1
|
Borella F, Fucina S, Seminara Y, Denti P, Ferraioli D, Bertero L, Gallio N, Cusato J, Valabrega G, Revelli A, Marozio L, Cosma S. Targeting TOP2A in Ovarian Cancer: Biological and Clinical Implications. Curr Oncol 2024; 31:8054-8074. [PMID: 39727717 DOI: 10.3390/curroncol31120594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/09/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024] Open
Abstract
The enzyme topoisomerase II alpha (TOP2A) plays a critical role in DNA replication and cell proliferation, making it a promising target for cancer therapy. In epithelial ovarian cancer (EOC), TOP2A overexpression is associated with poor prognosis and resistance to conventional treatments. This review explores the biological functions of TOP2A in EOC and discusses its potential as a therapeutic target. We highlight studies on the mechanisms through which TOP2A contributes to tumor progression and recurrence. Additionally, we evaluate the clinical implications of targeting TOP2A, including the use of TOP2A inhibitors and their combination with novel drugs. We provide a comprehensive overview of the current understanding and future directions for targeting TOP2A in the management of EOC.
Collapse
Affiliation(s)
- Fulvio Borella
- Gynecology and Obstetrics 1U, Department of Surgical Sciences, University of Turin, 10126 Turin, Italy
| | - Stefano Fucina
- Gynecology and Obstetrics 1U, Department of Surgical Sciences, University of Turin, 10126 Turin, Italy
| | - Ylenia Seminara
- Gynecology and Obstetrics 1U, Department of Surgical Sciences, University of Turin, 10126 Turin, Italy
| | - Pietro Denti
- Gynecology and Obstetrics 1U, Department of Surgical Sciences, University of Turin, 10126 Turin, Italy
| | - Domenico Ferraioli
- Department of Gynecology, Léon Bérard, Comprehensive Cancer Centre, 69008 Lyon, France
| | - Luca Bertero
- Pathology Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy
| | - Niccolò Gallio
- Gynecology and Obstetrics 2U, Department of Surgical Sciences, University of Turin, 10126 Turin, Italy
| | - Jessica Cusato
- Laboratory of Clinical Pharmacology and Pharmacogenetics, Department of Medical Sciences, University of Turin, 10149 Turin, Italy
| | - Giorgio Valabrega
- Department of Oncology, University of Turin, Medical Oncology, Ordine Mauriziano Hospital, 10128 Turin, Italy
| | - Alberto Revelli
- Gynecology and Obstetrics 2U, Department of Surgical Sciences, University of Turin, 10126 Turin, Italy
| | - Luca Marozio
- Gynecology and Obstetrics 1U, Department of Surgical Sciences, University of Turin, 10126 Turin, Italy
| | - Stefano Cosma
- Gynecology and Obstetrics 1U, Department of Surgical Sciences, University of Turin, 10126 Turin, Italy
| |
Collapse
|
2
|
Belger C, Abrahams C, Imamdin A, Lecour S. Doxorubicin-induced cardiotoxicity and risk factors. IJC HEART & VASCULATURE 2024; 50:101332. [PMID: 38222069 PMCID: PMC10784684 DOI: 10.1016/j.ijcha.2023.101332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 12/14/2023] [Accepted: 12/16/2023] [Indexed: 01/16/2024]
Abstract
Doxorubicin (DOX) is an anthracycline antibiotic widely used as a chemotherapeutic agent to treat solid tumours and hematologic malignancies. Although useful in the treatment of cancers, the benefit of DOX is limited due to its cardiotoxic effect that is observed in a large number of patients. In the literature, there is evidence that the presence of various factors may increase the risk of developing DOX-induced cardiotoxicity. A better understanding of the role of these different factors in DOX-induced cardiotoxicity may facilitate the choice of the therapeutic approach in cancer patients suffering from various cardiovascular risk factors. In this review, we therefore discuss the latest findings in both preclinical and clinical research suggesting a link between DOX-induced cardiotoxicity and various risk factors including sex, age, ethnicity, diabetes, dyslipidaemia, obesity, hypertension, cardiovascular disease and co-medications.
