18 results on '"Moslehi J"'
Search Results
2. Sex-Specific Cardiovascular Risks of Cancer and Its Therapies.
- Author
-
Wilcox NS, Rotz SJ, Mullen M, Song EJ, Ky Hamilton B, Moslehi J, Armenian SH, Wu JC, Rhee JW, and Ky B
- Subjects
- Anthracyclines adverse effects, Cardiotoxicity diagnosis, Cardiotoxicity epidemiology, Cardiovascular Diseases diagnosis, Female, Hematopoietic Stem Cell Transplantation adverse effects, Hormone Replacement Therapy adverse effects, Humans, Immunotherapy adverse effects, Male, Neoplasms drug therapy, Antineoplastic Agents adverse effects, Cardiovascular Diseases chemically induced, Cardiovascular Diseases epidemiology, Heart Disease Risk Factors, Neoplasms epidemiology, Sex Characteristics
- Abstract
In both cardiovascular disease and cancer, there are established sex-based differences in prevalence and outcomes. Males and females may also differ in terms of risk of cardiotoxicity following cancer therapy, including heart failure, cardiomyopathy, atherosclerosis, thromboembolism, arrhythmias, and myocarditis. Here, we describe sex-based differences in the epidemiology and pathophysiology of cardiotoxicity associated with anthracyclines, hematopoietic stem cell transplant (HCT), hormone therapy and immune therapy. Relative to males, the risk of anthracycline-induced cardiotoxicity is higher in prepubertal females, lower in premenopausal females, and similar in postmenopausal females. For autologous hematopoietic cell transplant, several studies suggest an increased risk of late heart failure in female lymphoma patients, but sex-based differences have not been shown for allogeneic hematopoietic cell transplant. Hormone therapies including GnRH (gonadotropin-releasing hormone) modulators, androgen receptor antagonists, selective estrogen receptor modulators, and aromatase inhibitors are associated with cardiotoxicity, including arrhythmia and venous thromboembolism. However, sex-based differences have not yet been elucidated. Evaluation of sex differences in cardiotoxicity related to immune therapy is limited, in part, due to low participation of females in relevant clinical trials. However, some studies suggest that females are at increased risk of immune checkpoint inhibitor myocarditis, although this has not been consistently demonstrated. For each of the aforementioned cancer therapies, we consider sex-based differences according to cardiotoxicity management. We identify knowledge gaps to guide future mechanistic and prospective clinical studies. Furthering our understanding of sex-based differences in cancer therapy cardiotoxicity can advance the development of targeted preventive and therapeutic cardioprotective strategies.
- Published
- 2022
- Full Text
- View/download PDF
3. Identification of anticancer drugs associated with atrial fibrillation: analysis of the WHO pharmacovigilance database.
- Author
-
Alexandre J, Salem JE, Moslehi J, Sassier M, Ropert C, Cautela J, Thuny F, Ederhy S, Cohen A, Damaj G, Vilque JP, Plane AF, Legallois D, Champ-Rigot L, Milliez P, Funck-Brentano C, and Dolladille C
- Subjects
- Adverse Drug Reaction Reporting Systems, Humans, Pharmacovigilance, Prospective Studies, United States, World Health Organization, Antineoplastic Agents adverse effects, Atrial Fibrillation diagnosis, Atrial Fibrillation drug therapy, Atrial Fibrillation epidemiology
- Abstract
Aims: The explosion of novel anticancer therapies has meant emergence of cardiotoxicity signals including atrial fibrillation (AF). Reliable data concerning the liability of anticancer drugs in inducing AF are scarce. Using the World Health Organization individual case safety report database, VigiBase®, we aimed to determine the association between anticancer drugs and AF., Methods and Results: A disproportionality analysis evaluating the multivariable-adjusted reporting odds ratios for AF with their 99.97% confidence intervals was performed for 176 U.S. Food and Drug Administration (FDA)- or European Medicines Agency (EMA)-labelled anticancer drugs in VigiBase®, followed by a descriptive analysis of AF cases for the anticancer drugs identified in VigiBase®. ClinicalTrial registration number: NCT03530215. A total of 11 757 AF cases associated with at least one anticancer drug were identified in VigiBase® of which 95.8% were deemed serious. Nineteen anticancer drugs were significantly associated with AF of which 14 (74%) are used in haematologic malignancies and 9 (45%) represented new AF associations not previously confirmed in literature including immunomodulating agents (lenalidomide, pomalidomide), several kinase inhibitors (nilotinib, ponatinib, midostaurin), antimetabolites (azacytidine, clofarabine), docetaxel (taxane), and obinutuzumab, an anti-CD20 monoclonal antibody., Conclusion: Although cancer malignancy itself may generate AF, we identified 19 anticancer drugs significantly associated with a significant increase in AF over-reporting. This pharmacovigilance study provides evidence that anticancer drugs themselves could represent independent risk factors for AF development. Dedicated prospective clinical trials are now required to confirm these 19 associations. This list of suspected anticancer drugs should be known by physicians when confronted to AF in cancer patients, particularly in case of haematologic malignancies., (Published on behalf of the European Society of Cardiology. All rights reserved. © The Author(s) 2020. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2021
