1
|
Tomuleasa C, Tigu AB, Munteanu R, Moldovan CS, Kegyes D, Onaciu A, Gulei D, Ghiaur G, Einsele H, Croce CM. Therapeutic advances of targeting receptor tyrosine kinases in cancer. Signal Transduct Target Ther 2024; 9:201. [PMID: 39138146 PMCID: PMC11323831 DOI: 10.1038/s41392-024-01899-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 05/29/2024] [Accepted: 06/14/2024] [Indexed: 08/15/2024] Open
Abstract
Receptor tyrosine kinases (RTKs), a category of transmembrane receptors, have gained significant clinical attention in oncology due to their central role in cancer pathogenesis. Genetic alterations, including mutations, amplifications, and overexpression of certain RTKs, are critical in creating environments conducive to tumor development. Following their discovery, extensive research has revealed how RTK dysregulation contributes to oncogenesis, with many cancer subtypes showing dependency on aberrant RTK signaling for their proliferation, survival and progression. These findings paved the way for targeted therapies that aim to inhibit crucial biological pathways in cancer. As a result, RTKs have emerged as primary targets in anticancer therapeutic development. Over the past two decades, this has led to the synthesis and clinical validation of numerous small molecule tyrosine kinase inhibitors (TKIs), now effectively utilized in treating various cancer types. In this manuscript we aim to provide a comprehensive understanding of the RTKs in the context of cancer. We explored the various alterations and overexpression of specific receptors across different malignancies, with special attention dedicated to the examination of current RTK inhibitors, highlighting their role as potential targeted therapies. By integrating the latest research findings and clinical evidence, we seek to elucidate the pivotal role of RTKs in cancer biology and the therapeutic efficacy of RTK inhibition with promising treatment outcomes.
Collapse
Affiliation(s)
- Ciprian Tomuleasa
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania.
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj Napoca, Romania.
- Academy of Romanian Scientists, Ilfov 3, 050044, Bucharest, Romania.
| | - Adrian-Bogdan Tigu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Academy of Romanian Scientists, Ilfov 3, 050044, Bucharest, Romania
| | - Raluca Munteanu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
- Academy of Romanian Scientists, Ilfov 3, 050044, Bucharest, Romania
| | - Cristian-Silviu Moldovan
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - David Kegyes
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
- Academy of Romanian Scientists, Ilfov 3, 050044, Bucharest, Romania
| | - Anca Onaciu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Diana Gulei
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Gabriel Ghiaur
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
- Department of Leukemia, Sidney Kimmel Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hermann Einsele
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
- Universitätsklinikum Würzburg, Medizinische Klinik II, Würzburg, Germany
| | - Carlo M Croce
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
2
|
Kunitake M, Goto A, Sakai Y, Higuchi K, Muto I, Ueda K, Hamada T, Hiraoka K, Murotani K. Exploring Predictors of Hypertension Development With Pazopanib and Examining Predictive Performance Over Time. In Vivo 2024; 38:1882-1890. [PMID: 38936947 PMCID: PMC11215618 DOI: 10.21873/invivo.13643] [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: 02/20/2024] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND/AIM Hypertension occurs frequently in patients taking pazopanib. Therefore, this study aimed to clarify the predictive factors for pazopanib-induced hypertension. PATIENTS AND METHODS In total, 47 patients who started pazopanib treatment for renal cell carcinoma or soft tissue sarcoma during hospitalization at Kurume University Hospital from November 2012 to February 2020 were included in the study. Patient background factors associated with pazopanib-induced hypertension were analyzed using a logistic regression model. Subsequently, a time-dependent receiver operating characteristic (ROC) analysis was performed to evaluate changes in the predictive performance of predictors of pazopanib-induced hypertension over time. RESULTS Logistic regression analysis showed that total bilirubin (t-bil) and sex are predictors of pazopanib-induced hypertension, along with systolic blood pressure (SBP) before pazopanib introduction. Additionally, evaluation of area under the curve (AUC) changes over time during the first 20 days of pazopanib treatment using time-dependent ROC showed that the AUC tended to be higher in the first half for SBP and in the second half for t-bil. Moreover, models including these two factors (SBP+t-bil and SBP+t-bil+sex) maintained a higher AUC from the early to late stages of the treatment period. CONCLUSION Total bilirubin and sex can serve as predictors of pazopanib-induced hypertension. Total bilirubin may contribute to the prediction of the development of hypertension after day 5.
