1
|
Pawlonka J, Buchalska B, Buczma K, Borzuta H, Kamińska K, Cudnoch-Jędrzejewska A. Targeting the Renin-angiotensin-aldosterone System (RAAS) for Cardiovascular Protection and Enhanced Oncological Outcomes: Review. Curr Treat Options Oncol 2024; 25:1406-1427. [PMID: 39422794 PMCID: PMC11541340 DOI: 10.1007/s11864-024-01270-9] [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] [Accepted: 09/30/2024] [Indexed: 10/19/2024]
Abstract
OPINION STATEMENT The renin-angiotensin-aldosterone system (RAAS) is a crucial regulator of the cardiovascular system and a target for widely used therapeutic drugs. Dysregulation of RAAS, implicated in prevalent diseases like hypertension and heart failure, has recently gained attention in oncological contexts due to its role in tumor biology and cardiovascular toxicities (CVTs). Thus, RAAS inhibitors (RAASi) may be used as potential supplementary therapies in cancer treatment and CVT prevention. Oncological treatments have evolved significantly, impacting patient survival and safety profiles. However, they pose cardiovascular risks, necessitating strategies for mitigating adverse effects. The main drug classes used in oncology include anthracyclines, anti-HER2 therapies, immune checkpoint inhibitors (ICIs), and vascular endothelial growth factor (VEGF) signaling pathway inhibitors (VSPI). While effective against cancer, these drugs induce varying CVTs. RAASi adjunctive therapy shows promise in enhancing clinical outcomes and protecting the cardiovascular system. Understanding RAAS involvement in cancer and CVT can inform personalized treatment approaches and improve patient care.
Collapse
Affiliation(s)
- J Pawlonka
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland.
| | - B Buchalska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - K Buczma
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - H Borzuta
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - K Kamińska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - A Cudnoch-Jędrzejewska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
2
|
Szponar J, Niziński P, Dudka J, Kasprzak-Drozd K, Oniszczuk A. Natural Products for Preventing and Managing Anthracycline-Induced Cardiotoxicity: A Comprehensive Review. Cells 2024; 13:1151. [PMID: 38995002 PMCID: PMC11240786 DOI: 10.3390/cells13131151] [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: 05/11/2024] [Revised: 07/03/2024] [Accepted: 07/04/2024] [Indexed: 07/13/2024] Open
Abstract
Doxorubicin (DOX) is an anthracycline anticancer agent that is highly effective in the treatment of solid tumors. Given the multiplicity of mechanisms involved in doxorubicin-induced cardiotoxicity, it is difficult to identify a precise molecular target for toxicity. The findings of a literature review suggest that natural products may offer cardioprotective benefits against doxorubicin-induced cardiotoxicity, both in vitro and in vivo. However, further confirmatory studies are required to substantiate this claim. It is of the utmost importance to direct greater attention towards the intricate signaling networks that are of paramount importance for the survival and dysfunction of cardiomyocytes. Notwithstanding encouraging progress made in preclinical studies of natural products for the prevention of DOX-induced cardiotoxicity, these have not yet been translated for clinical use. One of the most significant obstacles hindering the development of cardioprotective adjuvants based on natural products is the lack of adequate bioavailability in humans. This review presents an overview of current knowledge on doxorubicin DOX-induced cardiotoxicity, with a focus on the potential benefits of natural compounds and herbal preparations in preventing this adverse effect. As literature search engines, the browsers in the Scopus, PubMed, Web of Science databases and the ClinicalTrials.gov register were used.
Collapse
Affiliation(s)
- Jarosław Szponar
- Clinical Department of Toxicology and Cardiology, Toxicology Clinic, Stefan Wyszyński Regional Specialist Hospital, Medical University of Lublin, 20-718 Lublin, Poland;
| | - Przemysław Niziński
- Department of Pharmacology, Medical University of Lublin, Radziwiłłowska 11 Street, 20-080 Lublin, Poland;
| | - Jarosław Dudka
- Chair and Department of Toxicology, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland;
| | - Kamila Kasprzak-Drozd
- Department of Inorganic Chemistry, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland;
| | - Anna Oniszczuk
- Department of Inorganic Chemistry, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland;
| |
Collapse
|
3
|
Berlanga-Acosta J, Cibrian D, Valiente-Mustelier J, Suárez-Alba J, García-Ojalvo A, Falcón-Cama V, Jiang B, Wang L, Guillén-Nieto G. Growth hormone releasing peptide-6 (GHRP-6) prevents doxorubicin-induced myocardial and extra-myocardial damages by activating prosurvival mechanisms. Front Pharmacol 2024; 15:1402138. [PMID: 38873418 PMCID: PMC11169835 DOI: 10.3389/fphar.2024.1402138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 05/06/2024] [Indexed: 06/15/2024] Open
Abstract
Introduction: Dilated cardiomyopathy (DCM) is a fatal myocardial condition with ventricular structural changes and functional deficits, leading to systolic dysfunction and heart failure (HF). DCM is a frequent complication in oncologic patients receiving Doxorubicin (Dox). Dox is a highly cardiotoxic drug, whereas its damaging spectrum affects most of the organs by multiple pathogenic cascades. Experimentally reproduced DCM/HF through Dox administrations has shed light on the pathogenic drivers of cardiotoxicity. Growth hormone (GH) releasing peptide 6 (GHRP-6) is a GH secretagogue with expanding and promising cardioprotective pharmacological properties. Here we examined whether GHRP-6 administration concomitant to Dox prevented the onset of DCM/HF and multiple organs damages in otherwise healthy rats. Methods: Myocardial changes were sequentially evaluated by transthoracic echocardiography. Autopsy was conducted at the end of the administration period when ventricular dilation was established. Semiquantitative histopathologic study included heart and other internal organs samples. Myocardial tissue fragments were also addressed for electron microscopy study, and characterization of the transcriptional expression ratio between Bcl-2 and Bax. Serum samples were destined for REDOX system balance assessment. Results and discussion: GHRP-6 administration in parallel to Dox prevented myocardial fibers consumption and ventricular dilation, accounting for an effective preservation of the LV systolic function. GHRP-6 also attenuated extracardiac toxicity preserving epithelial organs integrity, inhibiting interstitial fibrosis, and ultimately reducing morbidity and mortality. Mechanistically, GHRP-6 proved to sustain cellular antioxidant defense, upregulate prosurvival gene Bcl-2, and preserve cardiomyocyte mitochondrial integrity. These evidences contribute to pave potential avenues for the clinical use of GHRP-6 in Dox-treated subjects.
Collapse
Affiliation(s)
| | - Danay Cibrian
- Center for Genetic Engineering and Biotechnology, Playa, Cuba
| | | | | | | | | | - Baohong Jiang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Linlin Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | | |
Collapse
|
4
|
Chen M, Yi Y, Chen B, Zhang H, Dong M, Yuan L, Zhou H, Jiang H, Ma Z. Metformin inhibits OCTN1- and OCTN2-mediated hepatic accumulation of doxorubicin and alleviates its hepatotoxicity in mice. Toxicology 2024; 503:153757. [PMID: 38364893 DOI: 10.1016/j.tox.2024.153757] [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: 09/26/2023] [Revised: 02/13/2024] [Accepted: 02/14/2024] [Indexed: 02/18/2024]
Abstract
Doxorubicin (DOX) is a widely used antitumor agent; however, its clinical application is limited by dose-related organ damage. Because organic cation/carnitine transporters (OCTN1 and OCTN2), which are critical for DOX uptake, are highly expressed in hepatocytes, we aimed to elucidate the role of these transporters in hepatic DOX uptake. The results indicated that inhibitors and RNA interference both significantly reduced DOX accumulation in HepG2 and HepaRG cells, suggesting that OCTN1/2 contribute substantially to DOX uptake by hepatocytes. To determine whether metformin (MET, an inhibitor of OCTN1 and OCTN2) ameliorates DOX-induced hepatotoxicity, we conducted in vitro and in vivo studies. MET (1-100 μM) inhibited DOX (500 nM) accumulation and cytotoxicity in vitro in a concentration-dependent manner. Furthermore, intravenous MET administration at 250 or 500 mg/kg or by gavage at 50, 100, or 200 mg/kg reduced DOX (8 mg/kg) accumulation in a dose-dependent manner in the mouse liver and attenuated the release of alanine aminotransferase, aspartate aminotransferase, and carboxylesterase 1. Additionally, MET reduced the distribution of DOX in the heart, liver, and kidney and enhanced the urinary elimination of DOX; however, it did not increase the nephric toxicity of DOX. In conclusion, our study demonstrated that MET alleviates DOX hepatotoxicity by inhibiting OCTN1- and OCTN2-mediated DOX uptake in vitro (mouse hepatocytes and HepaRG or HepG2 cells) and in mice.