- Full Text
- View/download PDF
4. Ibrutinib-Mediated Atrial Fibrillation Attributable to Inhibition of C-Terminal Src Kinase.
- Author
-
Xiao L, Salem JE, Clauss S, Hanley A, Bapat A, Hulsmans M, Iwamoto Y, Wojtkiewicz G, Cetinbas M, Schloss MJ, Tedeschi J, Lebrun-Vignes B, Lundby A, Sadreyev RI, Moslehi J, Nahrendorf M, Ellinor PT, and Milan DJ
- Subjects
- Action Potentials drug effects, Adenine toxicity, Agammaglobulinaemia Tyrosine Kinase deficiency, Agammaglobulinaemia Tyrosine Kinase genetics, Animals, Atrial Fibrillation enzymology, Atrial Fibrillation physiopathology, CSK Tyrosine-Protein Kinase genetics, CSK Tyrosine-Protein Kinase metabolism, Databases, Genetic, Heart Atria enzymology, Heart Atria physiopathology, Humans, Mice, 129 Strain, Mice, Inbred C57BL, Mice, Inbred CBA, Mice, Knockout, Risk Assessment, Risk Factors, Adenine analogs & derivatives, Antineoplastic Agents toxicity, Atrial Fibrillation chemically induced, Atrial Function, Left drug effects, CSK Tyrosine-Protein Kinase antagonists & inhibitors, Heart Atria drug effects, Heart Rate drug effects, Piperidines toxicity, Protein Kinase Inhibitors toxicity
- Abstract
Background: Ibrutinib is a Bruton tyrosine kinase inhibitor with remarkable efficacy against B-cell cancers. Ibrutinib also increases the risk of atrial fibrillation (AF), which remains poorly understood., Methods: We performed electrophysiology studies on mice treated with ibrutinib to assess inducibility of AF. Chemoproteomic analysis of cardiac lysates identified candidate ibrutinib targets, which were further evaluated in genetic mouse models and additional pharmacological experiments. The pharmacovigilance database, VigiBase, was queried to determine whether drug inhibition of an identified candidate kinase was associated with increased reporting of AF., Results: We demonstrate that treatment of mice with ibrutinib for 4 weeks results in inducible AF, left atrial enlargement, myocardial fibrosis, and inflammation. This effect was reproduced in mice lacking Bruton tyrosine kinase, but not in mice treated with 4 weeks of acalabrutinib, a more specific Bruton tyrosine kinase inhibitor, demonstrating that AF is an off-target side effect. Chemoproteomic profiling identified a short list of candidate kinases that was narrowed by additional experimentation leaving CSK (C-terminal Src kinase) as the strongest candidate for ibrutinib-induced AF. Cardiac-specific Csk knockout in mice led to increased AF, left atrial enlargement, fibrosis, and inflammation, phenocopying ibrutinib treatment. Disproportionality analyses in VigiBase confirmed increased reporting of AF associated with kinase inhibitors blocking Csk versus non-Csk inhibitors, with a reporting odds ratio of 8.0 (95% CI, 7.3-8.7; P <0.0001)., Conclusions: These data identify Csk inhibition as the mechanism through which ibrutinib leads to AF. Registration: URL: https://ww.clinicaltrials.gov; Unique identifier: NCT03530215.
