associated with a change in mortality. METHODS We used administrative medical registries to identify a cohort of all Danish patients diagnosed with a first primary cancer from 2006 to 2017. We examined temporal changes in 1-year risks of venous thromboembolism and in mortality risks at 30 days and 1 year after venous thromboembolism. Cox regression was used to assess changes in mortality rate ratios over time. RESULTS We included 350,272 cancer patients (median age 68 years, 49.1% female), of whom 8167 developed venous thromboembolism within 1 year after cancer diagnosis. The cumulative 1-year risk of venous thromboembolism was 1.8% in 2006-2008, increasing to 2.8% for patients diagnosed in 2015-2017. The 30-day mortality after venous thromboembolism decreased from 15.1% in 2006-2008 to 12.7% in 2015-2017, and the 1-year mortality decreased from 52.4% to 45.8%, equivalent to a hazard ratio (HR) of 0.83 (95% confidence interval [CI], 0.75-0.90). This pattern of declining 1-year mortality was consistent for patients with pulmonary embolism, HR 0.79 (95% CI, 0.69-0.90), and deep venous thrombosis, HR 0.76 (95% CI, 0.67-0.87). Lower mortality over time was evident across all strata of cancer stage, cancer type, and cancer treatment. CONCLUSIONS The 1-year risk of venous thromboembolism after a first primary cancer diagnosis in Denmark increased during 2006-2017. This increase was accompanied by declining mortality.
Collapse
Affiliation(s)
| | - Flemming Skjøth
- Unit Clinical Biostatistics, Aalborg University Hospital, Aalborg, Denmark; Aalborg Thrombosis Research Unit, Department of Clinical Medicine, Faculty of Medicine, Aalborg University, Aalborg, Denmark
| | - Mette Søgaard
- Unit for Thrombosis and Drug Research, Department of Cardiology; Aalborg Thrombosis Research Unit, Department of Clinical Medicine, Faculty of Medicine, Aalborg University, Aalborg, Denmark
| | | | - Thure Filskov Overvad
- Unit for Thrombosis and Drug Research, Department of Cardiology; Department of Oncology, Aalborg University Hospital, Aalborg, Denmark
| | - Simon Noble
- Marie Curie Palliative Care Research Centre, Cardiff University, Cardiff, UK
| | - Samuel Zachary Goldhaber
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Torben Bjerregaard Larsen
- Unit for Thrombosis and Drug Research, Department of Cardiology; Aalborg Thrombosis Research Unit, Department of Clinical Medicine, Faculty of Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
86
|
Prevalence, clinical characteristics, and impact of active cancer in patients with acute myocardial infarction: data from an all-comer registry. J Cardiol 2021; 78:193-200. [PMID: 34167885 DOI: 10.1016/j.jjcc.2021.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/30/2021] [Accepted: 04/02/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND Although a history of cancer is a poor prognostic factor in patients with acute myocardial infarction (AMI), the clinical importance of coexisting active cancer remains unclear. METHODS In this single-center retrospective study, we reviewed an AMI registry and assessed the prevalence and predictors of active cancer, 1-year incidence of cardiac death or major bleeding events (defined as a Bleeding Academy Research Consortium type 3 or 5), and the impact of coexisting active cancer on clinical outcomes. Active cancer was defined as either an already-diagnosed or undiagnosed occult cancer. RESULTS Between January 2012 and December 2017, 1140 AMI patients (median age, 69 years; male, 76.0%) were enrolled. Active and historical cancers were diagnosed in 63 patients (5.5%) and 50 patients (4.4%), respectively. The most common location was the urinary tract (n=21). In the Kaplan-Meier analysis, the active cancer group had a higher incidence of 1-year cardiac death (17.5% vs. 5.3%, p < 0.001) and major bleeding events (19.0% vs. 5.6%, p < 0.001) than the non-cancer group. In the multivariate Cox proportional hazards regression models, active cancer was an independent predictor of both cardiac death and major bleeding at 1 year. Specifically, gastrointestinal tract and advanced-stage cancers had the poorest outcomes. Compared to the non-cancer group, the 1-year major bleeding rate was higher for all cancer types and stages. In contrast, early-stage cancers had a weaker impact on the 1-year cardiac mortality compared to advanced-stage cancers. Similarly, cardiac death during 1-year also occurred less frequently in occult cancers than in already-known cancers. CONCLUSIONS In patients with AMI, coexisting active cancer was rare, but it significantly impacted cardiac death and major bleeding events.
Collapse
|
87
|
Fleming MR, Xiao L, Jackson KD, Beckman JA, Barac A, Moslehi JJ. Vascular Impact of Cancer Therapies: The Case of BTK (Bruton Tyrosine Kinase) Inhibitors. Circ Res 2021; 128:1973-1987. [PMID: 34110908 PMCID: PMC10185355 DOI: 10.1161/circresaha.121.318259] [Citation(s) in RCA: 10] [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: 01/15/2023]
Abstract
Novel targeted cancer therapies have revolutionized oncology therapies, but these treatments can have cardiovascular complications, which include heterogeneous cardiac, metabolic, and vascular sequelae. Vascular side effects have emerged as important considerations in both cancer patients undergoing active treatment and cancer survivors. Here, we provide an overview of vascular effects of cancer therapies, focusing on small-molecule kinase inhibitors and specifically inhibitors of BTK (Bruton tyrosine kinase), which have revolutionized treatment and prognosis for B-cell malignancies. Cardiovascular side effects of BTK inhibitors include atrial fibrillation, increased risk of bleeding, and hypertension, with the former 2 especially providing a treatment challenge for the clinician. Cardiovascular complications of small-molecule kinase inhibitors can occur through either on-target (targeting intended target kinase) or off-target kinase inhibition. We will review these concepts and focus on the case of BTK inhibitors, highlight the emerging data suggesting an off-target effect that may provide insights into development of arrhythmias, specifically atrial fibrillation. We believe that cardiac and vascular sequelae of novel targeted cancer therapies can provide insights into human cardiovascular biology.