Collapse
Affiliation(s)
- Mingyang Chen
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yaodong Yi
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Binxin Chen
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Hengbin Zhang
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Minlei Dong
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Luexiang Yuan
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Hui Zhou
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; Jinhua Institute of Zhejiang University, Jinhua, China
| | - Huidi Jiang
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; Jinhua Institute of Zhejiang University, Jinhua, China.
| | - Zhiyuan Ma
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China.
| |
Collapse
|
5
|
Yi Y, Zhang H, Chen M, Chen B, Chen Y, Li P, Zhou H, Ma Z, Jiang H. Inhibition of multiple uptake transporters in cardiomyocytes/mitochondria alleviates doxorubicin-induced cardiotoxicity. Chem Biol Interact 2023; 382:110627. [PMID: 37453608 DOI: 10.1016/j.cbi.2023.110627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 06/30/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
Doxorubicin (DOX) has been widely used to treat various tumors; however, DOX-induced cardiotoxicity limits its utilization. Since high accumulation of DOX in cardiomyocytes/mitochondria is the key reason, we aimed to clarify the mechanisms of DOX uptake and explore whether selectively inhibiting DOX uptake transporters would attenuate DOX accumulation and cardiotoxicity. Our results demonstrated that OCTN1/OCTN2/PMAT (organic cation/carnitine transporter 1/2 or plasma membrane monoamine transporter), especially OCTN2, played crucial roles in DOX uptake in cardiomyocytes, while OCTN2 and OCTN1 contributed to DOX transmembrane transport in mitochondria. Metformin (1-100 μM) concentration-dependently reduced DOX (5 μM for accumulation, 500 nM for cytotoxicity) concentration and toxicity in cardiomyocytes/mitochondria via inhibition of OCTN1-, OCTN2- and PMAT-mediated DOX uptake but did not affect its efflux. Furthermore, metformin (iv: 250 and 500 mg/kg or ig: 50, 100 and 200 mg/kg) could dose-dependently reduce DOX (8 mg/kg) accumulation in mouse myocardium and attenuated its cardiotoxicity. In addition, metformin (1-100 μM) did not impair DOX efficacy in breast cancer or leukemia cells. In conclusion, our study clarified the role of multiple transporters, especially OCTN2, in DOX uptake in cardiomyocytes/mitochondria; metformin alleviated DOX-induced cardiotoxicity without compromising its antitumor efficacy by selective inhibition of multiple transporters mediated DOX accumulation in myocardium/mitochondria.
Collapse
Affiliation(s)
- Yaodong Yi
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Hengbin Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Mingyang Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Binxin Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Yingchun Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Ping Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Hui Zhou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Jinhua Institute of Zhejiang University, PR China
| | - Zhiyuan Ma
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.
| | - Huidi Jiang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Jinhua Institute of Zhejiang University, PR China.
| |
Collapse
|
6
|
Zhao Y, Jia H, Hua X, An T, Song J. Cardio-oncology: Shared Genetic, Metabolic, and Pharmacologic Mechanism. Curr Cardiol Rep 2023; 25:863-878. [PMID: 37493874 PMCID: PMC10403418 DOI: 10.1007/s11886-023-01906-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/11/2023] [Indexed: 07/27/2023]
Abstract
PURPOSE OF REVIEW The article aims to investigate the complex relationship between cancer and cardiovascular disease (CVD), with a focus on the effects of cancer treatment on cardiac health. RECENT FINDINGS Advances in cancer treatment have improved long-term survival rates, but CVD has emerged as a leading cause of morbidity and mortality in cancer patients. The interplay between cancer itself, treatment methods, homeostatic changes, and lifestyle modifications contributes to this comorbidity. Recent research in the field of cardio-oncology has revealed common genetic mutations, risk factors, and metabolic features associated with the co-occurrence of cancer and CVD. This article provides a comprehensive review of the latest research in cardio-oncology, including common genetic mutations, risk factors, and metabolic features, and explores the interactions between cancer treatment and CVD drugs, proposing novel approaches for the management of cancer and CVD.
Collapse
Affiliation(s)
- Yiqi Zhao
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, 100037 Beijing, China
| | - Hao Jia
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, 100037 Beijing, China
| | - Xiumeng Hua
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, 100037 Beijing, China
| | - Tao An
- Department of Cardiology, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiangping Song
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, 100037 Beijing, China
| |
Collapse
|
7
|
Fan D, Jin Z, Cao J, Li Y, He T, Zhang W, Peng L, Liu H, Wu X, Chen M, Fan Y, He B, Yu W, Wang H, Hu X, Lu Z. Leucine zipper protein 1 prevents doxorubicin-induced cardiotoxicity in mice. Redox Biol 2023; 64:102780. [PMID: 37354826 DOI: 10.1016/j.redox.2023.102780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/25/2023] [Accepted: 06/08/2023] [Indexed: 06/26/2023] Open
Abstract
OBJECTIVE Doxorubicin (DOX) is commonly used for chemotherapy; however, its clinical value is extremely dampened because of the fatal cardiotoxicity. Leucine zipper protein 1 (LUZP1) plays critical roles in cardiovascular development, and this study is designed for determining its function and mechanism in DOX-induced cardiotoxicity. METHODS Cardiac-specific Luzp1 knockout (cKO) and transgenic (cTG) mice received a single or repeated DOX injections to establish acute and chronic cardiotoxicity. Biomarkers of inflammation, oxidative damage and cell apoptosis were evaluated. Transcriptome and co-immunoprecipitation analysis were used to screen the underlying molecular pathways. Meanwhile, primary cardiomyocytes were applied to confirm the beneficial effects of LUZP1 in depth. RESULTS LUZP1 was upregulated in DOX-injured hearts and cardiomyocytes. Cardiac-specific LUZP1 deficiency aggravated, while cardiac-specific LUZP1 overexpression attenuated DOX-associated inflammation, oxidative damage, cell apoptosis and acute cardiac injury. Mechanistic studies revealed that LUZP1 ameliorated DOX-induced cardiotoxicity through activating 5'-AMP-activated protein kinase (AMPK) pathway, and AMPK deficiency abolished the cardioprotection of LUZP1. Further findings suggested that LUZP1 interacted with protein phosphatase 1 to activate AMPK pathway. Moreover, we determined that cardiac-specific LUZP1 overexpression could also attenuate DOX-associated chronic cardiac injury in mice. CONCLUSION LUZP1 attenuates DOX-induced inflammation, oxidative damage, cell apoptosis and ventricular impairment through regulating AMPK pathway, and gene therapy targeting LUZP1 may provide novel therapeutic approached to treat DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Di Fan
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430062, China; Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, 430062, China
| | - Zhili Jin
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430062, China; Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, 430062, China
| | - Jianlei Cao
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430062, China; Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, 430062, China
| | - Yi Li
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430062, China; Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, 430062, China
| | - Tao He
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430062, China; Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, 430062, China
| | - Wei Zhang
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430062, China; Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, 430062, China
| | - Li Peng
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430062, China; Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, 430062, China
| | - Huixia Liu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430062, China; Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, 430062, China
| | - Xiaoyan Wu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430062, China; Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, 430062, China
| | - Ming Chen
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430062, China; Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, 430062, China
| | - Yongzhen Fan
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430062, China; Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, 430062, China
| | - Bo He
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430062, China; Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, 430062, China
| | - Wenxi Yu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430062, China; Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, 430062, China
| | - Hairong Wang
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430062, China; Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, 430062, China
| | - Xiaorong Hu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430062, China; Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, 430062, China.
| | - Zhibing Lu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430062, China; Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, 430062, China.
| |
Collapse
|
8
|
Dong XL, Yuan BH, Yu SZ, Liu H, Pan XH, Sun J, Pan LL. Adriamycin induces cardiac fibrosis in mice via PRMT5-mediated cardiac fibroblast activation. Acta Pharmacol Sin 2023; 44:573-583. [PMID: 36056082 PMCID: PMC9958096 DOI: 10.1038/s41401-022-00963-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 07/21/2022] [Indexed: 11/08/2022]
Abstract
Long-term treatment with adriamycin (ADR) is associated with higher incidences of cumulative cardiotoxicity manifest as heart failure. ADR-induced cardiomyopathy is characterized by extensive fibrosis that is caused by cardiac fibroblast activation. To date, however, no specific treatment is available to alleviate ADR-induced cardiotoxicity. Protein arginine methyltransferase 5 (PRMT5), a major enzyme responsible for methylation of arginine, regulates numerous cellular processes such as cell differentiation. In the present study we investigated the role of PRMT5 in cardiac fibrosis. Mice were administered ADR (3 mg/kg, i.p., every 2 days) for 2 weeks. We showed that aberrant PRMT5 expression was largely co-localized with α-SMA-positive activated cardiac fibroblasts in ADR-injected mice and in ADR-treated cardiac fibroblasts in vitro. PRMT5-overexpression exacerbated, whereas PRMT5 knockdown alleviated ADR-induced cardiac fibrosis in vivo and TGF-β1-induced cardiac fibroblast activation in vitro. We demonstrated that PRMT5-overexpression enhanced methylated-Smad3 levels in vivo and in vitro. Pretreatment with a specific PRMT5 inhibitor EPZ015666 (5 nM) or overexpression of a catalytically inactive mutant of PRMT5, PRMT5(E444Q), reduced PRMT5-induced methylation of Smad3, thus suppressing PRMT5-mediated cardiac fibroblast activation in vitro. Furthermore, ADR activated cardiac fibroblasts was depending on autocrine TGF-β1. Taken together, our results demonstrate that PRMT5 promotes ADR-induced cardiac fibrosis via activating cardiac fibroblasts, suggesting that it may be a potential therapeutic target of ADR-caused cardiotoxicity.