- Published
- 2020
- Full Text
- View/download PDF
5. The cancer patient and cardiology.
- Author
-
Zamorano JL, Gottfridsson C, Asteggiano R, Atar D, Badimon L, Bax JJ, Cardinale D, Cardone A, Feijen EAM, Ferdinandy P, López-Fernández T, Gale CP, Maduro JH, Moslehi J, Omland T, Plana Gomez JC, Scott J, Suter TM, and Minotti G
- Subjects
- Aftercare, Antineoplastic Agents therapeutic use, Humans, Risk Assessment, Risk Factors, Antineoplastic Agents adverse effects, Cardiotoxicity diagnosis, Cardiotoxicity etiology, Cardiotoxicity prevention & control, Cardiotoxicity therapy, Neoplasms drug therapy, Neoplasms radiotherapy, Radiotherapy adverse effects
- Abstract
Advances in cancer treatments have improved clinical outcomes, leading to an increasing population of cancer survivors. However, this success is associated with high rates of short- and long-term cardiovascular (CV) toxicities. The number and variety of cancer drugs and CV toxicity types make long-term care a complex undertaking. This requires a multidisciplinary approach that includes expertise in oncology, cardiology and other related specialties, and has led to the development of the cardio-oncology subspecialty. This paper aims to provide an overview of the main adverse events, risk assessment and risk mitigation strategies, early diagnosis, medical and complementary strategies for prevention and management, and long-term follow-up strategies for patients at risk of cancer therapy-related cardiotoxicities. Research to better define strategies for early identification, follow-up and management is highly necessary. Although the academic cardio-oncology community may be the best vehicle to foster awareness and research in this field, additional stakeholders (industry, government agencies and patient organizations) must be involved to facilitate cross-discipline interactions and help in the design and funding of cardio-oncology trials. The overarching goals of cardio-oncology are to assist clinicians in providing optimal care for patients with cancer and cancer survivors, to provide insight into future areas of research and to search for collaborations with industry, funding bodies and patient advocates. However, many unmet needs remain. This document is the product of brainstorming presentations and active discussions held at the Cardiovascular Round Table workshop organized in January 2020 by the European Society of Cardiology., (© 2020 European Society of Cardiology.)
- Published
- 2020
- Full Text
- View/download PDF
6. Role of serum biomarkers in cancer patients receiving cardiotoxic cancer therapies: a position statement from the Cardio-Oncology Study Group of the Heart Failure Association and the Cardio-Oncology Council of the European Society of Cardiology.
- Author
-
Pudil R, Mueller C, Čelutkienė J, Henriksen PA, Lenihan D, Dent S, Barac A, Stanway S, Moslehi J, Suter TM, Ky B, Štěrba M, Cardinale D, Cohen-Solal A, Tocchetti CG, Farmakis D, Bergler-Klein J, Anker MS, Von Haehling S, Belenkov Y, Iakobishvili Z, Maack C, Ciardiello F, Ruschitzka F, Coats AJS, Seferovic P, Lainscak M, Piepoli MF, Chioncel O, Bax J, Hulot JS, Skouri H, Hägler-Laube ES, Asteggiano R, Fernandez TL, de Boer RA, and Lyon AR
- Subjects
- Biomarkers, Tumor blood, Cardiotonic Agents administration & dosage, Humans, Antineoplastic Agents administration & dosage, Antineoplastic Agents adverse effects, Cardiotoxicity blood, Cardiotoxicity diagnosis, Cardiotoxicity etiology, Heart Failure blood, Heart Failure chemically induced, Heart Failure diagnosis, Neoplasms blood, Neoplasms drug therapy
- Abstract
Serum biomarkers are an important tool in the baseline risk assessment and diagnosis of cardiovascular disease in cancer patients receiving cardiotoxic cancer treatments. Increases in cardiac biomarkers including cardiac troponin and natriuretic peptides can be used to guide initiation of cardioprotective treatments for cancer patients during treatment and to monitor the response to cardioprotective treatments, and they also offer prognostic value. This position statement examines the role of cardiac biomarkers in the management of cancer patients. The Cardio-Oncology Study Group of the Heart Failure Association (HFA) of the European Society of Cardiology (ESC) in collaboration with the Cardio-Oncology Council of the ESC have evaluated the current evidence for the role of cardiovascular biomarkers in cancer patients before, during and after cardiotoxic cancer therapies. The characteristics of the main two biomarkers troponin and natriuretic peptides are discussed, the link to the mechanisms of cardiovascular toxicity, and the evidence for their clinical use in surveillance during and after anthracycline chemotherapy, trastuzumab and HER2-targeted therapies, vascular endothelial growth factor inhibitors, proteasome inhibitors, immune checkpoint inhibitors, cyclophosphamide and radiotherapy. Novel surveillance clinical pathways integrating cardiac biomarkers for cancer patients receiving anthracycline chemotherapy or trastuzumab biomarkers are presented and future direction in cardio-oncology biomarker research is discussed., (© 2020 European Society of Cardiology.)