Collapse
Affiliation(s)
- Matthew R Fleming
- Division of Cardiovascular Medicine (M.R.F., J.A.B., J.J.M.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Ling Xiao
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston (L.X.)
| | - Klarissa D Jackson
- UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill (K.D.J.)
| | - Joshua A Beckman
- Division of Cardiovascular Medicine (M.R.F., J.A.B., J.J.M.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Ana Barac
- Georgetown University and MedStar Heart and Vascular Institute, MedStar Washing Hospital Center, DC (A.B.)
| | - Javid J Moslehi
- Division of Cardiovascular Medicine (M.R.F., J.A.B., J.J.M.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Cardio-Oncology Program (J.J.M.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
88
|
Nonaka M, Hosoda H, Uezono Y. Cancer treatment-related cardiovascular disease: Current status and future research priorities. Biochem Pharmacol 2021; 190:114599. [PMID: 33989656 DOI: 10.1016/j.bcp.2021.114599] [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: 03/26/2021] [Revised: 05/07/2021] [Accepted: 05/07/2021] [Indexed: 10/21/2022]
Abstract
With the development of new drugs, such as molecular-targeted drugs, and multidisciplinary therapies, cancer treatment outcomes have improved, and the number of cancer survivors is increasing every year. However, some chemotherapeutic agents cause cardiovascular complications (cancer treatment-related cardiovascular disease, CTRCVD), which affect the life prognosis and quality of life (QOL) of cancer patients. Therefore, it is necessary to select treatment methods that take into account the prognosis and QOL of cancer patients, and to take measures against CTRCVD. The mechanism of cardiotoxicity of high-risk drugs, such as doxorubicin and HER2 inhibitors, are still unclear; genetic factors, and cardiovascular disease risk factors (e.g., hypertension, dyslipidemia, and diabetes) are associated with CTRCVD progression. The establishment of methods for prevention, early diagnosis, and treatment of CTRCVD and the generation of evidence for these methods are needed. It is also necessary to develop screening methods for chemotherapy cardiotoxicity. In this review, we discuss the current status of CTRCVD, its complications, and expected countermeasures.
Collapse
Affiliation(s)
- Miki Nonaka
- Department of Pain Control Research, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan(1)
| | - Hiroshi Hosoda
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute, 6-1, Kishibe-Shimmachi, Suita-City, Osaka 564-8565, Japan(1)
| | - Yasuhito Uezono
- Department of Pain Control Research, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan(1); Department of Pain Medicine, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; Supportive and Palliative Care Research Support Office, National Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa-City, Chiba 277-8577, Japan; Project for Supportive Care Research, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, 5-1-1, Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.
| |
Collapse
|
89
|
Baldassarre LA, Yang EH, Cheng RK, DeCara JM, Dent S, Liu JE, Rudski LG, Strom JB, Thavendiranathan P, Barac A, Zaha VG, Bucciarelli-Ducci C, Ellahham S, Deswal A, Lenneman C, Villarraga HR, Blaes AH, Ismail-Khan R, Ky B, Leja MJ, Scherrer-Crosbie M. Cardiovascular Care of the Oncology Patient During COVID-19: An Expert Consensus Document From the ACC Cardio-Oncology and Imaging Councils. J Natl Cancer Inst 2021; 113:513-522. [PMID: 33179744 PMCID: PMC7717327 DOI: 10.1093/jnci/djaa177] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 09/21/2020] [Accepted: 11/02/2020] [Indexed: 12/15/2022] Open
Abstract
In response to the coronavirus disease 2019 (COVID-19) pandemic, the Cardio-Oncology and Imaging Councils of the American College of Cardiology offers recommendations to clinicians regarding the cardiovascular care of cardio-oncology patients in this expert consensus statement. Cardio-oncology patients-individuals with an active or prior cancer history and with or at risk of cardiovascular disease-are a rapidly growing population who are at increased risk of infection, and experiencing severe and/or lethal complications by COVID-19. Recommendations for optimizing screening and monitoring visits to detect cardiac dysfunction are discussed. In addition, judicious use of multimodality imaging and biomarkers are proposed to identify myocardial, valvular, vascular, and pericardial involvement in cancer patients. The difficulties of diagnosing the etiology of cardiovascular complications in patients with cancer and COVID-19 are outlined, along with weighing the advantages against risks of exposure, with the modification of existing cardiovascular treatments and cardiotoxicity surveillance in patients with cancer during the COVID-19 pandemic.