Collapse
Affiliation(s)
- Xiao-Liang Dong
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Bao-Hui Yuan
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Sheng-Zhou Yu
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - He Liu
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Xiao-Hua Pan
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Jia Sun
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China.
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China.
| | - Li-Long Pan
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China.
| |
Collapse
|
9
|
Kaur N, Sharma RK, Singh Kushwah A, Singh N, Thakur S. A Comprehensive Review of Dilated Cardiomyopathy in Pre-clinical Animal Models in Addition to Herbal Treatment Options and Multi-modality Imaging Strategies. Cardiovasc Hematol Disord Drug Targets 2023; 22:207-225. [PMID: 36734898 DOI: 10.2174/1871529x23666230123122808] [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: 08/26/2022] [Revised: 11/05/2022] [Accepted: 11/17/2022] [Indexed: 02/01/2023]
Abstract
Dilated cardiomyopathy (DCM) is distinguished by ventricular chamber expansion, systolic dysfunction, and normal left ventricular (LV) wall thickness, and is mainly caused due to genetic or environmental factors; however, its aetiology is undetermined in the majority of patients. The focus of this work is on pathogenesis, small animal models, as well as the herbal medicinal approach, and the most recent advances in imaging modalities for patients with dilated cardiomyopathy. Several small animal models have been proposed over the last few years to mimic various pathomechanisms that contribute to dilated cardiomyopathy. Surgical procedures, gene mutations, and drug therapies are all characteristic features of these models. The pros and cons, including heart failure stimulation of extensively established small animal models for dilated cardiomyopathy, are illustrated, as these models tend to procure key insights and contribute to the development of innovative treatment techniques for patients. Traditional medicinal plants used as treatment in these models are also discussed, along with contemporary developments in herbal therapies. In the last few decades, accurate diagnosis, proper recognition of the underlying disease, specific risk stratification, and forecasting of clinical outcome, have indeed improved the health of DCM patients. Cardiac magnetic resonance (CMR) is the bullion criterion for assessing ventricular volume and ejection fraction in a reliable and consistent direction. Other technologies, like strain analysis and 3D echocardiography, have enhanced this technique's predictive and therapeutic potential. Nuclear imaging potentially helps doctors pinpoint the causative factors of left ventricular dysfunction, as with cardiac sarcoidosis and amyloidosis.
Collapse
Affiliation(s)
- Navneet Kaur
- Department of Pharmacology, Amar Shaheed Baba Ajit Singh Jujhar Singh Memorial College of Pharmacy, Bela, Ropar, Punjab, India
| | - Rahul Kumar Sharma
- Department of Pharmacology, Amar Shaheed Baba Ajit Singh Jujhar Singh Memorial College of Pharmacy, Bela, Ropar, Punjab, India
| | - Ajay Singh Kushwah
- Department of Pharmacology, Amar Shaheed Baba Ajit Singh Jujhar Singh Memorial College of Pharmacy, Bela, Ropar, Punjab, India
| | - Nisha Singh
- Department of Pharmacology, Amar Shaheed Baba Ajit Singh Jujhar Singh Memorial College of Pharmacy, Bela, Ropar, Punjab, India
| | - Shilpa Thakur
- Department of Pharmacology, Amar Shaheed Baba Ajit Singh Jujhar Singh Memorial College of Pharmacy, Bela, Ropar, Punjab, India
| |
Collapse
|
10
|
Han Y, Xian Y, Gao X, Qiang P, Hao J, Yang F, Shimosawa T, Chang Y, Xu Q. Eplerenone inhibits the macrophage-to-myofibroblast transition in rats with UUO-induced type 4 cardiorenal syndrome through the MR/CTGF pathway. Int Immunopharmacol 2022; 113:109396. [PMID: 36461595 DOI: 10.1016/j.intimp.2022.109396] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 10/15/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022]
Abstract
Cardiovascular complications are the leading causes of death in patients with chronic kidney disease (CKD), accounting for approximately 50% of deaths. Despite significant advances in the understanding of cardiac disease due to CKD, the underlying mechanisms involved in many pathological changes have not been fully elucidated. In our previous study, we observed severe fibrosis in the contralateral kidney of a 6-month-old rat UUO model. In the present experiment, we also observed severe fibrosis in the hearts of rats subjected to UUO and the macrophage-to-myofibroblast transition (MMT). These effects were inhibited by the mineralocorticoid receptor (MR) blocker eplerenone. Notably, in vitro, aldosterone-activated MR induced the MMT and subsequently promoted the secretion of CTGF, the target of MR, from macrophages; these changes were inhibited by eplerenone. The CTGF also induced the MMT and both the aldosterone and CTGF-induced MMT could be alleviated by the CTGF blocker. In conclusion, our results suggest that targeting the MR/CTGF pathway to inhibit the MMT may be an effective therapeutic strategy for the treatment of cardiac fibrosis.
Collapse
|
11
|
Vuong JT, Stein-Merlob AF, Cheng RK, Yang EH. Novel Therapeutics for Anthracycline Induced Cardiotoxicity. Front Cardiovasc Med 2022; 9:863314. [PMID: 35528842 PMCID: PMC9072636 DOI: 10.3389/fcvm.2022.863314] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/14/2022] [Indexed: 01/04/2023] Open
Abstract
Anthracyclines remain an essential component of the treatment of many hematologic and solid organ malignancies, but has important implications on cardiovascular disease. Anthracycline induced cardiotoxicity (AIC) ranges from asymptomatic LV dysfunction to highly morbid end- stage heart failure. As cancer survivorship improves, the detection and treatment of AIC becomes more crucial to improve patient outcomes. Current treatment modalities for AIC have been largely extrapolated from treatment of conventional heart failure, but developing effective therapies specific to AIC is an area of growing research interest. This review summarizes the current evidence behind the use of neurohormonal agents, dexrazoxane, and resynchronization therapy in AIC, evaluates the clinical outcomes of advanced therapy and heart transplantation in AIC, and explores future horizons for treatment utilizing gene therapy, stem cell therapy, and mechanism-specific targets.
Collapse
Affiliation(s)
- Jacqueline T. Vuong
- Department of Medicine, Ronald Reagan UCLA Medical Center, Los Angeles, CA, United States
| | - Ashley F. Stein-Merlob
- Division of Cardiology, Department of Medicine, Ronald Reagan UCLA Medical Center, Los Angeles, CA, United States
| | - Richard K. Cheng
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Eric H. Yang
- Division of Cardiology, Department of Medicine, Ronald Reagan UCLA Medical Center, Los Angeles, CA, United States
- UCLA Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- *Correspondence: Eric H. Yang,
| |
Collapse
|
12
|
Bauersachs J, Lother A. Mineralocorticoid receptor activation and antagonism in cardiovascular disease: cellular and molecular mechanisms. Kidney Int Suppl (2011) 2022; 12:19-26. [DOI: 10.1016/j.kisu.2021.11.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/15/2021] [Accepted: 11/08/2021] [Indexed: 02/08/2023] Open
|
13
|
L'Abbate S, Chianca M, Fabiani I, Del Franco A, Giannoni A, Vergaro G, Grigoratos C, Kusmic C, Passino C, D'Alessandra Y, Burchielli S, Emdin M, Cardinale DM. In Vivo Murine Models of Cardiotoxicity Due to Anticancer Drugs: Challenges and Opportunities for Clinical Translation. J Cardiovasc Transl Res 2022; 15:1143-1162. [PMID: 35312959 DOI: 10.1007/s12265-022-10231-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/04/2022] [Indexed: 12/13/2022]
Abstract
Modern therapeutic approaches have led to an improvement in the chances of surviving a diagnosis of cancer. However, this may come with side effects, with patients experiencing adverse cardiovascular events or exacerbation of underlying cardiovascular disease related to their cancer treatment. Rodent models of chemotherapy-induced cardiotoxicity are useful to define pathophysiological mechanisms of cardiac damage and to identify potential therapeutic targets. The key mechanisms involved in cardiotoxicity induced by specific different antineoplastic agents are summarized in this state-of-the-art review, as well as the rodent models of cardiotoxicity by different classes of anticancer drugs, along with the strategies tested for primary and secondary cardioprotection. Current approaches for early detection of cardiotoxicity in preclinical studies with a focus on the application of advanced imaging modalities and biomarker strategies are also discussed. Potential applications of cardiotoxicity modelling in rodents are illustrated in relation to the advancements of promising research topics of cardiotoxicity. Created with BioRender.com.