Collapse
Affiliation(s)
- Masayuki Kunitake
- Department of Pharmacy, Kurume University Hospital, Kurume, Japan;
- Biostatistics Center, Graduate School of Medicine, Kurume University, Kurume, Japan
| | - Aya Goto
- Department of Pharmacy, Kurume University Hospital, Kurume, Japan
| | - Yoshiro Sakai
- Department of Pharmacy, Kurume University Hospital, Kurume, Japan
| | - Kyoko Higuchi
- Department of Pharmacy, Kurume University Hospital, Kurume, Japan
| | - Ikko Muto
- Department of Dermatology, Kurume University School of Medicine, Kurume, Japan
| | - Kosuke Ueda
- Department of Urology, Kurume University School of Medicine, Kurume, Japan
| | - Tetsuya Hamada
- Department of Orthopedic Surgery, Kurume University School of Medicine, Kurume, Japan
| | - Koji Hiraoka
- Department of Orthopedic Surgery, Kurume University School of Medicine, Kurume, Japan
| | | |
Collapse
|
3
|
Zheng Y, Liu Y, Chen Z, Zhang Y, Qi Z, Wu N, Zhao Z, Tse G, Wang Y, Hu H, Niu Y, Liu T. Cardiovascular disease burden in patients with urological cancers: The new discipline of uro-cardio-oncology. CANCER INNOVATION 2024; 3:e108. [PMID: 38946935 PMCID: PMC11212304 DOI: 10.1002/cai2.108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/07/2023] [Accepted: 11/28/2023] [Indexed: 07/02/2024]
Abstract
Cancer remains a major cause of mortality worldwide, and urological cancers are the most common cancers among men. Several therapeutic agents have been used to treat urological cancer, leading to improved survival for patients. However, this has been accompanied by an increase in the frequency of survivors with cardiovascular complications caused by anticancer medications. Here, we propose the novel discipline of uro-cardio-oncology, an evolving subspecialty focused on the complex interactions between cardiovascular disease and urological cancer. In this comprehensive review, we discuss the various cardiovascular toxicities induced by different classes of antineoplastic agents used to treat urological cancers, including androgen deprivation therapy, vascular endothelial growth factor receptor tyrosine kinase inhibitors, immune checkpoint inhibitors, and chemotherapeutics. In addition, we discuss possible mechanisms underlying the cardiovascular toxicity associated with anticancer therapy and outline strategies for the surveillance, diagnosis, and effective management of cardiovascular complications. Finally, we provide an analysis of future perspectives in this emerging specialty, identifying areas in need of further research.
Collapse
Affiliation(s)
- Yi Zheng
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of CardiologySecond Hospital of Tianjin Medical UniversityTianjinChina
| | - Ying Liu
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of CardiologySecond Hospital of Tianjin Medical UniversityTianjinChina
| | - Ziliang Chen
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of CardiologySecond Hospital of Tianjin Medical UniversityTianjinChina
| | - Yunpeng Zhang
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of CardiologySecond Hospital of Tianjin Medical UniversityTianjinChina
| | - Zuo Qi
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of CardiologySecond Hospital of Tianjin Medical UniversityTianjinChina
| | - Ning Wu
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of CardiologySecond Hospital of Tianjin Medical UniversityTianjinChina
| | - Zhiqiang Zhao
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of CardiologySecond Hospital of Tianjin Medical UniversityTianjinChina
| | - Gary Tse
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of CardiologySecond Hospital of Tianjin Medical UniversityTianjinChina
- School of Nursing and Health StudiesHong Kong Metropolitan UniversityHong KongChina
| | - Yong Wang
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of CardiologySecond Hospital of Tianjin Medical UniversityTianjinChina
| | - Hailong Hu
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of CardiologySecond Hospital of Tianjin Medical UniversityTianjinChina
| | - Yuanjie Niu
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of CardiologySecond Hospital of Tianjin Medical UniversityTianjinChina
| | - Tong Liu
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of CardiologySecond Hospital of Tianjin Medical UniversityTianjinChina
| |
Collapse
|
4
|
Wang X, Nakano K, Shiga T, Ohmoto A, Oyakawa T, Ebihara A, Sato Y, Fukuda N, Nishizawa M, Urasaki T, Ono M, Yunokawa M, Tomomatsu J, Takahashi S. Assessment of Pazopanib-Related Heart Failure in Patients With Advanced Soft Tissue Sarcoma - A Single Institute Analysis. Circ J 2024; 88:228-233. [PMID: 35314578 DOI: 10.1253/circj.cj-21-0808] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2024]
Abstract
BACKGROUND Heart failure (HF) is one of the potential adverse events of pazopanib treatment for soft tissue sarcoma (STS), but detailed reports of such HF cases are scarce. This study determined the incidence and risk factors of HF following pazopanib treatment for STS at our Institute and the clinical outcomes. METHODS AND RESULTS This study retrospectively analyzed the cases of STS patients treated with pazopanib (n=151) between 2012 and 2020. HF occurred in 6 patients (3.9%) at the median onset of 137 (range 14-468) days after the treatment initiation. When their HF was diagnosed, pazopanib was interrupted in all 6 patients. No patients experienced HF-related death, and HF development was not a significant factor for poor overall survival. The cumulative doses of anthracyclines (>225 mg/m2) before pazopanib initiation (83% vs. 37%, P=0.031), pazopanib initiation at age ≥60 years (83% vs. 35%, P=0.026), and the baseline B-type natriuretic peptide (BNP) concentration (≥50 pg/mL) before pazopanib (67% vs. 11%, P=0.002) initiation were predictive factors for post-pazopanib treatment HF. CONCLUSIONS The study findings highlight the effect of past anthracycline exposure and baseline BNP for pazopanib-associated HF. Although the study patients' clinical outcomes were generally favorable, periodic monitoring of cardiac function using ultrasonic echocardiography or serum markers is essential to detect events early and begin therapeutic intervention appropriately under a cardiologist's instructions.