Collapse
Affiliation(s)
- Lauren A Baldassarre
- Affiliations of authors: Section of Cardiovascular Medicine, Department of Medicine, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Eric H Yang
- UCLA Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Richard K Cheng
- Cardio-Oncology Program, Department of Medicine, Division of Cardiology and Department of Radiology, University of Washington, Seattle, WA, USA
| | - Jeanne M DeCara
- Section of Cardiology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Susan Dent
- Duke Cancer Institute, Department of Medicine, Duke University, Durham, NC, USA
| | - Jennifer E Liu
- Cardiology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lawrence G Rudski
- Azrieli Heart Center, Department of Medicine, Jewish General Hospital, McGill University, Montreal, QC, Canada
| | - Jordan B Strom
- Richard A. and Susan F. Smith Center for Outcomes Research in Cardiology, Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Paaladinesh Thavendiranathan
- Ted Rogers Program in Cardiotoxicity Prevention, Division of Cardiology, Peter Munk Cardiac Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Ana Barac
- Medstar Heart and Vascular Institute, Georgetown University, Washington, DC, USA
| | - Vlad G Zaha
- Cardio-Oncology Program, Harold C. Simmons Comprehensive Cancer Center, Division of Cardiology, Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Chiara Bucciarelli-Ducci
- Bristol Heart Institute, Bristol National Institute of Health Research (NIHR) Biomedical Research Centre, University Hospitals Bristol NHS Trust and University of Bristol, Bristol, UK
| | - Samer Ellahham
- Heart and Vascular Institute, Cleveland Clinic-Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Anita Deswal
- Department of Cardiology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carrie Lenneman
- Division of Cardiology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hector R Villarraga
- Department of Cardiovascular Diseases, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Anne H Blaes
- Division of Hematology and Oncology, University of Minnesota, Minneapolis, MN, USA
| | - Roohi Ismail-Khan
- Cardio-Oncology Program, Division of Oncologic Sciences, H. Lee Moffitt Cancer Center, University of South Florida, Tampa, FL, USA
| | - Bonnie Ky
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Monika J Leja
- Division of Cardiovascular Medicine, Department of Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Marielle Scherrer-Crosbie
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
90
|
Modeling Precision Cardio-Oncology: Using Human-Induced Pluripotent Stem Cells for Risk Stratification and Prevention. Curr Oncol Rep 2021; 23:77. [PMID: 33937943 PMCID: PMC8088904 DOI: 10.1007/s11912-021-01066-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2021] [Indexed: 11/12/2022]
Abstract
Purpose of Review Cardiovascular toxicity is a leading cause of mortality among cancer survivors and has become increasingly prevalent due to improved cancer survival rates. In this review, we synthesize evidence illustrating how common cancer therapeutic agents, such as anthracyclines, human epidermal growth factors receptors (HER2) monoclonal antibodies, and tyrosine kinase inhibitors (TKIs), have been evaluated in cardiomyocytes (CMs) derived from human-induced pluripotent stem cells (hiPSCs) to understand the underlying mechanisms of cardiovascular toxicity. We place this in the context of precision cardio-oncology, an emerging concept for personalizing the prevention and management of cardiovascular toxicities from cancer therapies, accounting for each individual patient’s unique factors. We outline steps that will need to be addressed by multidisciplinary teams of cardiologists and oncologists in partnership with regulators to implement future applications of hiPSCs in precision cardio-oncology. Recent Findings Current prevention of cardiovascular toxicity involves routine screenings and management of modifiable risk factors for cancer patients, as well as the initiation of cardioprotective medications. Despite recent advancements in precision cardio-oncology, knowledge gaps remain and limit our ability to appropriately predict with precision which patients will develop cardiovascular toxicity. Investigations using patient-specific CMs facilitate pharmacological discovery, mechanistic toxicity studies, and the identification of cardioprotective pathways. Studies with hiPSCs demonstrate that patients with comorbidities have more frequent adverse responses, compared to their counterparts without cardiac disease. Further studies utilizing hiPSC modeling should be considered, to evaluate the impact and mitigation of known cardiovascular risk factors, including blood pressure, body mass index (BMI), smoking status, diabetes, and physical activity in their role in cardiovascular toxicity after cancer therapy. Future real-world applications will depend on understanding the current use of hiPSC modeling in order for oncologists and cardiologists together to inform their potential to improve our clinical collaborative practice in cardio-oncology. Summary When applying such in vitro characterization, it is hypothesized that a safety score can be assigned to each individual to determine who has a greater probability of developing cardiovascular toxicity. Using hiPSCs to create personalized models and ultimately evaluate the cardiovascular toxicity of individuals’ treatments may one day lead to more patient-specific treatment plans in precision cardio-oncology while reducing cardiovascular disease (CVD) morbidity and mortality.
Collapse
|
91
|
Estratificación, monitorización y control del riesgo cardiovascular en pacientes con cáncer. Documento de consenso de SEC, FEC, SEOM, SEOR, SEHH, SEMG, AEEMT, AEEC y AECC. Rev Esp Cardiol 2021. [DOI: 10.1016/j.recesp.2020.11.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
92
|
Ren C, Sun K, Zhang Y, Hu Y, Hu B, Zhao J, He Z, Ding R, Wang W, Liang C. Sodium-Glucose CoTransporter-2 Inhibitor Empagliflozin Ameliorates Sunitinib-Induced Cardiac Dysfunction via Regulation of AMPK-mTOR Signaling Pathway-Mediated Autophagy. Front Pharmacol 2021; 12:664181. [PMID: 33995090 PMCID: PMC8116890 DOI: 10.3389/fphar.2021.664181] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/12/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Sodium–glucose cotransporter-2 (SGLT2) inhibitors have been shown to decrease the adverse cardiac events and risks of cardiovascular mortality among patients with or without diabetes, which has made these drugs promising treatment options for patients with chronic heart failure. Cardiac dysfunction is a common and severe side effect induced by cancer chemotherapies, which seriously affects the prognosis and life quality of tumor patients. However, it is not clear whether SGLT2 inhibitors have cardiovascular benefits in patients with cancer chemotherapy–related cardiac dysfunction. We aimed to determine whether empagliflozin (EMPA), an SGLT2 inhibitor, has a protective role against sunitinib (SNT)-induced cardiac dysfunction in a mouse model. Methods: Male C57BL/6J mice were randomized into control (control, n = 8), empagliflozin (EMPA, n = 8), sunitinib (SNT, n = 12), or sunitinib and empagliflozin coadministration (SNT + EMPA, n = 12) groups. EMPA, SNT, or SNT-combined EMPA was given via oral gavage for consecutive 28 days. Cardiovascular functions and pathological changes were examined, and the underlying mechanisms of EMPA’s effects were investigated in H9c2 cardiomyocytes. Results: Mice in the SNT group exhibited dramatically elevated blood pressure (systolic blood pressure [SBP] 134.30 ± 6.455 mmHg vs. 114.85 ± 6.30 mmHg) and impaired left ventricular function (left ventricular ejection fraction [LVEF] 50.24 ± 3.06% vs. 84.92 ± 2.02%), as compared with those of the control group. However, EMPA could ameliorate SNT-induced cardiotoxicity, both in terms of SBP (117.51 ± 5.28 mmHg vs. 134.30 ± 6.455 mmHg) and LVEF (76.18 ± 5.16% vs. 50.24 ± 3.06 %). In H9c2 cardiomyocytes, SNT-induced cardiomyocyte death and cell viability loss as well as dysfunction of adenosine 5’-monophosphate–activated protein kinase–mammalian target of rapamycin (AMPK-mTOR) signaling–mediated autophagy were restored by EMPA. However, these favorable effects mediated by EMPA were blocked by the inhibition of AMPK or autophagy. Conclusion: EMPA could ameliorate SNT-induced cardiac dysfunction via regulating cardiomyocyte autophagy, which was mediated by the AMPK-mTOR signaling pathway. These findings supported that SGLT2 inhibitor therapy could be a potential cardioprotective approach for cardiovascular complications among patients receiving SNT. However, these favorable effects still need to be validated in clinical trials.