Collapse
Affiliation(s)
- Serena L'Abbate
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Michela Chianca
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Iacopo Fabiani
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy.
| | - Annamaria Del Franco
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy.,Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Alberto Giannoni
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy.,Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Giuseppe Vergaro
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy.,Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | | | | | - Claudio Passino
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy.,Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Yuri D'Alessandra
- Cardiovascular Proteomics Unit, Centro Cardiologico Monzino I.R.C.C.S., Milan, Italy
| | | | - Michele Emdin
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy.,Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Daniela Maria Cardinale
- Cardioncology Unit, Cardiology Division, European Institute of Oncology, I.R.C.C.S., Milan, Italy
| |
Collapse
|
14
|
Chen DS, Yan J, Yang PZ. Cardiomyocyte Atrophy, an Underestimated Contributor in Doxorubicin-Induced Cardiotoxicity. Front Cardiovasc Med 2022; 9:812578. [PMID: 35282350 PMCID: PMC8913904 DOI: 10.3389/fcvm.2022.812578] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/11/2022] [Indexed: 12/21/2022] Open
Abstract
Left ventricular (LV) mass loss is prevalent in doxorubicin (DOX)-induced cardiotoxicity and is responsible for the progressive decline of cardiac function. Comparing with the well-studied role of cell death, the part of cardiomyocyte atrophy (CMA) playing in the LV mass loss is underestimated and the knowledge of the underlying mechanism is still limited. In this review, we summarized the recent advances in the DOX-induced CMA. We found that the CMA caused by DOX is associated with the upregulation of FOXOs and “atrogenes,” the activation of transient receptor potential canonical 3-NADPH oxidase 2 (TRPC3-Nox2) axis, and the suppression of IGF-1-PI3K signaling pathway. The imbalance of anabolic and catabolic process may be the common final pathway of these mechanisms. At last, we provided some strategies that have been demonstrated to alleviate the DOX-induced CMA in animal models.
Collapse
Affiliation(s)
- De-Shu Chen
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, China
- Heart Center of Zhujiang Hospital, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, China
| | - Jing Yan
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, China
- Heart Center of Zhujiang Hospital, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, China
- Jing Yan
| | - Ping-Zhen Yang
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, China
- Heart Center of Zhujiang Hospital, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, China
- *Correspondence: Ping-Zhen Yang
| |
Collapse
|
15
|
Wu BB, Leung KT, Poon ENY. Mitochondrial-Targeted Therapy for Doxorubicin-Induced Cardiotoxicity. Int J Mol Sci 2022; 23:1912. [PMID: 35163838 PMCID: PMC8837080 DOI: 10.3390/ijms23031912] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/27/2022] [Accepted: 02/01/2022] [Indexed: 01/27/2023] Open
Abstract
Anthracyclines, such as doxorubicin, are effective chemotherapeutic agents for the treatment of cancer, but their clinical use is associated with severe and potentially life-threatening cardiotoxicity. Despite decades of research, treatment options remain limited. The mitochondria is commonly considered to be the main target of doxorubicin and mitochondrial dysfunction is the hallmark of doxorubicin-induced cardiotoxicity. Here, we review the pathogenic mechanisms of doxorubicin-induced cardiotoxicity and present an update on cardioprotective strategies for this disorder. Specifically, we focus on strategies that can protect the mitochondria and cover different therapeutic modalities encompassing small molecules, post-transcriptional regulators, and mitochondrial transfer. We also discuss the shortcomings of existing models of doxorubicin-induced cardiotoxicity and explore advances in the use of human pluripotent stem cell derived cardiomyocytes as a platform to facilitate the identification of novel treatments against this disorder.
Collapse
Affiliation(s)
- Bin Bin Wu
- Centre for Cardiovascular Genomics and Medicine, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China;
- Hong Kong Hub of Paediatric Excellence (HK HOPE), The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China;
| | - Kam Tong Leung
- Hong Kong Hub of Paediatric Excellence (HK HOPE), The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China;
- Department of Paediatrics, The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
| | - Ellen Ngar-Yun Poon
- Centre for Cardiovascular Genomics and Medicine, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China;
- Hong Kong Hub of Paediatric Excellence (HK HOPE), The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China;
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
- School of Biomedical Sciences, The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
| |
Collapse
|
16
|
Bauersachs J, López-Andrés N. Mineralocorticoid receptor in cardiovascular diseases-Clinical trials and mechanistic insights. Br J Pharmacol 2021; 179:3119-3134. [PMID: 34643952 DOI: 10.1111/bph.15708] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/07/2021] [Accepted: 09/27/2021] [Indexed: 12/19/2022] Open
Abstract
Aldosterone binds to the mineralocorticoid receptor (NR3C2), a transcription factor of the nuclear receptor family, present in the kidney and in various other non-epithelial cells including the heart and the vasculature. Indeed, extra-renal pathophysiological effects of this hormone have been characterized, extending its actions to the cardiovascular system. A growing body of clinical and pre-clinical evidence suggests that mineralocorticoid receptor overactivation plays an important pathophysiological role in cardiovascular remodelling by promoting cardiac hypertrophy, fibrosis, arterial stiffness and in inflammation and oxidative stress. The following review article outlines the role of mineralocorticoid receptor in cardiovascular disease with a focus on myocardial remodelling and heart failure (HF) including clinical trials as well as cellular and animal studies.
Collapse
Affiliation(s)
- Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Natalia López-Andrés
- Cardiovascular Translational Research. Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| |
Collapse
|
17
|
Furihata T, Maekawa S, Takada S, Kakutani N, Nambu H, Shirakawa R, Yokota T, Kinugawa S. Premedication with pioglitazone prevents doxorubicin-induced left ventricular dysfunction in mice. BMC Pharmacol Toxicol 2021; 22:27. [PMID: 33962676 PMCID: PMC8103594 DOI: 10.1186/s40360-021-00495-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 04/26/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Doxorubicin (DOX) is widely used as an effective chemotherapeutic agent for cancers; however, DOX induces cardiac toxicity, called DOX-induced cardiomyopathy. Although DOX-induced cardiomyopathy is known to be associated with a high cumulative dose of DOX, the mechanisms of its long-term effects have not been completely elucidated. Pioglitazone (Pio) is presently contraindicated in patients with symptomatic heart failure owing to the side effects. The concept of drug repositioning led us to hypothesize the potential effects of Pio as a premedication before DOX treatment, and to analyze this hypothesis in mice. METHODS First, for the hyperacute (day 1) and acute (day 7) DOX-induced dysfunction models, mice were fed a standard diet with or without 0.02% (wt/wt) Pio for 5 days before DOX treatment (15 mg/kg body weight [BW] via intraperitoneal [i.p.] administration). The following 3 treatment groups were analyzed: standard diet + vehicle (Vehicle), standard diet + DOX (DOX), and Pio + DOX. Next, for the chronic model (day 35), the mice were administrated DOX once a week for 5 weeks (5 mg/kg BW/week, i.p.). RESULTS In the acute phase after DOX treatment, the percent fractional shortening of the left ventricle (LV) was significantly decreased in DOX mice. This cardiac malfunction was improved in Pio + DOX mice. In the chronic phase, we observed that LV function was preserved in Pio + DOX mice. CONCLUSIONS Our findings may provide a new pathophysiological explanation by which Pio plays a role in the treatment of DOX-induced cardiomyopathy, but the molecular links between Pio and DOX-induced LV dysfunction remain largely elusive.