Collapse
Affiliation(s)
- Xiaofei Wang
- Department of Medical Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research
| | - Kenji Nakano
- Department of Medical Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research
| | - Taro Shiga
- Department of Onco-Cardiology/Cardiovascular Medicine, The Cancer Institute Hospital of Japanese Foundation for Cancer Research
| | - Akihiro Ohmoto
- Department of Medical Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research
| | - Takuya Oyakawa
- Department of Onco-Cardiology/Cardiovascular Medicine, The Cancer Institute Hospital of Japanese Foundation for Cancer Research
| | - Aya Ebihara
- Department of Onco-Cardiology/Cardiovascular Medicine, The Cancer Institute Hospital of Japanese Foundation for Cancer Research
| | - Yasuyoshi Sato
- Department of Medical Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research
| | - Naoki Fukuda
- Department of Medical Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research
| | - Masatoshi Nishizawa
- Department of Medical Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research
- Next Generation Development of Genome and Cellular Therapy Program, Research Institute for Radiation Biology and Medicine, Hiroshima University
| | - Tsuya Urasaki
- Department of Medical Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research
| | - Makiko Ono
- Department of Medical Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research
| | - Mayu Yunokawa
- Department of Medical Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research
| | - Junichi Tomomatsu
- Department of Medical Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research
| | - Shunji Takahashi
- Department of Medical Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research
| |
Collapse
|
5
|
Shyam Sunder S, Sharma UC, Pokharel S. Adverse effects of tyrosine kinase inhibitors in cancer therapy: pathophysiology, mechanisms and clinical management. Signal Transduct Target Ther 2023; 8:262. [PMID: 37414756 PMCID: PMC10326056 DOI: 10.1038/s41392-023-01469-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 04/06/2023] [Accepted: 04/23/2023] [Indexed: 07/08/2023] Open
Abstract
Since their invention in the early 2000s, tyrosine kinase inhibitors (TKIs) have gained prominence as the most effective pathway-directed anti-cancer agents. TKIs have shown significant utility in the treatment of multiple hematological malignancies and solid tumors, including chronic myelogenous leukemia, non-small cell lung cancers, gastrointestinal stromal tumors, and HER2-positive breast cancers. Given their widespread applications, an increasing frequency of TKI-induced adverse effects has been reported. Although TKIs are known to affect multiple organs in the body including the lungs, liver, gastrointestinal tract, kidneys, thyroid, blood, and skin, cardiac involvement accounts for some of the most serious complications. The most frequently reported cardiovascular side effects range from hypertension, atrial fibrillation, reduced cardiac function, and heart failure to sudden death. The potential mechanisms of these side effects are unclear, leading to critical knowledge gaps in the development of effective therapy and treatment guidelines. There are limited data to infer the best clinical approaches for the early detection and therapeutic modulation of TKI-induced side effects, and universal consensus regarding various management guidelines is yet to be reached. In this state-of-the-art review, we examine multiple pre-clinical and clinical studies and curate evidence on the pathophysiology, mechanisms, and clinical management of these adverse reactions. We expect that this review will provide researchers and allied healthcare providers with the most up-to-date information on the pathophysiology, natural history, risk stratification, and management of emerging TKI-induced side effects in cancer patients.