Collapse
Affiliation(s)
- Changzhen Ren
- Department of Cardiology, Changzheng Hospital, Naval Medical University, Shanghai, China.,Department of General Practice, 960th Hospital of PLA, Jinan, China
| | - Kaiqiang Sun
- Department of Spine Surgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yanda Zhang
- Department of Cardiology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yangxi Hu
- Department of Cardiology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Bowen Hu
- Department of Cardiology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jian Zhao
- Department of Cardiology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Zhiqing He
- Department of Cardiology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Ru Ding
- Department of Cardiology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Weizhong Wang
- Department of Marine Biomedicine and Polar Medicine, Naval Medical Center of People's Liberation Army (PLA), Naval Medical University, Shanghai, China
| | - Chun Liang
- Department of Cardiology, Changzheng Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
93
|
Nishikawa T, Morishima T, Okawa S, Fujii Y, Otsuka T, Kudo T, Fujita T, Kamada R, Yasui T, Shioyama W, Oka T, Tabuchi T, Fujita M, Miyashiro I. Multicentre cohort study of the impact of percutaneous coronary intervention on patients with concurrent cancer and ischaemic heart disease. BMC Cardiovasc Disord 2021; 21:177. [PMID: 33849438 PMCID: PMC8045293 DOI: 10.1186/s12872-021-01968-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 03/23/2021] [Indexed: 01/22/2023] Open
Abstract
Background The incidence of concurrent cancer and ischaemic heart disease (IHD) is increasing; however, the long-term patient prognoses remain unclear. Methods Five-year all-cause mortality data pertaining to patients in the Osaka Cancer Registry, who were diagnosed with colorectal, lung, prostate, and gastric cancers between 2010 and 2015, were retrieved and analysed together with linked patient administrative data. Patient characteristics (cancer type, stage, and treatment; coronary risk factors; medications; and time from cancer diagnosis to index admission for percutaneous coronary intervention [PCI] or IHD diagnosis) were adjusted for propensity score matching. Three groups were identified: patients who underwent PCI within 3 years of cancer diagnosis (n = 564, PCI + group), patients diagnosed with IHD within 3 years of cancer diagnosis who did not undergo PCI (n = 3058, PCI-/IHD + group), and patients without IHD (n = 27,392, PCI-/IHD- group). Kaplan–Meier analysis was used for comparisons. Results After propensity score matching, the PCI + group had better prognosis (n = 489 in both groups, hazard ratio 0.64, 95% confidence interval 0.51–0.81, P < 0.001) than the PCI-/IHD + group. PCI + patients (n = 282) had significantly higher mortality than those without IHD (n = 280 in each group, hazard ratio 2.88, 95% confidence interval 1.90–4.38, P < 0.001). Conclusions PCI might improve the long-term prognosis in cancer patients with IHD. However, these patients could have significantly worse long-term prognosis than cancer patients without IHD. Since the present study has some limitations, further research will be needed on this important topic in cardio-oncology. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-021-01968-w.
Collapse
Affiliation(s)
- Tatsuya Nishikawa
- Department of Onco-Cardiology, Osaka International Cancer Institute, 3-1-69, Otemae, Chuo-ku, Osaka City, 541-8567, Japan.
| | | | - Sumiyo Okawa
- Cancer Control Centre, Osaka International Cancer Institute, Osaka, Japan
| | - Yuki Fujii
- Department of Onco-Cardiology, Osaka International Cancer Institute, 3-1-69, Otemae, Chuo-ku, Osaka City, 541-8567, Japan
| | - Tomoyuki Otsuka
- Department of Clinical Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Toshihiro Kudo
- Department of Clinical Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Takeshi Fujita
- Department of Onco-Cardiology, Osaka International Cancer Institute, 3-1-69, Otemae, Chuo-ku, Osaka City, 541-8567, Japan
| | - Risa Kamada
- Department of Onco-Cardiology, Osaka International Cancer Institute, 3-1-69, Otemae, Chuo-ku, Osaka City, 541-8567, Japan
| | - Taku Yasui
- Department of Onco-Cardiology, Osaka International Cancer Institute, 3-1-69, Otemae, Chuo-ku, Osaka City, 541-8567, Japan
| | - Wataru Shioyama
- Department of Onco-Cardiology, Osaka International Cancer Institute, 3-1-69, Otemae, Chuo-ku, Osaka City, 541-8567, Japan
| | - Toru Oka
- Department of Onco-Cardiology, Osaka International Cancer Institute, 3-1-69, Otemae, Chuo-ku, Osaka City, 541-8567, Japan
| | - Takahiro Tabuchi
- Cancer Control Centre, Osaka International Cancer Institute, Osaka, Japan
| | - Masashi Fujita
- Department of Onco-Cardiology, Osaka International Cancer Institute, 3-1-69, Otemae, Chuo-ku, Osaka City, 541-8567, Japan.