Collapse
Affiliation(s)
- Takaaki Furihata
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo, 060-8638, Japan.
| | - Satoshi Maekawa
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Shingo Takada
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo, 060-8638, Japan
- Faculty of Lifelong Sport, Department of Sports Education, Hokusho University, Ebetsu, 069-8511, Japan
| | - Naoya Kakutani
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo, 060-8638, Japan
- Research Fellow of the Japan Society for the Promotion of Science, Tokyo, Japan
| | - Hideo Nambu
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Ryosuke Shirakawa
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Takashi Yokota
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Shintaro Kinugawa
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo, 060-8638, Japan
| |
Collapse
|
18
|
Lother A, Bondareva O, Saadatmand AR, Pollmeier L, Härdtner C, Hilgendorf I, Weichenhan D, Eckstein V, Plass C, Bode C, Backs J, Hein L, Gilsbach R. Diabetes changes gene expression but not DNA methylation in cardiac cells. J Mol Cell Cardiol 2021; 151:74-87. [PMID: 33197445 DOI: 10.1016/j.yjmcc.2020.11.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 10/22/2020] [Accepted: 11/08/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Diabetes mellitus is a worldwide epidemic that causes high mortality due to cardiovascular complications, in particular heart failure. Diabetes is associated with profound pathophysiological changes in the heart. The aim of this study was to investigate the impact of diabetes on gene expression and DNA methylation in cardiac cells. METHODS AND RESULTS Transcriptome analysis of heart tissue from mice with streptozotocin-induced diabetes revealed only 39 genes regulated, whereas cell type-specific analysis of the diabetic heart was more sensitive and more specific than heart tissue analysis and revealed a total of 3205 differentially regulated genes in five cell types. Whole genome DNA methylation analysis with basepair resolution of distinct cardiac cell types identified highly specific DNA methylation signatures of genic and regulatory regions. Interestingly, despite marked changes in gene expression, DNA methylation remained stable in streptozotocin-induced diabetes. Integrated analysis of cell type-specific gene expression enabled us to assign the particular contribution of single cell types to the pathophysiology of the diabetic heart. Finally, analysis of gene regulation revealed ligand-receptor pairs as potential mediators of heterocellular interaction in the diabetic heart, with fibroblasts and monocytes showing the highest degree of interaction. CONCLUSION In summary, cell type-specific analysis reveals differentially regulated gene programs that are associated with distinct biological processes in diabetes. Interestingly, despite these changes in gene expression, cell type-specific DNA methylation signatures of genic and regulatory regions remain stable in diabetes. Analysis of heterocellular interactions in the diabetic heart suggest that the interplay between fibroblasts and monocytes is of pivotal importance.
Collapse
Affiliation(s)
- Achim Lother
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Germany; Heart Center Freiburg University, Department of Cardiology and Angiology I, Faculty of Medicine, University of Freiburg, Germany
| | - Olga Bondareva
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Germany
| | - Ali R Saadatmand
- Institute of Experimental Cardiology, Heidelberg University, Heidelberg, Germany; German Centre for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Luisa Pollmeier
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Germany
| | - Carmen Härdtner
- Heart Center Freiburg University, Department of Cardiology and Angiology I, Faculty of Medicine, University of Freiburg, Germany
| | - Ingo Hilgendorf
- Heart Center Freiburg University, Department of Cardiology and Angiology I, Faculty of Medicine, University of Freiburg, Germany
| | - Dieter Weichenhan
- Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Volker Eckstein
- Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Christoph Plass
- Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christoph Bode
- Heart Center Freiburg University, Department of Cardiology and Angiology I, Faculty of Medicine, University of Freiburg, Germany
| | - Johannes Backs
- Institute of Experimental Cardiology, Heidelberg University, Heidelberg, Germany; German Centre for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Lutz Hein
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Germany; BIOSS Centre for Biological Signalling Studies, University of Freiburg, Germany
| | - Ralf Gilsbach
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Germany; Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main, Germany; German Centre of Cardiovascular Research (DZHK), partner site RheinMain, Frankfurt am Main, Germany.
| |
Collapse
|
19
|
Lódi M, Bánhegyi V, Bódi B, Gyöngyösi A, Kovács Á, Árokszállási A, Hamdani N, Fagyas M, Édes I, Csanádi Z, Czuriga I, Kisvárday Z, Lekli I, Bai P, Tóth A, Papp Z, Czuriga D. Prophylactic, single-drug cardioprotection in a comparative, experimental study of doxorubicin-induced cardiomyopathy. J Transl Med 2020; 18:470. [PMID: 33298102 PMCID: PMC7725221 DOI: 10.1186/s12967-020-02564-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 10/10/2020] [Indexed: 12/28/2022] Open
Abstract
Background Cardiomyopathy is a common side effect of doxorubicin (DOX) chemotherapy. Despite intensive research efforts in the field, there is still no evidence available for routine cardioprotective prophylaxis to prevent cardiotoxicity in the majority of oncological patients at low risk of cardiovascular disease. We have recently demonstrated the advantages of a prophylactic, combined heart failure therapy in an experimental model of DOX-induced cardiomyopathy. In the current work, we focus on individually applied prophylactic medications studied in the same translational environment to clarify their distinct roles in the prevention of DOX cardiotoxicity. Methods Twelve-week-old male Wistar rats were divided into 5 subgroups. Prophylactic β-blocker (BB, bisoprolol), angiotensin-converting enzyme inhibitor (ACEI, perindopril) or aldosterone antagonist (AA, eplerenone) treatments were applied 1 week before DOX administration, then 6 cycles of intravenous DOX chemotherapy were administered. Rats receiving only intravenous DOX or saline served as positive and negative controls. Blood pressure, heart rate, body weight, and echocardiographic parameters were monitored in vivo. Two months after the last DOX administration, the animals were sacrificed, and their heart and serum samples were frozen in liquid nitrogen for histological, mechanical, and biochemical measurements. Results All prophylactic treatments increased the survival of DOX-receiving animals. The lowest mortality rates were seen in the BB and ACEI groups. The left ventricular ejection fraction was only preserved in the BB group. The DOX-induced increase in the isovolumetric relaxation time could not be prevented by any prophylactic treatment. A decreased number of apoptotic nuclei and a preserved myocardial ultrastructure were found in all groups receiving prophylactic cardioprotection, while the DOX-induced fibrotic remodelling and the increase in caspase-3 levels could only be substantially prevented by the BB and ACEI treatments. Conclusion Primary prophylaxis with cardioprotective agents like BB or ACEI has a key role in the prevention of DOX-induced cardiotoxicity in healthy rats. Future human studies are necessary to implement this finding in the clinical management of oncological patients free of cardiovascular risk factors.
Collapse
Affiliation(s)
- Mária Lódi
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | - Viktor Bánhegyi
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | - Beáta Bódi
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | - Alexandra Gyöngyösi
- Department of Pharmacology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Árpád Kovács
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Anita Árokszállási
- Department of Oncology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Nazha Hamdani
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany
| | - Miklós Fagyas
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - István Édes
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Csanádi
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - István Czuriga
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Kisvárday
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - István Lekli
- Department of Pharmacology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Péter Bai
- MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, Hungary
| | - Attila Tóth
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Papp
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Dániel Czuriga
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
20
|
de Boer RA, Hulot J, Tocchetti CG, Aboumsallem JP, Ameri P, Anker SD, Bauersachs J, Bertero E, Coats AJ, Čelutkienė J, Chioncel O, Dodion P, Eschenhagen T, Farmakis D, Bayes‐Genis A, Jäger D, Jankowska EA, Kitsis RN, Konety SH, Larkin J, Lehmann L, Lenihan DJ, Maack C, Moslehi JJ, Müller OJ, Nowak‐Sliwinska P, Piepoli MF, Ponikowski P, Pudil R, Rainer PP, Ruschitzka F, Sawyer D, Seferovic PM, Suter T, Thum T, van der Meer P, Van Laake LW, von Haehling S, Heymans S, Lyon AR, Backs J. Common mechanistic pathways in cancer and heart failure. A scientific roadmap on behalf of the Translational Research Committee of the Heart Failure Association (HFA) of the European Society of Cardiology (ESC). Eur J Heart Fail 2020; 22:2272-2289. [PMID: 33094495 PMCID: PMC7894564 DOI: 10.1002/ejhf.2029] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/13/2020] [Accepted: 10/18/2020] [Indexed: 12/18/2022] Open
Abstract
The co-occurrence of cancer and heart failure (HF) represents a significant clinical drawback as each disease interferes with the treatment of the other. In addition to shared risk factors, a growing body of experimental and clinical evidence reveals numerous commonalities in the biology underlying both pathologies. Inflammation emerges as a common hallmark for both diseases as it contributes to the initiation and progression of both HF and cancer. Under stress, malignant and cardiac cells change their metabolic preferences to survive, which makes these metabolic derangements a great basis to develop intersection strategies and therapies to combat both diseases. Furthermore, genetic predisposition and clonal haematopoiesis are common drivers for both conditions and they hold great clinical relevance in the context of personalized medicine. Additionally, altered angiogenesis is a common hallmark for failing hearts and tumours and represents a promising substrate to target in both diseases. Cardiac cells and malignant cells interact with their surrounding environment called stroma. This interaction mediates the progression of the two pathologies and understanding the structure and function of each stromal component may pave the way for innovative therapeutic strategies and improved outcomes in patients. The interdisciplinary collaboration between cardiologists and oncologists is essential to establish unified guidelines. To this aim, pre-clinical models that mimic the human situation, where both pathologies coexist, are needed to understand all the aspects of the bidirectional relationship between cancer and HF. Finally, adequately powered clinical studies, including patients from all ages, and men and women, with proper adjudication of both cancer and cardiovascular endpoints, are essential to accurately study these two pathologies at the same time.