Collapse
Affiliation(s)
| | | | - Aqeela Imamdin
- Cardioprotection Group, Cape Heart Institute, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Sandrine Lecour
- Cardioprotection Group, Cape Heart Institute, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
3
|
Yu Y, Zhou T, Cao L. Use and application of organ-on-a-chip platforms in cancer research. J Cell Commun Signal 2023:10.1007/s12079-023-00790-7. [PMID: 38032444 DOI: 10.1007/s12079-023-00790-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 10/31/2023] [Indexed: 12/01/2023] Open
Abstract
Tumors are a major cause of death worldwide, and much effort has been made to develop appropriate anti-tumor therapies. Existing in vitro and in vivo tumor models cannot reflect the critical features of cancer. The development of organ-on-a-chip models has enabled the integration of organoids, microfluidics, tissue engineering, biomaterials research, and microfabrication, offering conditions that mimic tumor physiology. Three-dimensional in vitro human tumor models that have been established as organ-on-a-chip models contain multiple cell types and a structure that is similar to the primary tumor. These models can be applied to various foci of oncology research. Moreover, the high-throughput features of microfluidic organ-on-a-chip models offer new opportunities for achieving large-scale drug screening and developing more personalized treatments. In this review of the literature, we explore the development of organ-on-a-chip technology and discuss its use as an innovative tool in basic and clinical applications and summarize its advancement of cancer research.
Collapse
Affiliation(s)
- Yifan Yu
- Department of Hepatobiliary and Transplant Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - TingTing Zhou
- The College of Basic Medical Science, Health Sciences Institute, Key Laboratory of Cell Biology of Ministry of Public Health, Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Liu Cao
- The College of Basic Medical Science, Health Sciences Institute, Key Laboratory of Cell Biology of Ministry of Public Health, Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China.
| |
Collapse
|
4
|
Afonso AI, Amaro-Leal Â, Machado F, Rocha I, Geraldes V. Doxorubicin Dose-Dependent Impact on Physiological Balance-A Holistic Approach in a Rat Model. BIOLOGY 2023; 12:1031. [PMID: 37508460 PMCID: PMC10376408 DOI: 10.3390/biology12071031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/13/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023]
Abstract
Doxorubicin (DOX) is commonly used in several chemotherapies to treat various cancers, but it is known to cause cardiotoxicity and cardiac symptoms. Autonomic dysfunction is thought to contribute to the cardiotoxic effects of DOX, but the specific dose required to disrupt homeostatic processes is still unclear and is influenced by numerous factors. This study aimed to investigate how the DOX dosage affects autonomic function and physiological parameters, to elucidate the neurocardiac mechanisms underlying the observed cardiovascular side effects. Wistar rats were treated with DOX for four weeks and divided into three dosing groups: DOX8 (2 mg/kg/week), DOX16 (4 mg/kg/week), and DOX20 (5 mg/kg/week). A control group received NaCl 0.9% saline (1 mL/kg/week). In an acute experiment, we recorded blood pressure (BP), electrocardiogram, heart rate (HR), and respiratory rate (RF). Baroreflex gain and chemoreflex sensitivity were calculated, and cardiac tissue was analyzed with picrosirius histochemistry to measure collagen content. Our results showed that the LF/HF ratio, indicative of autonomic activity, was altered along with hypotension and bradycardia at a cumulative DOX dose threshold of 16 mg/kg. We observed a positive correlation between DOX dose and BP, HR, urinary norepinephrine, LF/HF ratio, and fibrotic heart area. Lower LF/HF ratios were associated with high DOX doses, reflecting drug-induced impairment of autonomic control of HR. This study provides valuable insights into the dose-dependent effects of DOX on physiological parameters and the development of cardiovascular dysfunction. These findings are critical, which is important for optimizing the management and therapeutic strategies for patients undergoing DOX-based chemotherapy.