| | - Isao Miyashiro
- Cancer Control Centre, Osaka International Cancer Institute, Osaka, Japan
| |
Collapse
|
94
|
van Dorst DC, Dobbin SJ, Neves KB, Herrmann J, Herrmann SM, Versmissen J, Mathijssen RH, Danser AJ, Lang NN. Hypertension and Prohypertensive Antineoplastic Therapies in Cancer Patients. Circ Res 2021; 128:1040-1061. [PMID: 33793337 PMCID: PMC8011349 DOI: 10.1161/circresaha.121.318051] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The development of a wide range of novel antineoplastic therapies has improved the prognosis for patients with a wide range of malignancies, which has increased the number of cancer survivors substantially. Despite the oncological benefit, cancer survivors are exposed to short- and long-term adverse cardiovascular toxicities associated with anticancer therapies. Systemic hypertension, the most common comorbidity among cancer patients, is a major contributor to the increased risk for developing these adverse cardiovascular events. Cancer and hypertension have common risk factors, have overlapping pathophysiological mechanisms and hypertension may also be a risk factor for some tumor types. Many cancer therapies have prohypertensive effects. Although some of the mechanisms by which these antineoplastic agents lead to hypertension have been characterized, further preclinical and clinical studies are required to investigate the exact pathophysiology and the optimal management of hypertension associated with anticancer therapy. In this way, monitoring and management of hypertension before, during, and after cancer treatment can be improved to minimize cardiovascular risks. This is vital to optimize cardiovascular health in patients with cancer and survivors, and to ensure that advances in terms of cancer survivorship do not come at the expense of increased cardiovascular toxicities.
Collapse
Affiliation(s)
- Daan C.H. van Dorst
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (D.C.H.v.D., J.V., A.H.J.D.), Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Medical Oncology, Erasmus MC Cancer Institute (D.C.H.v.D., R.H.J.M.), Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Stephen J.H. Dobbin
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (S.J.H.D., K.B.N., N.N.L.)
| | - Karla B. Neves
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (S.J.H.D., K.B.N., N.N.L.)
| | - Joerg Herrmann
- Department of Cardiovascular Medicine (J.H.), Mayo Clinic, Rochester, MN
| | - Sandra M. Herrmann
- Division of Nephrology and Hypertension (S.M.H.), Mayo Clinic, Rochester, MN
| | - Jorie Versmissen
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (D.C.H.v.D., J.V., A.H.J.D.), Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Hospital Pharmacy (J.V.), Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Ron H.J. Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute (D.C.H.v.D., R.H.J.M.), Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - A.H. Jan Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (D.C.H.v.D., J.V., A.H.J.D.), Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Ninian N. Lang
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (S.J.H.D., K.B.N., N.N.L.)
| |
Collapse
|
95
|
Saunderson CED, Plein S, Manisty CH. Role of cardiovascular magnetic resonance imaging in cardio-oncology. Eur Heart J Cardiovasc Imaging 2021; 22:383-396. [PMID: 33404058 DOI: 10.1093/ehjci/jeaa345] [Citation(s) in RCA: 14] [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: 10/15/2020] [Accepted: 12/10/2020] [Indexed: 12/28/2022] Open
Abstract
Advances in cancer therapy have led to significantly longer cancer-free survival times over the last 40 years. Improved survivorship coupled with increasing recognition of an expanding range of adverse cardiovascular effects of many established and novel cancer therapies has highlighted the impact of cardiovascular disease in this population. This has led to the emergence of dedicated cardio-oncology services that can provide pre-treatment risk stratification, surveillance, diagnosis, and monitoring of cardiotoxicity during cancer therapies, and late effects screening following completion of treatment. Cardiovascular imaging and the development of imaging biomarkers that can accurately and reliably detect pre-clinical disease and enhance our understanding of the underlying pathophysiology of cancer treatment-related cardiotoxicity are becoming increasingly important. Multi-parametric cardiovascular magnetic resonance (CMR) is able to assess cardiac structure, function, and provide myocardial tissue characterization, and hence can be used to address a variety of important clinical questions in the emerging field of cardio-oncology. In this review, we discuss the current and potential future applications of CMR in the investigation and management of cancer patients.
Collapse
Affiliation(s)
- Christopher E D Saunderson
- Department of Biomedical Imaging Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Sven Plein
- Department of Biomedical Imaging Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Charlotte H Manisty
- Department of Cardio-Oncology, Barts Heart Centre, Barts Health NHS Trust, West Smithfield, London EC1A 7BE, UK
| |
Collapse
|
96
|
Martín García A, Mitroi C, Mazón Ramos P, García Sanz R, Virizuela JA, Arenas M, Egocheaga Cabello I, Albert D, Anguita Sánchez M, Arrarte Esteban VI, Ayala de la Peña F, Bonanand Lozano C, Castro A, Castro Fernández A, Córdoba R, Cosín-Sales J, Chaparro-Muñoz M, Dalmau R, Drak Hernández Y, Deiros Bronte L, Díez-Villanueva P, Escobar Cervantes C, Fernández Redondo C, García Rodríguez E, Lozano T, Marco Vera P, Martínez Monzonis A, Mesa D, Oristrell G, Palma Gámiz JL, Pedreira M, Reinoso-Barbero L, Rodríguez I, Serrano Antolín JM, Toral B, Torres Royo L, Velasco Del Castillo S, Vicente-Herrero T, Zatarain-Nicolás E, Tamargo J, López Fernández T. Stratification and management of cardiovascular risk in cancer patients. A consensus document of the SEC, FEC, SEOM, SEOR, SEHH, SEMG, AEEMT, AEEC, and AECC. ACTA ACUST UNITED AC 2021; 74:438-448. [PMID: 33712348 DOI: 10.1016/j.rec.2020.11.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/12/2020] [Indexed: 12/12/2022]
Abstract
Both cancer treatment and survival have significantly improved, but these advances have highlighted the deleterious effects of vascular complications associated with anticancer therapy. This consensus document aims to provide a coordinated, multidisciplinary and practical approach to the stratification, monitoring and treatment of cardiovascular risk in cancer patients. The document is promoted by the Working Group on Cardio Oncology of the Spanish Society of Cardiology (SEC) and was drafted in collaboration with experts from distinct areas of expertise of the SEC and the Spanish Society of Hematology and Hemotherapy (SEHH), the Spanish Society of Medical Oncology (SEOM), the Spanish Society of Radiation Oncology (SEOR), the Spanish Society of General and Family Physicians (SEMG), the Spanish Association of Specialists in Occupational Medicine (AEEMT), the Spanish Association of Cardiovascular Nursing (AEEC), the Spanish Heart Foundation (FEC), and the Spanish Cancer Association (AECC).