Collapse
Affiliation(s)
- Rudolf A. de Boer
- Department of CardiologyUniversity Medical Center GroningenGroningenThe Netherlands
| | - Jean‐Sébastien Hulot
- Université de Paris, PARCC, INSERMParisFrance
- CIC1418 and DMU CARTE, AP‐HP, Hôpital Européen Georges‐PompidouParisFrance
| | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences and Interdepartmental Center of Clinical and Translational ResearchFederico II UniversityNaplesItaly
| | | | - Pietro Ameri
- Department of Internal Medicine and Center of Excellence for Biomedical ResearchUniversity of GenovaGenoaItaly
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San MartinoGenoaItaly
| | - Stefan D. Anker
- Department of Cardiology & Berlin Institute of Health Center for Regenerative Therapies (BCRT), German Center for Cardiovascular Research (DZHK), Partner Site BerlinCharité‐Universitätsmedizin Berlin (Campus CVK)BerlinGermany
| | - Johann Bauersachs
- Department of Cardiology and AngiologyHannover Medical SchoolHannoverGermany
| | - Edoardo Bertero
- Comprehensive Heart Failure CenterUniversity Clinic WürzburgWürzburgGermany
| | | | - Jelena Čelutkienė
- Clinic of Cardiac and Vascular Diseases, Institute of Clinical Medicine, Faculty of MedicineVilnius UniversityVilniusLithuania
| | - Ovidiu Chioncel
- Emergency Institute for Cardiovascular Diseases ‘Prof. C.C. Iliescu’University of Medicine Carol DavilaBucharestRomania
| | | | - Thomas Eschenhagen
- Institute of Experimental Pharmacology and ToxicologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- Partner Site Hamburg/Kiel/Lübeck, DZHK (German Centre for Cardiovascular Research)HamburgGermany
| | - Dimitrios Farmakis
- University of Cyprus Medical SchoolNicosiaCyprus
- Cardio‐Oncology Clinic, Heart Failure Unit, Department of CardiologyAthens University Hospital ‘Attikon’, National and Kapodistrian University of Athens Medical SchoolAthensGreece
| | - Antoni Bayes‐Genis
- Heart Failure Unit and Cardiology DepartmentHospital Universitari Germans Trias i Pujol, CIBERCVBadalonaSpain
- Department of MedicineUniversitat Autònoma de BarcelonaBarcelonaSpain
- CIBER CardiovascularInstituto de Salud Carlos IIIMadridSpain
| | - Dirk Jäger
- Department of Medical Oncology, National Center for Tumor Diseases (NCT)University Hospital HeidelbergHeidelbergGermany
| | - Ewa A. Jankowska
- Department of Heart Diseases, Wroclaw Medical University, and Centre for Heart DiseasesUniversity HospitalWroclawPoland
| | - Richard N. Kitsis
- Departments of Medicine (Cardiology) and Cell BiologyWilf Family Cardiovascular Research Institute, Albert Einstein Cancer Center, Albert Einstein College of MedicineNew YorkNYUSA
| | - Suma H. Konety
- Cardiovascular Division, Cardio‐Oncology Program, Department of MedicineUniversity of Minnesota Medical SchoolMinneapolisMNUSA
| | | | - Lorenz Lehmann
- Cardio‐Oncology Unit, Department of CardiologyUniversity of HeidelbergHeidelbergGermany
- DZHK (German Centre for Cardiovascular Research), partner siteHeidelberg/MannheimGermany
- DKFZ (German Cancer Research Center)HeidelbergGermany
| | - Daniel J. Lenihan
- Cardio‐Oncology Center of Excellence, Cardiovascular DivisionWashington University in St. LouisSt. LouisMOUSA
| | - Christoph Maack
- Comprehensive Heart Failure CenterUniversity Clinic WürzburgWürzburgGermany
| | - Javid J. Moslehi
- Division of Cardiovascular Medicine and OncologyCardio‐Oncology Program, Vanderbilt University Medical Center and Vanderbilt‐Ingram Cancer CenterNashvilleTNUSA
| | - Oliver J. Müller
- Department of Internal Medicine IIIUniversity of KielKielGermany
- DZHK (German Centre for Cardiovascular Research), partner siteHamburg/Kiel/LübeckGermany
| | - Patrycja Nowak‐Sliwinska
- School of Pharmaceutical SciencesUniversity of Geneva, Institute of Pharmaceutical Sciences of Western Switzerland, University of GenevaGenevaSwitzerland
- Translational Research Center in OncohaematologyGenevaSwitzerland
| | | | - Piotr Ponikowski
- Department of Heart Diseases, Wroclaw Medical University, and Centre for Heart DiseasesUniversity HospitalWroclawPoland
| | - Radek Pudil
- 1st Department Medicine‐CardioangiologyUniversity Hospital and Medical FacultyHradec KraloveCzech Republic
| | - Peter P. Rainer
- Medical University of GrazUniversity Heart Center – Division of CardiologyGrazAustria
| | - Frank Ruschitzka
- Department of CardiologyUniversity Hospital Zurich, University Heart CenterZurichSwitzerland
| | - Douglas Sawyer
- Center for Molecular Medicine, Maine Medical Center Research InstituteMaine Medical CenterScarboroughMEUSA
| | - Petar M. Seferovic
- University of Belgrade Faculty of Medicine, Serbian Academy of Sciences and ArtsBelgradeSerbia
| | - Thomas Suter
- Swiss Cardiovascular CentreBern UniversityBernSwitzerland
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS)Hannover Medical SchoolHannoverGermany
| | - Peter van der Meer
- Department of CardiologyUniversity Medical Center GroningenGroningenThe Netherlands
| | - Linda W. Van Laake
- Division Heart and Lungs and Regenerative Medicine CentreUniversity Medical Centre Utrecht and Utrecht UniversityUtrechtThe Netherlands
| | - Stephan von Haehling
- Department of Cardiology and Pneumology, Heart CenterUniversity of Göttingen Medical CenterGöttingenGermany
- German Center for Cardiovascular Research (DZHK), partner site GöttingenGöttingenGermany
| | - Stephane Heymans
- Department of Cardiology, CARIM School for Cardiovascular Diseases Faculty of Health, Medicine and Life SciencesMaastricht UniversityMaastrichtThe Netherlands
- Department of Cardiovascular SciencesCentre for Molecular and Vascular Biology, KU LeuvenLeuvenBelgium
| | - Alexander R. Lyon
- Cardio‐Oncology Service, Royal Brompton Hospital, and National Heart and Lung Institute, Imperial College LondonLondonUK
| | - Johannes Backs
- Institute of Experimental CardiologyHeidelberg University HospitalHeidelbergGermany
- DZHK (German Centre for Cardiovascular Research), partner siteHeidelberg/MannheimGermany
| |
Collapse
|
21
|
Ma ZG, Kong CY, Wu HM, Song P, Zhang X, Yuan YP, Deng W, Tang QZ. Toll-like receptor 5 deficiency diminishes doxorubicin-induced acute cardiotoxicity in mice. Am J Cancer Res 2020; 10:11013-11025. [PMID: 33042267 PMCID: PMC7532690 DOI: 10.7150/thno.47516] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 08/21/2020] [Indexed: 12/24/2022] Open
Abstract
Rationale: Clinical application of doxorubicin (DOX) is limited by its toxic cardiovascular side effects. Our previous study found that toll-like receptor (TLR) 5 deficiency attenuated cardiac fibrosis in mice. However, the role of TLR5 in DOX-induced cardiotoxicity remains unclear. Methods: To further investigate this, TLR5-deficient mice were subjected to a single intraperitoneal injection of DOX to mimic an acute model. Results: Here, we reported that TLR5 expression was markedly increased in response to DOX injection. Moreover, TLR5 deficiency exerted potent protective effects against DOX-related cardiac injury, whereas activation of TLR5 by flagellin exacerbated DOX injection-induced cardiotoxicity. Mechanistically, the effects of TLR5 were largely attributed to direct interaction with spleen tyrosine kinase to activate NADPH oxidase (NOX) 2, increasing the production of superoxide and subsequent activation of p38. The toxic effects of TLR5 activation in DOX-related acute cardiac injury were abolished by NOX2 deficiency in mice. Our further study showed that neutralizing antibody-mediated TLR5 depletion also attenuated DOX-induced acute cardiotoxicity. Conclusion: These findings suggest that TLR5 deficiency attenuates DOX-induced cardiotoxicity in mice, and targeting TLR5 may provide feasible therapies for DOX-induced acute cardiotoxicity.