Collapse
Affiliation(s)
- Ana I Afonso
- Cardiovascular Centre of the University of Lisbon, 1649-028 Lisbon, Portugal
| | - Ângela Amaro-Leal
- Cardiovascular Centre of the University of Lisbon, 1649-028 Lisbon, Portugal
- Egas Moniz School of Health and Science, 2829-511 Caparica, Portugal
| | - Filipa Machado
- Cardiovascular Centre of the University of Lisbon, 1649-028 Lisbon, Portugal
| | - Isabel Rocha
- Cardiovascular Centre of the University of Lisbon, 1649-028 Lisbon, Portugal
- Institute of Physiology, Faculty of Medicine of the University of Lisbon, 1649-028 Lisbon, Portugal
| | - Vera Geraldes
- Cardiovascular Centre of the University of Lisbon, 1649-028 Lisbon, Portugal
- Institute of Physiology, Faculty of Medicine of the University of Lisbon, 1649-028 Lisbon, Portugal
| |
Collapse
|
5
|
Hwang S, Kim SH, Yoo KH, Chung MH, Lee JW, Son KH. Exogenous 8-hydroxydeoxyguanosine attenuates doxorubicin-induced cardiotoxicity by decreasing pyroptosis in H9c2 cardiomyocytes. BMC Mol Cell Biol 2022; 23:55. [DOI: 10.1186/s12860-022-00454-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/25/2022] [Indexed: 12/15/2022] Open
Abstract
AbstractDoxorubicin (DOX), which is widely used in cancer treatment, can induce cardiomyopathy. One of the main mechanisms whereby DOX induces cardiotoxicity involves pyroptosis through the NLR family pyrin domain containing 3 (NLRP3) inflammasome and gasdermin D (GSDMD). Increased NAPDH oxidase (NOX) and oxidative stress trigger pyroptosis. Exogenous 8-hydroxydeoxyguanosine (8-OHdG) decreases reactive oxygen species (ROS) production by inactivating NOX. Here, we examined whether 8-OHdG treatment can attenuate DOX-induced pyroptosis in H9c2 cardiomyocytes. Exposure to DOX increased the peroxidative glutathione redox status and NOX1/2/4, toll-like receptor (TLR)2/4, and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) expression, while an additional 8-OHdG treatment attenuated these effects. Furthermore, DOX induced higher expression of NLRP3 inflammasome components, including NLRP3, apoptosis-associated speck-like protein containing a c-terminal caspase recruitment domain (ASC), and pro-caspase-1. Moreover, it increased caspase-1 activity, a marker of pyroptosis, and interleukin (IL)-1β expression. All these effects were attenuated by 8-OHdG treatment. In addition, the expression of the cardiotoxicity markers, atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) was increased by DOX, whereas the increase of ANP and BNP induced by DOX treatment was reversed by 8-OHdG. In conclusion, exogenous 8-OHdG attenuated DOX-induced pyroptosis by decreasing the expression of NOX1/2/3, TLR2/4, and NF-κB. Thus, 8-OHdG may attenuate DOX-induced cardiotoxicity through the inhibition of pyroptosis.