- Published
- 2020
- Full Text
- View/download PDF
7. Vascular medicine and cardio-oncology - A new, evolving clinical frontier.
- Author
-
Versmissen J, Power JR, and Moslehi J
- Subjects
- Heart Diseases diagnosis, Heart Diseases epidemiology, Heart Diseases therapy, Humans, Neoplasms diagnosis, Neoplasms epidemiology, Prognosis, Risk Assessment, Risk Factors, Antineoplastic Agents adverse effects, Cancer Survivors, Cardiology trends, Heart Diseases chemically induced, Medical Oncology trends, Neoplasms drug therapy, Specialization trends
- Published
- 2020
- Full Text
- View/download PDF
8. Management of cardiac disease in cancer patients throughout oncological treatment: ESMO consensus recommendations.
- Author
-
Curigliano G, Lenihan D, Fradley M, Ganatra S, Barac A, Blaes A, Herrmann J, Porter C, Lyon AR, Lancellotti P, Patel A, DeCara J, Mitchell J, Harrison E, Moslehi J, Witteles R, Calabro MG, Orecchia R, de Azambuja E, Zamorano JL, Krone R, Iakobishvili Z, Carver J, Armenian S, Ky B, Cardinale D, Cipolla CM, Dent S, and Jordan K
- Subjects
- Humans, Consensus, Medical Oncology, Antineoplastic Agents adverse effects, Heart Diseases chemically induced, Heart Diseases epidemiology, Neoplasms complications, Neoplasms drug therapy, Neoplasms epidemiology
- Abstract
Cancer and cardiovascular (CV) disease are the most prevalent diseases in the developed world. Evidence increasingly shows that these conditions are interlinked through common risk factors, coincident in an ageing population, and are connected biologically through some deleterious effects of anticancer treatment on CV health. Anticancer therapies can cause a wide spectrum of short- and long-term cardiotoxic effects. An explosion of novel cancer therapies has revolutionised this field and dramatically altered cancer prognosis. Nevertheless, these new therapies have introduced unexpected CV complications beyond heart failure. Common CV toxicities related to cancer therapy are defined, along with suggested strategies for prevention, detection and treatment. This ESMO consensus article proposes to define CV toxicities related to cancer or its therapies and provide guidance regarding prevention, screening, monitoring and treatment of CV toxicity. The majority of anticancer therapies are associated with some CV toxicity, ranging from asymptomatic and transient to more clinically significant and long-lasting cardiac events. It is critical however, that concerns about potential CV damage resulting from anticancer therapies should be weighed against the potential benefits of cancer therapy, including benefits in overall survival. CV disease in patients with cancer is complex and treatment needs to be individualised. The scope of cardio-oncology is wide and includes prevention, detection, monitoring and treatment of CV toxicity related to cancer therapy, and also ensuring the safe development of future novel cancer treatments that minimise the impact on CV health. It is anticipated that the management strategies discussed herein will be suitable for the majority of patients. Nonetheless, the clinical judgment of physicians remains extremely important; hence, when using these best clinical practices to inform treatment options and decisions, practitioners should also consider the individual circumstances of their patients on a case-by-case basis., (Copyright © 2019 European Society for Medical Oncology. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