Collapse
|
22
|
Riehle C, Bauersachs J. Small animal models of heart failure. Cardiovasc Res 2020; 115:1838-1849. [PMID: 31243437 PMCID: PMC6803815 DOI: 10.1093/cvr/cvz161] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 04/30/2019] [Accepted: 06/24/2019] [Indexed: 12/11/2022] Open
Abstract
Heart disease is a major cause of death worldwide with increasing prevalence, which urges the development of new therapeutic strategies. Over the last few decades, numerous small animal models have been generated to mimic various pathomechanisms contributing to heart failure (HF). Despite some limitations, these animal models have greatly advanced our understanding of the pathogenesis of the different aetiologies of HF and paved the way to understanding the underlying mechanisms and development of successful treatments. These models utilize surgical techniques, genetic modifications, and pharmacological approaches. The present review discusses the strengths and limitations of commonly used small animal HF models, which continue to provide crucial insight and facilitate the development of new treatment strategies for patients with HF.
Collapse
Affiliation(s)
- Christian Riehle
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, Germany
| |
Collapse
|
23
|
Sobczuk P, Czerwińska M, Kleibert M, Cudnoch-Jędrzejewska A. Anthracycline-induced cardiotoxicity and renin-angiotensin-aldosterone system-from molecular mechanisms to therapeutic applications. Heart Fail Rev 2020; 27:295-319. [PMID: 32472524 PMCID: PMC8739307 DOI: 10.1007/s10741-020-09977-1] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Few millions of new cancer cases are diagnosed worldwide every year. Due to significant progress in understanding cancer biology and developing new therapies, the mortality rates are decreasing with many of patients that can be completely cured. However, vast majority of them require chemotherapy which comes with high medical costs in terms of adverse events, of which cardiotoxicity is one of the most serious and challenging. Anthracyclines (doxorubicin, epirubicin) are a class of cytotoxic agents used in treatment of breast cancer, sarcomas, or hematological malignancies that are associated with high risk of cardiotoxicity that is observed in even up to 30% of patients and can be diagnosed years after the therapy. The mechanism, in which anthracyclines cause cardiotoxicity are not well known, but it is proposed that dysregulation of renin-angiotensin-aldosterone system (RAAS), one of main humoral regulators of cardiovascular system, may play a significant role. There is increasing evidence that drugs targeting this system can be effective in the prevention and treatment of anthracycline-induced cardiotoxicity what has recently found reflection in the recommendation of some scientific societies. In this review, we comprehensively describe possible mechanisms how anthracyclines affect RAAS and lead to cardiotoxicity. Moreover, we critically review available preclinical and clinical data on use of RAAS inhibitors in the primary and secondary prevention and treatment of cardiac adverse events associated with anthracycline-based chemotherapy.
Collapse
Affiliation(s)
- Paweł Sobczuk
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland.,Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Magdalena Czerwińska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Marcin Kleibert
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Agnieszka Cudnoch-Jędrzejewska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland.
| |
Collapse
|
24
|
Luo L, Zeng F, Xie J, Fan J, Xiao S, Wang Z, Xie H, Liu B. A RBC membrane-camouflaged biomimetic nanoplatform for enhanced chemo-photothermal therapy of cervical cancer. J Mater Chem B 2020; 8:4080-4092. [PMID: 32239064 DOI: 10.1039/c9tb02937k] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Due to the untargeted release of chemical drugs, the efficacy of chemotherapy is often compromised along with serious side effects on patients. Recently, the development of targeted delivery systems using nanomaterials as carriers has provided more alternatives for chemical drug transportation. In this study, we developed a novel targeted nanocomplex of GOQD-ICG-DOX@RBCM-FA NPs (GID@RF NPs). First, PEG modified graphene oxide quantum dots (GOQDs) were used to co-load the photosensitizer of indocyanine green (ICG) and DOX, to form GOQD-ICG-DOX NPs (GID NPs). Then, the red blood cell membrane (RBCM) was applied for GID NP camouflage to avoid immune clearance. Finally, folic acid was used to endow the targeting ability of GID@RF NPs. MTT assay showed that the survival rate of HeLa cells reduced by 71% after treatment with GID@RF NPs and laser irradiation. Meanwhile, membrane camouflage significantly prolonged the blood circulation time and enhanced the immune evading ability of GID NPs. Moreover, the drug accumulation at tumor sites was significantly improved through the strong interaction between FA and FA receptor highly expressed on the tumor cells. In vivo assay demonstrated the strongest tumor growth inhibition ability of the combinational chemo/photothermal therapy. H&E analysis indicated no significant abnormalities in the major organs of mice undergoing GID@RF NPs treatment. The level of blood and biochemical parameters remained stable as compared to the control. In summary, this combinational therapy system provides a safe, rapid and effective alternative for the treatment of cervical cancer in the future.
Collapse
Affiliation(s)
- Lin Luo
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Institute of Cancer Research, School of Medicine, University of South China, Hengyang, Hunan 421001, China.
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Davis MK, Villa D, Tsang TS, Starovoytov A, Gelmon K, Virani SA. Effect of Eplerenone on Diastolic Function in Women Receiving Anthracycline-Based Chemotherapy for Breast Cancer. JACC CardioOncol 2019; 1:295-298. [PMID: 34396193 PMCID: PMC8352029 DOI: 10.1016/j.jaccao.2019.10.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Affiliation(s)
- Margot K. Davis
- Gordon & Leslie Diamond Health Care Centre, 2775 Laurel Street, 9th Floor-Cardiology, Vancouver, British Columbia V5Z 1M9, Canada @DktrV
| | | | | | | | | | | |
Collapse
|
26
|
Lódi M, Priksz D, Fülöp GÁ, Bódi B, Gyöngyösi A, Nagy L, Kovács Á, Kertész AB, Kocsis J, Édes I, Csanádi Z, Czuriga I, Kisvárday Z, Juhász B, Lekli I, Bai P, Tóth A, Papp Z, Czuriga D. Advantages of prophylactic versus conventionally scheduled heart failure therapy in an experimental model of doxorubicin-induced cardiomyopathy. J Transl Med 2019; 17:229. [PMID: 31324258 PMCID: PMC6642576 DOI: 10.1186/s12967-019-1978-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 07/08/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Chemotherapy-induced left ventricular dysfunction represents a major clinical problem, which is often only recognised at an advanced stage, when supportive therapy is ineffective. Although an early heart failure treatment could positively influence the health status and clinical outcome, there is still no evidence of routine prophylactic cardioprotection for the majority of patients without previous cardiovascular history awaiting potentially cardiotoxic chemotherapy. In this study, we set out to investigate whether a prophylactic cardioprotective therapy relative to a conventionally scheduled heart failure treatment is more effective in preventing cardiotoxicity in a rodent model of doxorubicin (DOX)-induced cardiomyopathy. METHODS Male Wistar rats (n = 7-11 per group) were divided into 4 subgroups, namely negative controls receiving intravenous saline (CON), positive controls receiving intravenous DOX (6 cycles; D-CON), and DOX-treated animals receiving either prophylactic (PRE, started 1 week before DOX) or conventionally applied (POST, started 1 month after DOX) combined heart failure therapy of oral bisoprolol, perindopril and eplerenone. Blood pressure, heart rate, body weight and echocardiographic parameters were monitored in vivo, whereas myocardial fibrosis, capillarisation, ultrastructure, myofilament function, apoptosis, oxidative stress and mitochondrial biogenesis were studied in vitro. RESULTS The survival rate in the PRE group was significantly improved compared to D-CON (p = 0.0207). DOX increased the heart rate of the animals (p = 0.0193), while the blood pressure (p ≤ 0.0105) and heart rate (p = 0.0029) were significantly reduced in the PRE group compared to D-CON and POST. The ejection fraction remained preserved in the PRE group compared to D-CON or POST (p ≤ 0.0237), while none of the treatments could prevent the DOX-induced increase in the isovolumetric relaxation time. DOX decreased the rate of the actin-myosin cross-bridge cycle, irrespective of any treatment applied (p ≤ 0.0433). The myocardium of the D-CON and POST animals displayed pronounced ultrastructural damage, which was not apparent in the PRE group (p ≤ 0.033). While the DOX-induced apoptotic activity could be reduced in both the PRE and POST groups (p ≤ 0.0433), no treatment was able to prevent fibrotic remodelling or the disturbed mitochondrial biogenesis. CONCLUSION For attenuating DOX-induced adverse myocardial effects, prophylactic cardioprotection has many advantages compared to a late-applied treatment.