Collapse
|
6
|
Yang JY, Woo HJ, Lee P, Kim SH. Induction of Apoptosis and Effect on the FAK/AKT/mTOR Signal Pathway by Evodiamine in Gastric Cancer Cells. Curr Issues Mol Biol 2022; 44:4339-4349. [PMID: 36135210 PMCID: PMC9497533 DOI: 10.3390/cimb44090298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 12/24/2022] Open
Abstract
Evodiamine isolated from Evodia rutaecarpa has been known to have anti-tumor activity against various cancer cell types. Although there have been reports showing the inhibitory effect of evodiamine on cell survival of gastric cancer cell, it is not clearly explained how evodiamine affects the expression and modification of proteins associated with apoptosis and upstream signal pathways. We confirmed the cytotoxic activity of evodiamine against AGS and MKN45 cells by a WST assay, cell morphological change, and clonogenic assay. The apoptotic cells were evaluated by Annexin V/PI analysis and Western blot and the expressions of apoptosis-related molecules were confirmed by Western blot. Evodiamine promoted apoptosis of AGS gastric cancer cells through both intrinsic and extrinsic signal pathways in a time- and dose-dependent manner. Evodiamine attenuated the expression of anti-apoptotic proteins, including Bcl-2, XIAP, and survivin, and elevated that of the pro-apoptotic protein Bax. Evodiamine also suppressed the FAK/AKT/mTOR signal pathway. Based on these results, we expect that the results from this study will further elucidate our understanding of evodiamine as an anti-cancer drug.
Collapse
Affiliation(s)
- Ji Yeong Yang
- Crop Foundation Research Division, National Institute of Crop Science (NICS), Rural Development Administration (RDA), Wanju 55365, Korea
| | - Hyun Jun Woo
- Department of Clinical Laboratory Science, Semyung University, Jecheon 27136, Korea
| | - Pyeongjae Lee
- School of Industrial Bio-Pharmaceutical Science, Semyung University, Jecheon 27136, Korea
- Correspondence: (P.L.); (S.-H.K.)
| | - Sa-Hyun Kim
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Korea
- Correspondence: (P.L.); (S.-H.K.)
| |
Collapse
|
7
|
Fu B, Lin HC, Chen N, Zhao P. Adenosine triphosphate/pH dual-responsive controlled drug release system with high cancer/normal cell selectivity and low side toxicity. J Biomater Appl 2022; 37:324-332. [DOI: 10.1177/08853282221087412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Most drug-delivery systems (DDS) suffer from poor selectivity to cancer/normal cells or the complicated synthetic process. Herein, we employed a novel facile method to develop an oligodeoxy nucleotides based DDS composed with adenosine-5′- triphosphate (ATP) aptamer and a pH responsive cytosine (C) DNA fragment for specific daunomycine (DNM) delivery. The DDS has ATP/pH dual-responsive drug release, can selectively internalize into tumor cell lines and thus has ultrahigh cancer/normal cell selectivity over the individual drug. The non-chemical synthesis, controllable dual-responsive intracellular drug release, and high cancer/normal cell selectivity endowed the DDS high biocompatibility and significant tumor suppression.
Collapse
Affiliation(s)
- Bo Fu
- College of Health Industry, Zhongshan Torch Polytechnic, Zhongshan, China
| | - Hui-Chao Lin
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Guangzhou, China
| | - Nian Chen
- College of Health Industry, Zhongshan Torch Polytechnic, Zhongshan, China
| | - Ping Zhao
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
8
|
Zhong R, Zhang F, Yang Z, Li Y, Xu Q, Lan H, Lang S, Cyganek L, Burgermeister E, El-Battrawy I, Zhou X, Akin I, Borggrefe M. Regulation of Ion Channel Function in Human-Induced Pluripotent Stem Cell-Derived Cardiomyocytes by Cancer Cell Secretion Through DNA Methylation. Front Cardiovasc Med 2022; 9:839104. [PMID: 35265687 PMCID: PMC8899119 DOI: 10.3389/fcvm.2022.839104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/28/2022] [Indexed: 12/24/2022] Open
Abstract
Background Cardiac dysfunction including arrhythmias appear frequently in patients with cancers, which are expected to be caused mainly by cardiotoxic effects of chemotherapy. Experimental studies investigating the effects of cancer cell secretion without chemotherapy on ion channel function in human cardiomyocytes are still lacking. Methods The human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) generated from three healthy donors were treated with gastrointestinal (GI) cancer (AGS and SW480 cells) medium for 48 h. The qPCR, patch-clamp, western blotting, immunostaining, dot blotting, bisulfite sequence, and overexpression of the ten-eleven translocation (TET) enzyme were performed for the study. Results After treated with cancer cell secretion, the maximum depolarization velocity and the action potential amplitude were reduced, the action potential duration prolonged, peak Na+ current, and the transient outward current were decreased, late Na+ and the slowly activating delayed rectifier K+ current were increased. Changes of mRNA and protein level of respective channels were detected along with altered DNA methylation level in CpG island in the promoter regions of ion channel genes and increased protein levels of DNA methyltransferases. Phosphoinositide 3-kinase (PI3K) inhibitor attenuated and transforming growth factor-β (TGF-β) mimicked the effects of cancer cell secretion. Conclusions GI cancer cell secretion could induce ion channel dysfunction, which may contribute to occurrence of arrhythmias in cancer patients. The ion channel dysfunction could result from DNA methylation of ion channel genes via activation of TGF-β/PI3K signaling. This study may provide new insights into pathogenesis of arrhythmia in cancer patients.