Collapse
Affiliation(s)
- Sunitha Shyam Sunder
- Cardio-Oncology Research Group, Department of Pathology and Laboratory Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Umesh C Sharma
- Division of Cardiovascular Medicine, Jacob's School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Saraswati Pokharel
- Cardio-Oncology Research Group, Department of Pathology and Laboratory Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
| |
Collapse
|
6
|
Harki O, Bouyon S, Sallé M, Arco-Hierves A, Lemarié E, Demory A, Chirica C, Vilgrain I, Pépin JL, Faury G, Briançon-Marjollet A. Inhibition of Vascular Endothelial Cadherin Cleavage Prevents Elastic Fiber Alterations and Atherosclerosis Induced by Intermittent Hypoxia in the Mouse Aorta. Int J Mol Sci 2022; 23:ijms23137012. [PMID: 35806017 PMCID: PMC9266969 DOI: 10.3390/ijms23137012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/17/2022] [Accepted: 06/21/2022] [Indexed: 01/27/2023] Open
Abstract
Intermittent hypoxia (IH), the major feature of obstructive sleep apnea syndrome (OSAS), induces atherosclerosis and elastic fiber alterations. VE-cadherin cleavage is increased in OSAS patients and in an IH-cellular model. It is mediated by HIF-1 and Src-tyr-kinases pathways and results in endothelial hyperpermeability. Our aim was to determine whether blocking VE-cadherin cleavage in vivo could be an efficient strategy to inhibit deleterious IH-induced vascular remodeling, elastic fiber defects and atherogenesis. VE-cadherin regulation, aortic remodeling and atherosclerosis were studied in IH-exposed C57Bl/6J or ApoE-/-mice treated or not with Src-tyr-kinases inhibitors (Saracatinib/Pazopanib) or a HIF-1 inhibitor (Acriflavine). Human aortic endothelial cells were exposed to IH and treated with the same inhibitors. LDL and the monocytes transendothelium passage were measured. In vitro, IH increased transendothelium LDL and monocytes passage, and the tested inhibitors prevented these effects. In mice, IH decreased VE-cadherin expression and increased plasmatic sVE level, intima-media thickness, elastic fiber alterations and atherosclerosis, while the inhibitors prevented these in vivo effects. In vivo inhibition of HIF-1 and Src tyr kinase pathways were associated with the prevention of IH-induced elastic fiber/lamella degradation and atherogenesis, which suggests that VE-cadherin could be an important target to limit atherogenesis and progression of arterial stiffness in OSAS.
Collapse
Affiliation(s)
- Olfa Harki
- Université Grenoble Alpes, INSERM U1300, CHU Grenoble Alpes, Laboratoire HP2, 38042 Grenoble, France; (O.H.); (S.B.); (M.S.); (A.A.-H.); (E.L.); (A.D.); (J.-L.P.); (A.B.-M.)
| | - Sophie Bouyon
- Université Grenoble Alpes, INSERM U1300, CHU Grenoble Alpes, Laboratoire HP2, 38042 Grenoble, France; (O.H.); (S.B.); (M.S.); (A.A.-H.); (E.L.); (A.D.); (J.-L.P.); (A.B.-M.)
| | - Marine Sallé
- Université Grenoble Alpes, INSERM U1300, CHU Grenoble Alpes, Laboratoire HP2, 38042 Grenoble, France; (O.H.); (S.B.); (M.S.); (A.A.-H.); (E.L.); (A.D.); (J.-L.P.); (A.B.-M.)
| | - Alejandro Arco-Hierves
- Université Grenoble Alpes, INSERM U1300, CHU Grenoble Alpes, Laboratoire HP2, 38042 Grenoble, France; (O.H.); (S.B.); (M.S.); (A.A.-H.); (E.L.); (A.D.); (J.-L.P.); (A.B.-M.)
| | - Emeline Lemarié
- Université Grenoble Alpes, INSERM U1300, CHU Grenoble Alpes, Laboratoire HP2, 38042 Grenoble, France; (O.H.); (S.B.); (M.S.); (A.A.-H.); (E.L.); (A.D.); (J.-L.P.); (A.B.-M.)
| | - Alexandra Demory
- Université Grenoble Alpes, INSERM U1300, CHU Grenoble Alpes, Laboratoire HP2, 38042 Grenoble, France; (O.H.); (S.B.); (M.S.); (A.A.-H.); (E.L.); (A.D.); (J.-L.P.); (A.B.-M.)
| | - Carole Chirica
- Unité Biochimie Immunoanalyse, Service de Biochimie SB2TE, CHU Grenoble Alpes, 38000 Grenoble, France;
| | - Isabelle Vilgrain
- Université Grenoble Alpes, INSERM U1292, CEA, 38042 Grenoble, France;
| | - Jean-Louis Pépin
- Université Grenoble Alpes, INSERM U1300, CHU Grenoble Alpes, Laboratoire HP2, 38042 Grenoble, France; (O.H.); (S.B.); (M.S.); (A.A.-H.); (E.L.); (A.D.); (J.-L.P.); (A.B.-M.)
| | - Gilles Faury
- Université Grenoble Alpes, INSERM U1300, CHU Grenoble Alpes, Laboratoire HP2, 38042 Grenoble, France; (O.H.); (S.B.); (M.S.); (A.A.-H.); (E.L.); (A.D.); (J.-L.P.); (A.B.-M.)