Collapse
Affiliation(s)
- Mária Lódi
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Dániel Priksz
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gábor Áron Fülöp
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Beáta Bódi
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Alexandra Gyöngyösi
- Department of Pharmacology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Lilla Nagy
- MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, Hungary
| | - Árpád Kovács
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Attila Béla Kertész
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Móricz Zs. krt. 22, H-4032 Debrecen, Hungary
| | - Judit Kocsis
- Department of 3rd Internal Medicine, Semmelweis University, Budapest, Hungary
- Oncoradiology Center, Bács-Kiskun County Hospital, Kecskemét, Hungary
| | - István Édes
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Móricz Zs. krt. 22, H-4032 Debrecen, Hungary
| | - Zoltán Csanádi
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Móricz Zs. krt. 22, H-4032 Debrecen, Hungary
| | - István Czuriga
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Móricz Zs. krt. 22, H-4032 Debrecen, Hungary
| | - Zoltán Kisvárday
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Béla Juhász
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - István Lekli
- Department of Pharmacology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Péter Bai
- MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, Hungary
| | - Attila Tóth
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Papp
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Dániel Czuriga
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Móricz Zs. krt. 22, H-4032 Debrecen, Hungary
| |
Collapse
|
27
|
Hullin R, Métrich M, Sarre A, Basquin D, Maillard M, Regamey J, Martin D. Diverging effects of enalapril or eplerenone in primary prevention against doxorubicin-induced cardiotoxicity. Cardiovasc Res 2019; 114:272-281. [PMID: 29016737 DOI: 10.1093/cvr/cvx162] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 08/10/2017] [Indexed: 12/12/2022] Open
Abstract
Aims Clinical studies suggest beneficial effects of renin-angiotensin system blockade for prevention of left ventricular (LV) dysfunction after chemotherapy. However, the efficacy of this strategy as primary prevention has been poorly studied. This study aimed at identifying the pathophysiological mechanisms by which mineralocorticoid receptor antagonism (MRA) or angiotensin converting enzyme inhibition (ACEi) provide protection against doxorubicin-induced cardiotoxicity (DIC) in mouse models of acute and chronic toxicity. Methods and results Acute DIC was induced by a single injection of Dox at 15 mg/kg and chronic DIC applied 5 injections of Dox at 4 mg/kg/week. MRA was achieved using eplerenone or cardiomyocyte-specific ablation of the MR gene in transgenic mice and ACEi using enalapril. Drugs were provided with the first dose of Dox and applied until the end of the study. In both model of DIC, Dox induced cardiac atrophy with decreased LV volume, reduced cardiomyocyte cell size, and cardiac dysfunction. In the acute model, neither MRA nor ACEi protected against these manifestations of DIC. In the chronic model, concomitant treatment with eplerenone did not protect against DIC and drastically increased plasma aldosterone levels and cardiac levels of angiotensin II type 1 receptor and of connective tissue growth factor (CTGF), as observed in acute DIC. Enalapril treatment in the chronic model, however, protected against cardiac dysfunction and cardiomyocyte atrophy and was associated with increased activation of the PI3K/AKT/mTOR pathway along with normal levels of CTGF. Conclusion Enalapril and eplerenone disparately impact on cellular signalling in DIC. Eplerenone, on top of Dox treatment was not protective and associated with increased levels of plasma aldosterone and of cardiac CTGF. In contrast, we show that primary prevention with enalapril preserves LV morphology and function in a clinically relevant model of chronic DIC, with increased stimulation of the PI3K/AKT/mTOR axis and normal CTGF levels suggesting potential therapeutic implications.
Collapse
Affiliation(s)
- Roger Hullin
- Service of Cardiology, Cardiovascular Department, Lausanne University Hospital (CHUV) Lausanne, Switzerland
| | - Mélanie Métrich
- Service of Cardiology, Cardiovascular Department, Lausanne University Hospital (CHUV) Lausanne, Switzerland
| | - Alexandre Sarre
- Cardiovascular Assessment Facility, University of Lausanne, Lausanne, Switzerland
| | - Denis Basquin
- Service of Cardiology, Cardiovascular Department, Lausanne University Hospital (CHUV) Lausanne, Switzerland
| | - Marc Maillard
- Service of Nephrology, Medicine Department, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Julien Regamey
- Service of Cardiology, Cardiovascular Department, Lausanne University Hospital (CHUV) Lausanne, Switzerland
| | - David Martin
- Service of Cardiology, Cardiovascular Department, Lausanne University Hospital (CHUV) Lausanne, Switzerland
| |
Collapse
|
28
|
Ameri P, Canepa M, Anker MS, Belenkov Y, Bergler-Klein J, Cohen-Solal A, Farmakis D, López-Fernández T, Lainscak M, Pudil R, Ruschitska F, Seferovic P, Filippatos G, Coats A, Suter T, Von Haehling S, Ciardiello F, de Boer RA, Lyon AR, Tocchetti CG. Cancer diagnosis in patients with heart failure: epidemiology, clinical implications and gaps in knowledge. Eur J Heart Fail 2018; 20:879-887. [PMID: 29464808 DOI: 10.1002/ejhf.1165] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/24/2017] [Accepted: 01/24/2018] [Indexed: 12/15/2022] Open
Abstract
Cancer and heart failure (HF) are common medical conditions with a steadily rising prevalence in industrialized countries, particularly in the elderly, and they both potentially carry a poor prognosis. A new diagnosis of malignancy in subjects with pre-existing HF is not infrequent, and challenges HF specialists as well as oncologists with complex questions relating to both HF and cancer management. An increased incidence of cancer in patients with established HF has also been suggested. This review paper summarizes the epidemiology and the prognostic implications of cancer occurrence in HF, the impact of pre-existing HF on cancer treatment decisions and the impact of cancer on HF therapeutic options, while providing some practical suggestions regarding patient care and highlighting gaps in knowledge.
Collapse
Affiliation(s)
- Pietro Ameri
- Department of Internal Medicine, University of Genova; and Cardiology Unit, IRCCS Policlinic Hospital San Martino, Genova, Italy
| | - Marco Canepa
- Department of Internal Medicine, University of Genova; and Cardiology Unit, IRCCS Policlinic Hospital San Martino, Genova, Italy
| | - Markus S Anker
- Department of Cardiology (CBF), Charité University Medicine, Berlin, Germany; AND Division of Cardiology and Metabolism - Heart Failure, Cachexia & Sarcopenia; Department of Internal Medicine & Cardiology; and Berlin-Brandenburg Center for Regenerative Therapies (BCRT), at Charité University Medicine, Berlin, Germany
| | | | | | - Alain Cohen-Solal
- Department of Cardiology, Lariboisière Hospital, Paris, France; U942 INSERM, BIOCANVAS (Biomarqueurs Cardiovasculaires), Paris, France;, Department of Cardiology, University of Paris VII Denis Diderot, Paris, France
| | - Dimitrios Farmakis
- Cardio-Oncology Clinic, Heart Failure Unit, Department of Cardiology, Athens University Hospital 'Attikon', National and Kapodistrian University of Athens, Athens, Greece
| | - Teresa López-Fernández
- Cardio-Oncology Unit, Cardiac Imaging Unit, Department of Cardiology, La Paz University Hospital, IdiPAz, Madrid, Spain
| | - Mitja Lainscak
- Department of Cardiology, Department of Research and Education, General Hospital Celje, Celje, Slovenia, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Radek Pudil
- 1st Department of Medicine - Cardioangiology, Medical Faculty and University Hospital Hradec Kralove, Czech Republic
| | - Frank Ruschitska
- University Heart Center, University Hospital Zurich, Zurich, Switzerland
| | | | - Gerasimos Filippatos
- Cardio-Oncology Clinic, Heart Failure Unit, Department of Cardiology, Athens University Hospital 'Attikon', National and Kapodistrian University of Athens, Athens, Greece
| | - Andrew Coats
- Monash University, Australia and University of Warwick, UK
| | - Thomas Suter
- Department of Cardiology, Cardio-Oncology, Bern University Hospital, Bern, Switzerland
| | - Stephan Von Haehling
- Klinik für Kardiologie und Pneumologie, Herzzentrum Göttingen, Universitätsmedizin Göttingen, Georg-August-Universität, Göttingen, Germany;, Deutsches Zentrum für Herz- und Kreislaufforschung, Standort Göttingen, Göttingen, Germany
| | - Fortunato Ciardiello
- Department of Clinical and Experimental Medicine "Flaviano Magrassi", Luigi Vanvitelli University of Campania, Naples, Italy
| | - Rudolf A de Boer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | - Carlo G Tocchetti
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | | |
Collapse
|
29
|
Lipshultz SE, Herman EH. Anthracycline cardiotoxicity: the importance of horizontally integrating pre-clinical and clinical research. Cardiovasc Res 2018; 114:205-209. [PMID: 29272330 PMCID: PMC5852510 DOI: 10.1093/cvr/cvx246] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Steven E Lipshultz
- Department of Pediatrics, Wayne State University School of Medicine, Karmanos Cancer Institute, and Children’s Hospital of Michigan, 3901 Beaubien Boulevard, Pediatric Administration-T121A, Detroit, MI 48201, USA
| | - Eugene H Herman
- Toxicology and Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, The National Cancer Institute, Rockville, MD 20850-9734, USA
| |
Collapse
|