Collapse
Affiliation(s)
- Ana Martín García
- Servicio de Cardiología, Complejo Asistencial Universitario de Salamanca (CAUSA), IBSAL, USAL, Salamanca, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV-ISCIII), Spain.
| | - Cristina Mitroi
- Servicio de Cardiología, Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain
| | - Pilar Mazón Ramos
- Servicio de Cardiología, Hospital Clínico Universitario de Santiago, Santiago de Compostela, A Coruña, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV-ISCIII), Spain
| | - Ramón García Sanz
- Servicio de Hematología, Complejo Asistencial Universitario de Salamanca (CAUSA), IBSAL, Salamanca, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC-ISCIII), Spain
| | | | - Meritxell Arenas
- Servicio de Oncología Radioterápica, Hospital Universitario San Juan de Reus, Universidad Rovira i Virgili, Reus, Tarragona, Spain
| | | | - Dimpna Albert
- Servicio de Cardiología Pediátrica, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | | | | | | | | | - Almudena Castro
- Servicio de Cardiología, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV-ISCIII), Spain
| | | | - Raúl Córdoba
- Servicio de Hematología, Fundación Jiménez Díaz, Madrid, Spain
| | - Juan Cosín-Sales
- Servicio de Cardiología, Hospital Arnau de Vilanova, Valencia, Spain
| | | | - Regina Dalmau
- Servicio de Cardiología, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV-ISCIII), Spain
| | | | - Lucía Deiros Bronte
- Servicio de Cardiología Pediátrica, Hospital Universitario La Paz, Madrid, Spain
| | | | - Carlos Escobar Cervantes
- Servicio de Cardiología, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV-ISCIII), Spain
| | | | | | - Teresa Lozano
- Servicio de Cardiología, Hospital General Universitario de Alicante, ISABIAL, Alicante, Spain
| | - Pascual Marco Vera
- Servicio de Hematología y Hemoterapia, Hospital General Universitario de Alicante, ISABIAL, Alicante, Spain
| | - Amparo Martínez Monzonis
- Servicio de Cardiología, Hospital Clínico Universitario de Santiago, Santiago de Compostela, A Coruña, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV-ISCIII), Spain
| | - Dolores Mesa
- Servicio de Cardiología, Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Gerard Oristrell
- Servicio de Cardiología, Hospital Universitario Vall d'Hebron, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV-ISCIII), Spain
| | | | - Milagros Pedreira
- Servicio de Cardiología, Hospital Clínico Universitario de Santiago, Santiago de Compostela, A Coruña, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV-ISCIII), Spain
| | | | - Isabel Rodríguez
- Servicio de Oncología Radioterápica, Hospital Universitario La Paz, Madrid, Spain
| | | | - Belén Toral
- Servicio de Cardiología Pediátrica, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Laura Torres Royo
- Servicio de Oncología Radioterápica, Hospital Universitario San Juan de Reus, Universidad Rovira i Virgili, Reus, Tarragona, Spain
| | | | | | - Eduardo Zatarain-Nicolás
- Servicio de Cardiología, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Universidad Complutense, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV-ISCIII), Spain
| | - Juan Tamargo
- Departamento de Farmacología, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV-ISCIII), Spain
| | - Teresa López Fernández
- Servicio de Cardiología, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV-ISCIII), Spain
| |
Collapse
|
97
|
Bik E, Mateuszuk L, Orleanska J, Baranska M, Chlopicki S, Majzner K. Chloroquine-Induced Accumulation of Autophagosomes and Lipids in the Endothelium. Int J Mol Sci 2021; 22:ijms22052401. [PMID: 33673688 PMCID: PMC7957661 DOI: 10.3390/ijms22052401] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 01/14/2023] Open
Abstract
Chloroquine (CQ) is an antimalarial drug known to inhibit autophagy flux by impairing autophagosome–lysosome fusion. We hypothesized that autophagy flux altered by CQ has a considerable influence on the lipid composition of endothelial cells. Thus, we investigated endothelial responses induced by CQ on human microvascular endothelial cells (HMEC-1). HMEC-1 cells after CQ exposure were measured using a combined methodology based on label-free Raman and fluorescence imaging. Raman spectroscopy was applied to characterize subtle chemical changes in lipid contents and their distribution in the cells, while the fluorescence staining (LipidTox, LysoTracker and LC3) was used as a reference method. The results showed that CQ was not toxic to endothelial cells and did not result in the endothelial inflammation at concentrations of 1–30 µM. Notwithstanding, it yielded an increased intensity of LipidTox, LysoTracker, and LC3 staining, suggesting changes in the content of neutral lipids, lysosomotropism, and autophagy inhibition, respectively. The CQ-induced endothelial response was associated with lipid accumulation and was characterized by Raman spectroscopy. CQ-induced autophagosome accumulation in the endothelium is featured by a pronounced alteration in the lipid profile, but not in the endothelial inflammation. Raman-based assessment of CQ-induced biochemical changes offers a better understanding of the autophagy mechanism in the endothelial cells.