- Correspondence:
| | - Anne Briançon-Marjollet
- Université Grenoble Alpes, INSERM U1300, CHU Grenoble Alpes, Laboratoire HP2, 38042 Grenoble, France; (O.H.); (S.B.); (M.S.); (A.A.-H.); (E.L.); (A.D.); (J.-L.P.); (A.B.-M.)
| |
Collapse
|
7
|
Sadeghi S, Kargar M. Is there association between clinically relevant toxicities of pazopanib and sunitinib with the use of weak CYP3A4 and P-gp inhibitors? Eur J Clin Pharmacol 2021; 77:1427-1428. [PMID: 33723657 DOI: 10.1007/s00228-021-03118-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 03/02/2021] [Indexed: 11/24/2022]
Affiliation(s)
- Setayesh Sadeghi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mona Kargar
- Research Center for Rational Use of Drugs, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Iacopo F, Branch M, Cardinale D, Middeldorp M, Sanders P, Cohen JB, Achirica MC, Jaiswal S, Brown SA. Preventive Cardio-Oncology: Cardiovascular Disease Prevention in Cancer Patients and Survivors. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2021. [DOI: 10.1007/s11936-020-00883-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
9
|
Abstract
Chemotherapy, alone or in association with radiation therapy, has represented the cornerstone of cancer treatment for decades. However, in the last several years, an unprecedented progress in the understanding of cancer biology and the discovery of novel therapeutic targets have led to a paradigm shift in the management of patients with neoplastic diseases. The introduction of tyrosine kinase inhibitors, vascular endothelial growth factor pathway inhibitors, immunomodulatory agents, proteasome inhibitors, immune checkpoint inhibitors, and chimeric antigen receptor T cells, among others, has been associated with prolonged survival in many forms of cancer. A common feature of both chemotherapy and novel cancer treatments is the frequent occurrence of vascular toxicity, mainly mediated by injury to the endothelium. While the mechanisms may vary between agents, the clinical manifestations may overlap and range from hypertension, vasospastic and thrombotic arterial events (myocardial ischemia and infarction, peripheral ischemia, and limb gangrene), venous thromboembolism (deep vein thrombosis and pulmonary embolism) to capillary leak syndrome. Therefore, the effective management of patients with cancer requires a multidisciplinary team approach in which oncologist and cardiovascular medicine specialists work together to prevent, detect, and minimize acute vascular toxicity and long-term consequences of cancer therapy.
Collapse
Affiliation(s)
- Umberto Campia
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
10
|
Jin Y, Xu Z, Yan H, He Q, Yang X, Luo P. A Comprehensive Review of Clinical Cardiotoxicity Incidence of FDA-Approved Small-Molecule Kinase Inhibitors. Front Pharmacol 2020; 11:891. [PMID: 32595510 PMCID: PMC7303342 DOI: 10.3389/fphar.2020.00891] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 05/29/2020] [Indexed: 11/24/2022] Open
Abstract
Numerous protein kinases encoded in the genome have become attractive targets for the treatment of different types of cancer. As of January 2020, a total of 52 small-molecule kinase inhibitors (SMKIs) have been approved by the FDA. With the numerous clinical trials and a heavy focus on drug safety, SMKI-induced cardiotoxicity, which is a life-threatening risk, has greatly attracted the attention of researchers. In this review, the SMKIs with cardiotoxicity incidence were described exhaustively. The data were collected from 42 clinical trials, 25 FDA-published documents, seven meta-analysis/systematic reviews, three case reports and more than 50 other types of articles. To date, 73% (38 of 52) of SMKIs have reported treatment-related cardiotoxicity. Among the 38 SMKIs with known cardiotoxicity, the rates of incidence of cardiac adverse events were QT prolongation: 47% (18 of 38), hypertension: 40% (15 of 38), left ventricular dysfunction: 34% (13 of 38), arrhythmia: 34% (13 of 38), heart failure: 26% (10 of 38) and ischemia or myocardial infarction: 29% (11 of 38). In the development process of novel SMKIs, more attention should be paid to balancing the treatment efficacy and the risk of cardiotoxicity. In preclinical drug studies, producing an accurate and reliable cardiotoxicity evaluation model is of key importance. To avoid the clinical potential cardiotoxicity risk and discontinuation of a highly effective drug, patients treated with SMKIs should be proactively monitored on the basis of a global standard. Moreover, the underlying mechanisms of SMKI-induced cardiotoxicity need to be further studied to develop new therapies for SMKI-induced cardiotoxicity.
Collapse
Affiliation(s)
| | | | | | | | - Xiaochun Yang
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Peihua Luo
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
11
|
Campia U, Moslehi JJ, Amiri-Kordestani L, Barac A, Beckman JA, Chism DD, Cohen P, Groarke JD, Herrmann J, Reilly CM, Weintraub NL. Cardio-Oncology: Vascular and Metabolic Perspectives: A Scientific Statement From the American Heart Association. Circulation 2019; 139:e579-e602. [PMID: 30786722 DOI: 10.1161/cir.0000000000000641] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cardio-oncology has organically developed as a new discipline within cardiovascular medicine as a result of the cardiac and vascular adverse sequelae of the major advances in cancer treatment. Patients with cancer and cancer survivors are at increased risk of vascular disease for a number of reasons. First, many new cancer therapies, including several targeted therapies, are associated with vascular and metabolic complications. Second, cancer itself serves as a risk factor for vascular disease, especially by increasing the risk for thromboembolic events. Finally, recent data suggest that common modifiable and genetic risk factors predispose to both malignancies and cardiovascular disease. Vascular complications in patients with cancer represent a new challenge for the clinician and a new frontier for research and investigation. Indeed, vascular sequelae of novel targeted therapies may provide insights into vascular signaling in humans. Clinically, emerging challenges are best addressed by a multidisciplinary approach in which cardiovascular medicine specialists and vascular biologists work closely with oncologists in the care of patients with cancer and cancer survivors. This novel approach realizes the goal of providing superior care through the creation of cardio-oncology consultative services and the training of a new generation of cardiovascular specialists with a broad understanding of cancer treatments.