Collapse
Affiliation(s)
- Rujia Zhong
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Feng Zhang
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Zhen Yang
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Yingrui Li
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Qiang Xu
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Huan Lan
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Siegfried Lang
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site, Mannheim, Germany
| | - Lukas Cyganek
- Stem Cell Unit, Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site, Göttingen, Germany
| | - Elke Burgermeister
- Second Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Ibrahim El-Battrawy
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site, Mannheim, Germany
| | - Xiaobo Zhou
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
- DZHK (German Center for Cardiovascular Research), Partner Site, Mannheim, Germany
- *Correspondence: Xiaobo Zhou
| | - Ibrahim Akin
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site, Mannheim, Germany
| | - Martin Borggrefe
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site, Mannheim, Germany
| |
Collapse
|
9
|
Béland J, Duverger JE, Comtois P. Novel Analysis Method for Beating Cells Videomicroscopy Data: Functional Characterization of Culture Samples. Front Physiol 2022; 13:733706. [PMID: 35242049 PMCID: PMC8886216 DOI: 10.3389/fphys.2022.733706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 01/06/2022] [Indexed: 11/13/2022] Open
Abstract
Cell culture of cardiac tissue analog is becoming increasingly interesting for regenerative medicine (cell therapy and tissue engineering) and is widely used for high throughput cardiotoxicity. As a cost-effective approach to rapidly discard new compounds with high toxicity risks, cardiotoxicity evaluation is firstly done in vitro requiring cells/tissue with physiological/pathological characteristics (close to in vivo properties). Studying multicellular electrophysiological and contractile properties is needed to assess drug effects. Techniques favoring process automation which could help in simplifying screening drug candidates are thus of central importance. A lot of effort has been made to ameliorate in vitro models including several in vitro platforms for engineering neonatal rat cardiac tissues. However, most of the initial evaluation is done by studying the rate of activity. In this study, we present new approaches that use the videomicroscopy video of monolayer activity to study contractile properties of beating cells in culture. Two new variables are proposed which are linked to the contraction dynamics and are dependent on the rhythm of activity. Methods for evaluation of regional synchronicity within the image field of view are also presented that can rapidly determine regions with abnormal activity or heterogeneity in contraction dynamics.
Collapse
Affiliation(s)
- Jonathan Béland
- Research Centre, Montreal Heart Institute, Montreal, QC, Canada
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada
| | - James Elber Duverger
- Research Centre, Montreal Heart Institute, Montreal, QC, Canada
- Institute of Biomedical Engineering, Université de Montréal, Montreal, QC, Canada
| | - Philippe Comtois
- Research Centre, Montreal Heart Institute, Montreal, QC, Canada
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada
- Institute of Biomedical Engineering, Université de Montréal, Montreal, QC, Canada
- *Correspondence: Philippe Comtois,
| |
Collapse
|
10
|
Biswas S, Datta LP, Kumar Das T. A bioinspired stimuli-responsive amino acid-based antibacterial drug delivery system in cancer therapy. NEW J CHEM 2022. [DOI: 10.1039/d2nj00815g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Design of tyrosine based stimuli responsive antibacterial drug delivery system with potential application in cancer therapy.