9. Mechanisms and clinical course of cardiovascular toxicity of cancer treatment I. Oncology.
- Author
-
Yeh ETH, Ewer MS, Moslehi J, Dlugosz-Danecka M, Banchs J, Chang HM, and Minotti G
- Subjects
- Antineoplastic Agents therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Cardiotoxicity, Cardiovascular Diseases diagnosis, DNA Topoisomerases, Type II, Humans, Medical Oncology, Molecular Targeted Therapy adverse effects, Molecular Targeted Therapy methods, Neoplasms therapy, Poly-ADP-Ribose Binding Proteins, Antineoplastic Agents adverse effects, Antineoplastic Combined Chemotherapy Protocols adverse effects, Cardiovascular Diseases etiology, Neoplasms complications
- Abstract
The opening session of Second International Colloquium on Cardio-Oncology addressed two areas of vital interest. The first reviewed new thoughts related to established agents. While anthracycline cardiotoxicity has been studied and reviewed extensively, ongoing research attempting to understand why it appears the mechanism(s) of toxicity differs from that of oncologic efficacy continue to evoke comment and intriguing speculation. Better understanding of the role of β-topoisomerase II in toxicity has advanced our understanding of the cascade of events that lead to heart failure. Additionally, the cardioprotective role of dexrazoxane fits well with our new understanding of how β-topoisomerase II works. Beyond the anthracyclines, new insight is providing us insight to better understand the impact on cardiac function seen with other agents including those targeting HER2 and several tyrosine-kinase inhibitors. Unlike the anthracyclines, these agents affect cardiac function in ways that are less direct, and therefore have different characteristics and should be thought of in alternate ways. This new knowledge regarding established agents furthers our understanding of the spectrum of cardiotoxicity and cardiac dysfunction in the cancer patient. The session also addressed cardiovascular toxicities of newer and established agents beyond myocardial dysfunction including effects on the vasculature. These agents cause changes that may be temporary or permanent, and that range from subclinical to life-threatening. The session ended with a discussion of the cardiac effects of immune checkpoint inhibitors. These agents can cause rare and sometimes fatal cardiac inflammation, for which long-term follow up may be required., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
10. Cancer therapy-induced cardiomyopathy: can human induced pluripotent stem cell modelling help prevent it?
- Author
-
Stack JP, Moslehi J, Sayed N, and Wu JC
- Subjects
- Animals, Antineoplastic Agents therapeutic use, Cardiotoxicity, Cardiotoxins adverse effects, Genomics, Humans, Mice, Neoplasms drug therapy, Precision Medicine, Antineoplastic Agents adverse effects, Cardiomyopathies chemically induced, Drug Evaluation, Preclinical methods, Induced Pluripotent Stem Cells cytology, Induced Pluripotent Stem Cells drug effects, Models, Biological
- Abstract
Cardiotoxic effects from cancer therapy are a major cause of morbidity during cancer treatment. Unexpected toxicity can occur during treatment and/or after completion of therapy, into the time of cancer survivorship. While older drugs such as anthracyclines have well-known cardiotoxic effects, newer drugs such as tyrosine kinase inhibitors, proteasome inhibitors, and immunotherapies also can cause diverse cardiovascular and metabolic complications. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are increasingly being used as instruments for disease modelling, drug discovery, and mechanistic toxicity studies. Promising results with hiPSC-CM chemotherapy studies are raising hopes for improving cancer therapies through personalized medicine and safer drug development. Here, we review the cardiotoxicity profiles of common chemotherapeutic agents as well as efforts to model them in vitro using hiPSC-CMs., (Published on behalf of the European Society of Cardiology. All rights reserved. © The Author(s) 2018. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2019
- Full Text
- View/download PDF
11. Cardio-oncology: a novel platform for basic and translational cardiovascular investigation driven by clinical need.
- Author
-
Moslehi J, Fujiwara K, and Guzik T
- Subjects
- Animals, Cardiotoxicity, Cardiovascular Diseases diagnosis, Cardiovascular Diseases pathology, Cardiovascular Diseases therapy, Diffusion of Innovation, Forecasting, Humans, Risk Factors, Antineoplastic Agents adverse effects, Cardiology trends, Cardiovascular Diseases chemically induced, Medical Oncology trends, Translational Research, Biomedical trends
- Published
- 2019
- Full Text
- View/download PDF
12. Characterization of atrial fibrillation adverse events reported in ibrutinib randomized controlled registration trials.