Collapse
Affiliation(s)
- Ewelina Bik
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland; (E.B.); (L.M.); (J.O.); (M.B.); (S.C.)
- Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Str., 30-387 Krakow, Poland
| | - Lukasz Mateuszuk
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland; (E.B.); (L.M.); (J.O.); (M.B.); (S.C.)
| | - Jagoda Orleanska
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland; (E.B.); (L.M.); (J.O.); (M.B.); (S.C.)
| | - Malgorzata Baranska
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland; (E.B.); (L.M.); (J.O.); (M.B.); (S.C.)
- Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Str., 30-387 Krakow, Poland
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland; (E.B.); (L.M.); (J.O.); (M.B.); (S.C.)
- Chair of Pharmacology, Jagiellonian University, 16 Grzegorzecka Str., 31-531 Krakow, Poland
| | - Katarzyna Majzner
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland; (E.B.); (L.M.); (J.O.); (M.B.); (S.C.)
- Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Str., 30-387 Krakow, Poland
- Correspondence:
| |
Collapse
|
98
|
Galán-Arriola C, Vílchez-Tschischke JP, Lobo M, López GJ, de Molina-Iracheta A, Pérez-Martínez C, Villena-Gutiérrez R, Macías Á, Díaz-Rengifo IA, Oliver E, Fuster V, Sánchez-González J, Ibanez B. Coronary microcirculation damage in anthracycline cardiotoxicity. Cardiovasc Res 2021; 118:531-541. [PMID: 33605403 PMCID: PMC8803079 DOI: 10.1093/cvr/cvab053] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/28/2020] [Accepted: 02/17/2021] [Indexed: 01/19/2023] Open
Abstract
AIMS The aim of this study was to study changes in coronary microcirculation status during and after several cycles of anthracycline treatment. METHODS AND RESULTS Large-White male pigs (n = 40) were included in different experimental protocols (ExPr.) according to anthracycline cumulative exposure (0.45 mg/kg intracoronary (IC) doxorubicin per injection) and follow-up: Control (no doxorubicin); Single injection and sacrifice either at 48 hours (ExPr. 1) or 2 weeks (ExPr. 2); Three injections two weeks apart (low cumulative dose) and sacrifice either 2 weeks (ExPr. 3) or 12 weeks (ExPr. 4) after third injection; Five injections two weeks apart (high cumulative dose) and sacrifice 8 weeks after fifth injection (ExPr. 5). All groups were assessed by serial cardiac magnetic resonance (CMR) to quantify perfusion and invasive measurement of coronary flow reserve (CFR). At the end of each protocol, animals were sacrificed for ex vivo analyses. Vascular function was further evaluated by myography in explanted coronary arteries of pigs undergoing ExPr. 3 and controls.A single doxorubicin injection had no impact on microcirculation status, excluding a direct chemical toxicity. A series of five fortnightly doxorubicin injections (high cumulative dose) triggered a progressive decline in microcirculation status, evidenced by reduced CMR-based myocardial perfusion and CFR-measured impaired functional microcirculation. In the high cumulative dose regime (ExPr. 5), microcirculation changes appeared long before any contractile defect became apparent. Low cumulative doxorubicin dose (3 biweekly injections) was not associated with any contractile defect across long-term follow-up, but provoked persistent microcirculation damage, evident soon after third dose injection. Histological and myograph evaluations confirmed structural damage to arteries of all calibers even in animals undergoing low cumulative dose regimes. Conversely, arteriole damage and capillary bed alteration occurred only after high cumulative dose regime. CONCLUSION Serial in vivo evaluations of microcirculation status using state-of-the-art CMR and invasive CFR show that anthracyclines treatment is associated with progressive and irreversible damage to the microcirculation. This long-persisting damage is present even in low cumulative dose regimes, which are not associated with cardiac contractile deficits. Microcirculation damage might explain some of the increased incidence of cardiovascular events in cancer survivors who received anthracyclines without showing cardiac contractile defects.
Collapse
Affiliation(s)
- Carlos Galán-Arriola
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Jean Paul Vílchez-Tschischke
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Complejo Hospitalario Ruber Juan Bravo, Madrid, Spain
| | - Manuel Lobo
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Complejo Hospitalario Ruber Juan Bravo, Madrid, Spain
| | - Gonzalo J López
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | | | - Rocio Villena-Gutiérrez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Spain
| | | | | | - Eduardo Oliver
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Valentin Fuster
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York
| | | | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Spain.,Cardiology Department, IIS-Fundación Jiménez Díaz Hospital, Madrid
| |
Collapse
|
99
|
Kim KH, Choi S, Kim K, Chang J, Kim SM, Kim SR, Cho Y, Oh YH, Lee G, Son JS, Park SM. Association between physical activity and subsequent cardiovascular disease among 5-year breast cancer survivors. Breast Cancer Res Treat 2021; 188:203-214. [PMID: 33599866 DOI: 10.1007/s10549-021-06140-8] [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: 08/17/2020] [Accepted: 02/06/2021] [Indexed: 12/18/2022]
Abstract
PURPOSE To examine the association of physical activity among long-term breast cancer survivors on the occurrence of subsequent cardiovascular disease (CVD). METHODS We investigated the risk of CVD among 39,775 breast cancer patients who were newly diagnosed in 2006 and survived until 2011 within the Korean National Health Insurance Service database. Patients were followed up from 5 years after breast cancer diagnosis to the date of CVD event, death, or December 31, 2018, whichever came earliest. Every 500 MET-mins/week correspond to 152, 125, and 62.5 min per week of light-, moderate-, and vigorous-intensity physical activity, respectively. Adjusted hazard ratios (aHRs) and 95% confidence intervals (CIs) for CVD were calculated using Cox proportional hazards regression by physical activity levels. RESULTS Compared with those with physical activity of 0 MET-min/week, those with 1-499 (aHR 0.82, 95% CI 0.69-0.98), 500-999 (aHR 0.75, 95% CI 0.63-0.90), and ≥ 1,000 (aHR 0.76, 95% CI 0.63-0.93) MET-min/week of PA had lower risk of CVD. Higher levels of PA were associated with lower risk of stroke (p for trend = 0.016). The benefits of PA on obese and overweight breast cancer survivors were smaller than those in normal weight survivors. The frequency of moderate-to-vigorous physical activity (MVPA) showed a reverse J-curve association with CVD, and the best benefit occurred in the 3-4 times MVPA per week group (aHR 0.59, 95% CI 0.46-0.74). CONCLUSIONS The study showed that even small amounts of PA may be beneficial in potentially decreasing the risk of CVD, CHD, and stroke in breast cancer survivors. Our result will be useful to prescribe and delivery exercise among long-term breast cancer survivors.