Collapse
Affiliation(s)
- Subharanjan Biswas
- Department of Biochemistry & Biophysics, University of Kalyani, Kalyani, Nadia - 741235, Nadia, West Bengal, India
- Institut Lavoisier de Versailles, UMR CNRS 8180, Université de Versailles St Quentin en Yvelines, Université Paris Saclay, 45 avenue des Etats-Unis, Versailles 78035, France
| | - Lakshmi Priya Datta
- Department of Biochemistry & Biophysics, University of Kalyani, Kalyani, Nadia - 741235, Nadia, West Bengal, India
| | - Tapan Kumar Das
- Department of Biochemistry & Biophysics, University of Kalyani, Kalyani, Nadia - 741235, Nadia, West Bengal, India
| |
Collapse
|
11
|
Adıyaman MŞ, Adıyaman ÖA, Dağlı AF, Karahan MZ, Dağlı MN. Prevention of doxorubicin-induced experimental cardiotoxicity by Nigella sativa in rats. Rev Port Cardiol 2022; 41:99-105. [DOI: 10.1016/j.repc.2020.12.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 12/17/2020] [Accepted: 12/30/2020] [Indexed: 11/26/2022] Open
|
12
|
Tereshkina YA, Torkhovskaya TI, Sanzhakov MA, Kostryukova LV, Khudoklinova YY, Tikhonova EG. [The effect of lipid derivative of anti-tumor drug sarcolysin embedded in phospholipid nanoparticles in the experiments in vivo]. BIOMEDITSINSKAIA KHIMIIA 2021; 67:491-499. [PMID: 34964443 DOI: 10.18097/pbmc20216706491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
To improve the therapeutic properties of the antitumor agent Sarcolysin, we have previously developed and characterized a dosage form representing its ester conjugate with decanol embedded in ultra-small phospholipid nanoparticles less than 30 nm in size ("Sarcolysin-NP"). The effect of the resulting composition was investigated in vivo in comparison with the free substance of sarcolysin. The composition intravenous administration to mice showed an improvement in the pharmacokinetic parameters of sarcolysin associated with its initial higher (by 22%) level in the blood and prolonged circulation, which was also observed in mice with P388 tumor. In mice with three types of tumors - lymphocytic leukemia P388, lymphocytic leukemia L1210, and adenocarcinoma of the mammary gland Ca755 - administration of two doses of sarcolysin over a period of 7 days showed its predominant antitumor effect. The maximum tumor growth inhibition was noted for lymphocytic leukemia L1210 and adenocarcinoma of the mouse mammary gland Ca755 (at a dose of Sarcolysin-NP - 8,4 mg/kg), which was higher in comparison with free substance by more than 24% and 17%, respectively. Differences in the life span of the treated animals were revealed significantly at a dose of 10 mg/kg and amounted to 25% and 17,4% for lymphocytic leukemia P388 and L1210, respectively, and 11% for adenocarcinoma Ca755. In an experiment on rats, acute toxicity of Sarcolysin-NP administered intravenously showed that an average LD50 value 2-3 times exceeded a similar parameter for commercial preparations of free sarcolysin (Melphalan and Alkeran), which indicates its lower toxicity.