- Author
-
Brown JR, Moslehi J, O'Brien S, Ghia P, Hillmen P, Cymbalista F, Shanafelt TD, Fraser G, Rule S, Kipps TJ, Coutre S, Dilhuydy MS, Cramer P, Tedeschi A, Jaeger U, Dreyling M, Byrd JC, Howes A, Todd M, Vermeulen J, James DF, Clow F, Styles L, Valentino R, Wildgust M, Mahler M, and Burger JA
- Subjects
- Adenine analogs & derivatives, Adult, Aged, Aged, 80 and over, Antineoplastic Agents therapeutic use, Atrial Fibrillation epidemiology, Atrial Fibrillation therapy, Disease Management, Female, Follow-Up Studies, Hemorrhage etiology, Humans, Incidence, Male, Middle Aged, Piperidines, Protein Kinase Inhibitors therapeutic use, Pyrazoles therapeutic use, Pyrimidines therapeutic use, Randomized Controlled Trials as Topic, Risk Factors, Time Factors, Antineoplastic Agents adverse effects, Atrial Fibrillation diagnosis, Atrial Fibrillation etiology, Protein Kinase Inhibitors adverse effects, Pyrazoles adverse effects, Pyrimidines adverse effects
- Abstract
The first-in-class Bruton's tyrosine kinase inhibitor ibrutinib has proven clinical benefit in B-cell malignancies; however, atrial fibrillation (AF) has been reported in 6-16% of ibrutinib patients. We pooled data from 1505 chronic lymphocytic leukemia and mantle cell lymphoma patients enrolled in four large, randomized, controlled studies to characterize AF with ibrutinib and its management. AF incidence was 6.5% [95% Confidence Interval (CI): 4.8, 8.5] for ibrutinib at 16.6-months versus 1.6% (95%CI: 0.8, 2.8) for comparator and 10.4% (95%CI: 8.4, 12.9) at the 36-month follow up; estimated cumulative incidence: 13.8% (95%CI: 11.2, 16.8). Ibrutinib treatment, prior history of AF and age 65 years or over were independent risk factors for AF. Multiple AF events were more common with ibrutinib (44.9%; comparator, 16.7%) among patients with AF. Most (85.7%) patients with AF did not discontinue ibrutinib, and more than half received common anticoagulant/antiplatelet medications on study. Low-grade bleeds were more frequent with ibrutinib, but serious bleeds were uncommon (ibrutinib, 2.9%; comparator, 2.0%). Although the AF rate among older non-trial patients with comorbidities is likely underestimated by this dataset, these results suggest that AF among clinical trial patients is generally manageable without ibrutinib discontinuation ( clinicaltrials.gov identifier: 01578707, 01722487, 01611090, 01646021 )., (Copyright© 2017 Ferrata Storti Foundation.)
- Published
- 2017
- Full Text
- View/download PDF
13. Beyond Anthracyclines: Preemptive Management of Cardiovascular Toxicity in the Era of Targeted Agents for Hematologic Malignancies.
- Author
-
Sethi TK, Basdag B, Bhatia N, Moslehi J, and Reddy NM
- Subjects
- Animals, Anthracyclines therapeutic use, Antineoplastic Agents therapeutic use, Cardiotoxicity prevention & control, Cardiovascular Diseases prevention & control, Cardiovascular Diseases therapy, Disease Management, Drug Evaluation, Preclinical, Hematologic Neoplasms drug therapy, Hematologic Neoplasms etiology, Humans, Protein Kinase Inhibitors adverse effects, Protein Kinase Inhibitors therapeutic use, Anthracyclines adverse effects, Antineoplastic Agents adverse effects, Cardiovascular Diseases diagnosis, Cardiovascular Diseases etiology, Hematologic Neoplasms complications, Molecular Targeted Therapy adverse effects
- Abstract
Advances in drug discovery have led to the use of effective targeted agents in the treatment of hematologic malignancies. Drugs such as proteasome inhibitors in multiple myeloma and tyrosine kinase inhibitors in chronic myeloid leukemia and non-Hodgkin lymphoma have changed the face of treatment of hematologic malignancies. There are several reports of cardiovascular adverse events related to these newer agents. Both "on-target" and "off-target" effects of these agents can cause organ-specific toxicity. The need for long-term administration for most of these agents requires continued monitoring of toxicity. Moreover, the patient population is older, often over 50 years of age, making them more susceptible to cardiovascular side effects. Additional factors such as prior exposure to anthracyclines often add to this toxicity. In light of their success and widespread use, it is important to recognize and manage the unique side effect profile of targeted agents used in hematologic malignancies. In this article, we review the current data for the incidence of cardiovascular side effects of targeted agents in hematologic malignancies and discuss a preemptive approach towards managing these toxicities.