Collapse
|
12
|
Cohen JB, Geara AS, Hogan JJ, Townsend RR. Hypertension in Cancer Patients and Survivors: Epidemiology, Diagnosis, and Management. JACC CardioOncol 2019; 1:238-251. [PMID: 32206762 PMCID: PMC7089580 DOI: 10.1016/j.jaccao.2019.11.009] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 11/01/2019] [Accepted: 11/04/2019] [Indexed: 12/28/2022] Open
Abstract
Cancer patients and survivors of cancer have a greater burden of cardiovascular disease compared to the general population. Much of the elevated cardiovascular risk in these individuals is likely attributable to hypertension, as individuals with cancer have a particularly high incidence of hypertension following cancer diagnosis. Treatment with chemotherapy is an independent risk factor for hypertension due to direct effects of many agents on endothelial function, sympathetic activity, and renin-angiotensin system activity as well as nephrotoxicity. Diagnosis and management of hypertension in cancer patients requires accurate blood pressure measurement and consideration of potential confounding factors, such as adjuvant treatments and acute pain, that can temporarily elevate blood pressure readings. Home blood pressure monitoring can be a useful tool to facilitate longitudinal blood pressure monitoring for titration of antihypertensive medications. Selection of antihypertensive agents in cancer patients should account for treatment-specific morbidities and target organ injury.
Collapse
Affiliation(s)
- Jordana B. Cohen
- Renal-Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Abdallah S. Geara
- Renal-Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jonathan J. Hogan
- Renal-Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Raymond R. Townsend
- Renal-Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
13
|
Karaağaç M, Eryılmaz MK. Pazopanib-induced fatal heart failure in a patient with unresectable soft tissue sarcoma and review of literature. J Oncol Pharm Pract 2019; 26:768-774. [PMID: 31547750 DOI: 10.1177/1078155219875797] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Pazopanib, an oral multi-targeted tyrosine kinase inhibitor, is associated with improved outcomes in patients with unresectable or metastatic soft tissue sarcoma. Pazopanib may cause cardiotoxicity such as heart failure. CASE REPORT A 50-year-old female patient with no cardiovascular risk factors other than the previous treatment with adriamycin had a baseline left ventricular ejection fraction of 60%. She was receiving pazopanib 800 mg once daily for advanced leiomyosarcoma of the presacral area. On the 60th day of treatment, she presented with fatigue, palpitation, and exertional dyspnea for several days. Echocardiography was performed, and left ventricular ejection fraction was measured as 25%. Pazopanib-induced heart failure was considered and all other possible preliminary diagnoses were excluded. MANAGEMENT AND OUTCOME Pazopanib was stopped immediately. Bisoprolol fumarate 5 mg orally once daily, spironolactone 100 mg orally once daily, furosemide 40 mg orally once daily, and ramipril 2.5 mg orally once daily were started. The patient's symptoms partially improved. Second echocardiography was performed after 15 days, and left ventricular ejection fraction was measured as 35%. But, despite pazopanib was not resumed and cardiac support treatment was administered, she died four weeks after discontinuation of pazopanib due to heart failure. DISCUSSION Pazopanib-induced heart failure may be fatal. Physicians and patients should be aware of the cardiotoxicity risk when managing the use of pazopanib in soft tissue sarcoma.
Collapse
Affiliation(s)
- Mustafa Karaağaç
- Meram Medical Faculty, Department of Medical Oncology, Necmettin Erbakan University, Konya, Turkey
| | - Melek Karakurt Eryılmaz
- Meram Medical Faculty, Department of Medical Oncology, Necmettin Erbakan University, Konya, Turkey
| |
Collapse
|
14
|
Duran-Moreno J, Kampoli K, Kapetanakis EI, Mademli M, Koufopoulos N, Foukas PG, Kostopanagiotou K, Tomos P, Koumarianou A. Pericardial Synovial Sarcoma: Case Report, Literature Review and Pooled Analysis. In Vivo 2019; 33:1531-1538. [PMID: 31471401 PMCID: PMC6754991 DOI: 10.21873/invivo.11633] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/01/2019] [Accepted: 07/08/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Pericardial synovial sarcomas (PSS) are very rare tumors, with dismal prognosis and limited data. We describe the clinical features and identify prognostic factors of primary PSS. CASE REPORT We describe the case of a 56-year-old male patient with PSS managed by the multidisciplinary team of thoracic oncology. The therapeutic plan comprised surgery, chemotherapy, stereotactic radiosurgery and targeted therapy, with excellent results. MATERIALS AND METHODS Data from 37 cases reported in English during the past 20 years were gathered and analyzed. PSS was found to occur at a mean age of 36±17.082 (range=13-67) years. Survival analysis was performed on 20 cases with follow-up of at least 6 months. CONCLUSION Only complete resection of the tumor seems to be an independent prognostic factor. To our knowledge, this is the first report on the safety and effectivity of pazopanib in PSS and may provide guidance for similar cases in the future.