Collapse
|
13
|
Keshmiri Neghab H, Soheilifar MH, Saboury AA, Goliaei B, Hong J, Esmaeeli Djavid G. Optogenetic Stimulation of Primary Cardiomyocytes Expressing ChR2. J Lasers Med Sci 2021; 12:e32. [PMID: 34733755 PMCID: PMC8558726 DOI: 10.34172/jlms.2021.32] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 12/02/2020] [Indexed: 11/09/2022]
Abstract
Introduction: Non-clinical cardiovascular drug safety assessment is the main step in the progress of new pharmaceutical products. Cardiac drug safety testing focuses on a delayed rectifier potassium channel block and QT interval prolongation, whereas optogenetics is a powerful technology for modulating the electrophysiological properties of excitable cells. Methods: For this purpose, the blue light-gated ion channel, channelrhodopsin-2 (ChR2), has been introduced into isolated primary neonatal cardiomyocytes via a lentiviral vector. After being subjected to optical stimulation, transmembrane potential and intracellular calcium were assessed. Results: Here, we generated cardiomyocytes expressing ChR2 (light-sensitive protein), that upon optical stimulation, the cardiomyocytes depolarized result from alterations of membrane voltage and intracellular calcium. Conclusion: This cell model was easily adapted to a cell culture system in a laboratory, making this method very attractive for therapeutic research on cardiac optogenetics.
Collapse
Affiliation(s)
- Hoda Keshmiri Neghab
- Department of Photo Healing and Regeneration, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran
- Institutes of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Mohammad Hasan Soheilifar
- Department of Medical Laser, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran
- Research Center for Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ali Akbar Saboury
- Institutes of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Bahram Goliaei
- Institutes of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Jun Hong
- School of Life Sciences, Henan University, China
| | - Gholamreza Esmaeeli Djavid
- Department of Photo Healing and Regeneration, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran
| |
Collapse
|
14
|
Xu D, Mo J, Xie X, Hu N. In-Cell Nanoelectronics: Opening the Door to Intracellular Electrophysiology. NANO-MICRO LETTERS 2021; 13:127. [PMID: 34138366 PMCID: PMC8124030 DOI: 10.1007/s40820-021-00655-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/13/2021] [Indexed: 05/07/2023]
Abstract
Establishing a reliable electrophysiological recording platform is crucial for cardiology and neuroscience research. Noninvasive and label-free planar multitransistors and multielectrode arrays are conducive to perform the large-scale cellular electrical activity recordings, but the signal attenuation limits these extracellular devices to record subthreshold activities. In recent decade, in-cell nanoelectronics have been rapidly developed to open the door to intracellular electrophysiology. With the unique three-dimensional nanotopography and advanced penetration strategies, high-throughput and high-fidelity action potential like signal recordings is expected to be realized. This review summarizes in-cell nanoelectronics from versatile nano-biointerfaces, penetration strategies, active/passive nanodevices, systematically analyses the applications in electrogenic cells and especially evaluates the influence of nanodevices on the high-quality intracellular electrophysiological signals. Further, the opportunities, challenges and broad prospects of in-cell nanoelectronics are prospected, expecting to promote the development of in-cell electrophysiological platforms to meet the demand of theoretical investigation and clinical application.
Collapse
Affiliation(s)
- Dongxin Xu
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China
| | - Jingshan Mo
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China
| | - Xi Xie
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China
- The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China
| | - Ning Hu
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China.
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai, 200050, People's Republic of China.
| |
Collapse
|
15
|
Cadeddu Dessalvi C, Deidda M, Noto A, Madeddu C, Cugusi L, Santoro C, López-Fernández T, Galderisi M, Mercuro G. Antioxidant Approach as a Cardioprotective Strategy in Chemotherapy-Induced Cardiotoxicity. Antioxid Redox Signal 2021; 34:572-588. [PMID: 32151144 DOI: 10.1089/ars.2020.8055] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Significance: Chemotherapy-induced cardiotoxicity (CTX) has been associated with redox signaling imbalance. In fact, redox reactions are crucial for normal heart physiology, whereas excessive oxidative stress can cause cardiomyocyte structural damage. Recent Advances: An antioxidant approach as a cardioprotective strategy in this setting has shown encouraging results in preventing anticancer drug-induced CTX. Critical Issues: In fact, traditional heart failure drugs as well as many other compounds and nonpharmacological strategies, with a partial effect in reducing oxidative stress, have been shown to counterbalance chemotherapy-induced CTX in this setting to some extent. Future Directions: Given the various pathways of toxicity involved in different chemotherapeutic schemes, interactions with redox balance need to be fine-tuned and a personalized cardioprotective approach seems to be required.