- Published
- 2017
- Full Text
- View/download PDF
14. QT Prolongation and Oncology Drug Development.
- Author
-
Fradley MG and Moslehi J
- Subjects
- Electrocardiography, Humans, Antineoplastic Agents adverse effects, Cardiotoxins adverse effects, Long QT Syndrome, Torsades de Pointes
- Abstract
Many pharmaceutical agents interact with cardiac ion channels resulting in abnormal ventricular repolarization and prolongation of the QT interval. In rare circumstances, this has resulted in the development of the potentially life-threatening arrhythmia, torsades de pointes. It is recognized, however, that accurate measurement of the QT interval is challenging, and it is a poor predictor for the development of this arrhythmia. Nevertheless, QT interval monitoring is an essential part of pharmaceutical development, and significant increases in the QT interval may prevent a drug from gaining approval., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
15. Cardiology patient page. Breast cancer chemotherapy and your heart.
- Author
-
Unitt C, Montazeri K, Tolaney S, and Moslehi J
- Subjects
- Anthracyclines adverse effects, Anthracyclines therapeutic use, Antibodies, Monoclonal, Humanized adverse effects, Antibodies, Monoclonal, Humanized therapeutic use, Antineoplastic Agents therapeutic use, Cardiomyopathies epidemiology, Female, Heart Failure epidemiology, Humans, Risk Factors, Trastuzumab, Antineoplastic Agents adverse effects, Breast Neoplasms drug therapy, Drug Therapy, Heart Diseases epidemiology
- Published
- 2014
- Full Text
- View/download PDF
16. Cardiovascular complications associated with novel angiogenesis inhibitors: emerging evidence and evolving perspectives.
- Author
-
Bair SM, Choueiri TK, and Moslehi J
- Subjects
- Cardiotoxins pharmacology, Humans, Medication Therapy Management, Neovascularization, Pathologic metabolism, Patient Care Team, Signal Transduction drug effects, Translational Research, Biomedical, Vascular Endothelial Growth Factor A metabolism, Angiogenesis Inhibitors pharmacology, Antineoplastic Agents pharmacology, Cardiovascular Diseases chemically induced, Cardiovascular Diseases classification, Cardiovascular Diseases metabolism, Cardiovascular Diseases prevention & control, Neovascularization, Pathologic drug therapy
- Abstract
Novel cancer therapies targeting tumor angiogenesis have revolutionized treatment options in a variety of tumors. Specifically, VEGF signaling pathway (VSP) inhibitors have been introduced into clinical practice at a rapid pace over the last decade. It is becoming increasingly clear that VSP inhibitors can cause cardiovascular toxicities including hypertension, thrombosis, and heart failure. This review highlights these toxicities and proposes several strategies in their prevention and treatment. However, we recognize the dearth of data in this area and advocate a multi-disciplinary approach involving cardiologists and oncologists, as well as clinical and translational studies, in understanding and treating VSP-inhibitor associated toxicities., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
17. Breast cancer therapies and cardiomyopathy.
- Author
-
Groarke J, Tong D, Khambhati J, Cheng S, and Moslehi J
- Subjects
- Adult, Antibodies, Monoclonal, Humanized adverse effects, Biomarkers blood, Cardiomyopathies diagnosis, Cardiomyopathies drug therapy, Echocardiography, Female, Gated Blood-Pool Imaging, Humans, Receptor, ErbB-2 antagonists & inhibitors, Risk Assessment, Trastuzumab, Anthracyclines adverse effects, Antineoplastic Agents adverse effects, Breast Neoplasms drug therapy, Cardiomyopathies chemically induced, Heart drug effects
- Abstract
The prevalence of chemotherapy-related cardiac disease is increasing and management demands a multidisciplinary approach from cardiologists and oncologists. Pretreatment identification of predisposing risk factors and assessment of cardiac function before and at intervals during and after therapy with cardiotoxic agents are necessary. In clinical practice, surveillance is largely performed using transthoracic echocardiography or multi-gated radionuclide angiography. Imaging strategies that detect cardiac injury before overt left ventricular systolic dysfunction provide an opportunity for early intervention and improved cardiac outcomes., (Copyright © 2012. Published by Elsevier Inc.)
- Published
- 2012
- Full Text
- View/download PDF
18. Reversible cardiomyopathy associated with sunitinib and sorafenib.
- Author
-
Uraizee I, Cheng S, and Moslehi J
- Subjects
- Aged, Female, Humans, Male, Middle Aged, Niacinamide analogs & derivatives, Phenylurea Compounds, Protein-Tyrosine Kinases antagonists & inhibitors, Sorafenib, Sunitinib, Antineoplastic Agents adverse effects, Benzenesulfonates adverse effects, Cardiomyopathies chemically induced, Indoles adverse effects, Pyridines adverse effects, Pyrroles adverse effects, Vascular Endothelial Growth Factor A antagonists & inhibitors
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.