Collapse
Affiliation(s)
- Jose Duran-Moreno
- Hematology Oncology Unit, Fourth Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Katerina Kampoli
- Hematology Oncology Unit, Fourth Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Emmanouil I Kapetanakis
- Department of Thoracic Surgery, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Mademli
- Second Department of Radiology, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Nektarios Koufopoulos
- Second Department of Pathology, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Periklis G Foukas
- Second Department of Pathology, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Kostas Kostopanagiotou
- Department of Thoracic Surgery, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Periklis Tomos
- Department of Thoracic Surgery, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Anna Koumarianou
- Hematology Oncology Unit, Fourth Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
15
|
Nair P, Prado M, Perea‐Gil I, Karakikes I. Concise Review: Precision Matchmaking: Induced Pluripotent Stem Cells Meet Cardio-Oncology. Stem Cells Transl Med 2019; 8:758-767. [PMID: 31020786 PMCID: PMC6646696 DOI: 10.1002/sctm.18-0279] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 03/12/2019] [Indexed: 12/15/2022] Open
Abstract
As common chemotherapeutic agents are associated with an increased risk of acute and chronic cardiovascular complications, a new clinical discipline, cardio-oncology, has recently emerged. At the same time, the development of preclinical human stem cell-derived cardiovascular models holds promise as a more faithful platform to predict the cardiovascular toxicity of common cancer therapies and advance our understanding of the underlying mechanisms contributing to the cardiotoxicity. In this article, we review the recent advances in preclinical cancer-related cardiotoxicity testing, focusing on new technologies, such as human induced pluripotent stem cell-derived cardiomyocytes and tissue engineering. We further discuss some of the limitations of these technologies and present future directions. Stem Cells Translational Medicine 2019;8:758&767.
Collapse
Affiliation(s)
- Pooja Nair
- Department of Cardiothoracic SurgeryStanford University School of MedicineStanfordCaliforniaUSA
- Cardiovascular InstituteStanford University School of MedicineStanfordCaliforniaUSA
| | - Maricela Prado
- Department of Cardiothoracic SurgeryStanford University School of MedicineStanfordCaliforniaUSA
- Cardiovascular InstituteStanford University School of MedicineStanfordCaliforniaUSA
| | - Isaac Perea‐Gil
- Department of Cardiothoracic SurgeryStanford University School of MedicineStanfordCaliforniaUSA
- Cardiovascular InstituteStanford University School of MedicineStanfordCaliforniaUSA
| | - Ioannis Karakikes
- Department of Cardiothoracic SurgeryStanford University School of MedicineStanfordCaliforniaUSA
- Cardiovascular InstituteStanford University School of MedicineStanfordCaliforniaUSA
| |
Collapse
|
16
|
Cooper SL, Carter JJ, March J, Woolard J. Long-term cardiovascular effects of vandetanib and pazopanib in normotensive rats. Pharmacol Res Perspect 2019; 7:e00477. [PMID: 31164986 PMCID: PMC6543457 DOI: 10.1002/prp2.477] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 04/08/2019] [Indexed: 01/24/2023] Open
Abstract
Vandetanib and pazopanib are clinically available, multi-targeted inhibitors of vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF) receptor tyrosine kinases. Short-term VEGF receptor inhibition is associated with hypertension in 15%-60% of patients, which may limit the use of these anticancer therapies over the longer term. To evaluate the longer-term cardiovascular implications of treatment, we investigated the "on"-treatment (21 days) and "off"-treatment (10 days) effects following daily administration of vandetanib, pazopanib, or vehicle, in conscious rats. Cardiovascular variables were monitored in unrestrained Sprague-Dawley rats instrumented with radiotelemetric devices. In Study 1, rats were randomly assigned to receive either daily intraperitoneal injections of vehicle (volume 0.5 mL; n = 5) or vandetanib 25 mg/kg/day (volume 0.5 mL; n = 6). In Study 2, rats received either vehicle (volume 0.5 mL; n = 4) or pazopanib 30 mg/kg/day (volume 0.5 mL; n = 7), dosed once every 24 hours for 21 days. All solutions were in 2% Tween, 5% propylene glycol in 0.9% saline solution. Vandetanib caused sustained increases in mean arterial pressure (MAP), systolic blood pressure (SBP), and diastolic blood pressure (DBP) compared to baseline and vehicle. Vandetanib also significantly altered the circadian cycling of MAP, SBP, and DBP. Elevations in SBP were detectable 162 hours after the last dose of vandetanib. Pazopanib also caused increases in MAP, SBP, and DBP. However, compared to vandetanib, these increases were of slower onset and a smaller magnitude. These data suggest that the cardiovascular consequences of vandetanib and pazopanib treatment are sustained, even after prolonged cessation of drug treatment.