Collapse
Affiliation(s)
- Kyae Hyung Kim
- Department of Family Medicine, Seoul National University Hospital, Seoul, South Korea.,Comprehensive Care Clinic, Seoul National University Hospital, Seoul, South Korea
| | - Seulggie Choi
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, South Korea
| | - Kyuwoong Kim
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, South Korea
| | - Jooyoung Chang
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, South Korea
| | - Sung Min Kim
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, South Korea
| | - Seong Rae Kim
- College of Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Yoosun Cho
- Department of Family Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Yun Hwan Oh
- Department of Family Medicine, Jeju National University School of Medicine, Jeju National University Hospital, Jeju, South Korea
| | - Gyeongsil Lee
- Department of Family Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Joung Sik Son
- Department of Family Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Sang Min Park
- Department of Family Medicine, Seoul National University Hospital, Seoul, South Korea. .,Department of Biomedical Sciences, Seoul National University Graduate School, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, South Korea.
| |
Collapse
|
100
|
Guha A, Dey AK, Al-Kindi S, Miller PE, Ghosh AK, Banerjee A, Lopez-Mattei J, Desai NR, Patel B, Oliveira GH, de Lima M, Fradley M, Addison D. Socio-Economic Burden of Myocardial Infarction Among Cancer Patients. Am J Cardiol 2021; 141:16-22. [PMID: 33217349 DOI: 10.1016/j.amjcard.2020.11.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/31/2020] [Accepted: 11/03/2020] [Indexed: 01/25/2023]
Abstract
Cancer patients face a higher risk of future myocardial infarction (MI), even after completion of anticancer therapies. MI is a critical source of physical and financial stress in noncancer patients, but its impacts associated with cancer patients also saddled with the worry (stress) of potential reoccurrence is unknown. Therefore, we aimed to quantify MI's stress and financial burden after surviving cancer and compare to those never diagnosed with cancer. Utilizing cross-sectional national survey data from 2013 to 2018 derived from publicly available United States datasets, the National Health Interview Survey , and economic data from the National Inpatient Sample , we compared the socio-economic outcomes in those with MI by cancer-status. We adjusted for social, demographic, and clinical factors. Overall, 19,504 (10.2%) of the 189,836 National Health Interview Survey responders reported having cancer for more than 1 year. There was an increased prevalence of MI in cancer survivors compared with noncancer patients (8.8% vs 3.2%, p <0.001). MI was associated with increased financial worry, food insecurity, and financial burden of medical bills (p <0.001, respectively); however, concurrent cancer did not seem to be an effect modifier (p >0.05). There was no difference in annual residual family income by cancer status; however, 3 lowest deciles of residual income representing 21.1% cancer-survivor with MI had a residual income of <$9,000. MI continues to represent an immense source of financial and perceived stress. In conclusion, although cancer patients face a higher risk of subsequent MI, this does not appear to advance their reported stress significantly.
Collapse
Affiliation(s)
- Avirup Guha
- Harrington Heart and Vascular Institute, Case Western Reserve University, Cleveland, Ohio; Cardio-Oncology Program, Division of Cardiology, The Ohio State University Medical Center, Columbus, Ohio
| | - Amit Kumar Dey
- National Heart, Lung and Blood Institute, Bethesda, Maryland
| | - Sadeer Al-Kindi
- Harrington Heart and Vascular Institute, Case Western Reserve University, Cleveland, Ohio
| | - P Elliott Miller
- Division of Cardiology, Yale University School of Medicine, New Haven, Connecticut; Yale National Clinician Scholars Program, New Haven, Connecticut
| | - Arjun K Ghosh
- Cardio-Oncology Service, Bart's Heart Centre and University College London Hospital, Hatter Cardiovascular Institute, London, United Kingdom
| | - Amitava Banerjee
- Institute of Health Informatics, University College London, London, United Kingdom
| | - Juan Lopez-Mattei
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nihar R Desai
- Division of Cardiology, Yale University School of Medicine, New Haven, Connecticut; Center for Outcomes Research and Evaluation, New Haven, Connecticut
| | - Brijesh Patel
- Division of Cardiology, West Virginia University, Morgantown, West Virginia
| | - Guilherme H Oliveira
- Cardio-Oncology Program, Division of Cardiovascular Medicine, University of South Florida Morsani College of Medicine and H. Lee Moffitt Cancer Center and Research Institute and, Tampa, Florida
| | - Marcos de Lima
- Adult Hematologic Malignancies and Stem Cell Transplant Program, Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Michael Fradley
- Cardio-Oncology Program, Division of Cardiovascular Medicine, University of Pennsylvania, Philadelphia, Pennysylvania
| | - Daniel Addison
- Cardio-Oncology Program, Division of Cardiology, The Ohio State University Medical Center, Columbus, Ohio; Cancer Control Program, Department of Internal Medicine, Ohio State University Comprehensive Cancer Center, Columbus, Ohio.
| |
Collapse
|