Collapse
Affiliation(s)
- Samantha L. Cooper
- Division of Physiology, Pharmacology and NeuroscienceSchool of Life SciencesQueen's Medical CentreUniversity of NottinghamNottinghamUK
- Centre of Membrane Proteins and Receptors (COMPARE)University of Birmingham and University of NottinghamMidlandsUK
| | - Joanne J. Carter
- Division of Physiology, Pharmacology and NeuroscienceSchool of Life SciencesQueen's Medical CentreUniversity of NottinghamNottinghamUK
| | - Julie March
- Division of Physiology, Pharmacology and NeuroscienceSchool of Life SciencesQueen's Medical CentreUniversity of NottinghamNottinghamUK
| | - Jeanette Woolard
- Division of Physiology, Pharmacology and NeuroscienceSchool of Life SciencesQueen's Medical CentreUniversity of NottinghamNottinghamUK
- Centre of Membrane Proteins and Receptors (COMPARE)University of Birmingham and University of NottinghamMidlandsUK
| |
Collapse
|
17
|
Justice CN, Derbala MH, Baich TM, Kempton AN, Guo AS, Ho TH, Smith SA. The Impact of Pazopanib on the Cardiovascular System. J Cardiovasc Pharmacol Ther 2018; 23:387-398. [PMID: 29706106 PMCID: PMC6257996 DOI: 10.1177/1074248418769612] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Pazopanib is an approved treatment for renal cell carcinoma and a second-line treatment for nonadipocytic soft-tissue sarcoma. However, its clinical efficacy is limited by its cardiovascular side effects. Pazopanib and other vascular endothelial growth factor receptor tyrosine kinase inhibitors have been associated with the development of hypertension, QT interval prolongation, and other cardiovascular events; however, these mechanisms are largely unknown. Gaining a deeper understanding of these mechanisms is essential for the development of appropriate surveillance strategies and possible diagnostic biomarkers to allow us to monitor patients and modulate therapy prior to significant cardiac insult. This approach will be vital in keeping patients on these life-saving therapies and may be applicable to other tyrosine kinase inhibitors as well. In this review, we provide a comprehensive overview of the preclinical and clinical side effects of pazopanib with a focus on the mechanisms responsible for its toxicity to the cardiovascular system.
Collapse
Affiliation(s)
- Cody N. Justice
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Mohamed H. Derbala
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Tesla M. Baich
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Amber N. Kempton
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Aaron S. Guo
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Thai H. Ho
- Mayo Clinic Scottsdale, Arizona, Phoenix, AZ, USA
| | - Sakima A. Smith
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
- Division of Cardiology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| |
Collapse
|
18
|
Agarwal M, Thareja N, Benjamin M, Akhondi A, Mitchell GD. Tyrosine Kinase Inhibitor-Induced Hypertension. Curr Oncol Rep 2018; 20:65. [DOI: 10.1007/s11912-018-0708-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
19
|
Chemotherapeutic-Induced Cardiovascular Dysfunction: Physiological Effects, Early Detection-The Role of Telomerase to Counteract Mitochondrial Defects and Oxidative Stress. Int J Mol Sci 2018. [PMID: 29534446 PMCID: PMC5877658 DOI: 10.3390/ijms19030797] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Although chemotherapeutics can be highly effective at targeting malignancies, their ability to trigger cardiovascular morbidity is clinically significant. Chemotherapy can adversely affect cardiovascular physiology, resulting in the development of cardiomyopathy, heart failure and microvascular defects. Specifically, anthracyclines are known to cause an excessive buildup of free radical species and mitochondrial DNA damage (mtDNA) that can lead to oxidative stress-induced cardiovascular apoptosis. Therefore, oncologists and cardiologists maintain a network of communication when dealing with patients during treatment in order to treat and prevent chemotherapy-induced cardiovascular damage; however, there is a need to discover more accurate biomarkers and therapeutics to combat and predict the onset of cardiovascular side effects. Telomerase, originally discovered to promote cellular proliferation, has recently emerged as a potential mechanism to counteract mitochondrial defects and restore healthy mitochondrial vascular phenotypes. This review details mechanisms currently used to assess cardiovascular damage, such as C-reactive protein (CRP) and troponin levels, while also unearthing recently researched biomarkers, including circulating mtDNA, telomere length and telomerase activity. Further, we explore a potential role of telomerase in the mitigation of mitochondrial reactive oxygen species and maintenance of mtDNA integrity. Telomerase activity presents a promising indicator for the early detection and treatment of chemotherapy-derived cardiac damage.
Collapse
|