Collapse
Affiliation(s)
| | - Martino Deidda
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Antonio Noto
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Clelia Madeddu
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Lucia Cugusi
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Ciro Santoro
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Teresa López-Fernández
- Cardiology Service, Cardio-Oncology Unit, La Paz University Hospital, IdiPAz Research Institute, Ciber CV, Madrid, Spain
| | - Maurizio Galderisi
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Giuseppe Mercuro
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| |
Collapse
|
16
|
Carrasco R, Castillo RL, Gormaz JG, Carrillo M, Thavendiranathan P. Role of Oxidative Stress in the Mechanisms of Anthracycline-Induced Cardiotoxicity: Effects of Preventive Strategies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8863789. [PMID: 33574985 PMCID: PMC7857913 DOI: 10.1155/2021/8863789] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/29/2020] [Accepted: 12/31/2020] [Indexed: 12/15/2022]
Abstract
Anthracycline-induced cardiotoxicity (AIC) persists as a significant cause of morbidity and mortality in cancer survivors. Although many protective strategies have been evaluated, cardiotoxicity remains an ongoing threat. The mechanisms of AIC remain unclear; however, several pathways have been proposed, suggesting a multifactorial origin. When the central role of topoisomerase 2β in the pathophysiology of AIC was described some years ago, the classical reactive oxygen species (ROS) hypothesis shifted to a secondary position. However, new insights have reemphasized the importance of the role of oxidative stress-mediated signaling as a common pathway and a critical modulator of the different mechanisms involved in AIC. A better understanding of the mechanisms of cardiotoxicity is crucial for the development of treatment strategies. It has been suggested that the available therapeutic interventions for AIC could act on the modulation of oxidative balance, leading to a reduction in oxidative stress injury. These indirect antioxidant effects make them an option for the primary prevention of AIC. In this review, our objective is to provide an update of the accumulated knowledge on the role of oxidative stress in AIC and the modulation of the redox balance by potential preventive strategies.
Collapse
Affiliation(s)
- Rodrigo Carrasco
- Division of Cardiology, Peter Munk Cardiac Centre and the Ted Rogers Centre for Heart Research, University Health Network, Toronto, Ontario, Canada
| | - Rodrigo L. Castillo
- Medicine Department, East Division, Faculty of Medicine, University of Chile. Santiago, Chile; Critical Care Patient Unit, Hospital Salvador, Santiago, Chile
| | - Juan G. Gormaz
- Faculty of Medicine, University of Chile, Santiago, Chile
| | - Montserrat Carrillo
- Division of Cardiology, Peter Munk Cardiac Centre and the Ted Rogers Centre for Heart Research, University Health Network, Toronto, Ontario, Canada
| | - Paaladinesh Thavendiranathan
- Division of Cardiology, Peter Munk Cardiac Centre and the Ted Rogers Centre for Heart Research, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
17
|
Cardiotoxicity: A Major Setback in Childhood Leukemia Treatment. DISEASE MARKERS 2021; 2021:8828410. [PMID: 33505537 PMCID: PMC7810535 DOI: 10.1155/2021/8828410] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 11/15/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023]
Abstract
Ongoing research in the field of pediatric oncology has led to an increased number of childhood cancer survivors reaching adulthood. Therefore, ensuring a good quality of life for these patients has become a rising priority. Considering this, the following review focuses on summarizing the most recent research in anthracycline-induced cardiac toxicity in children treated for leukemia. For pediatric cancers, anthracyclines are one of the most used anticancer drugs, with over half of the childhood cancer survivors believed to have been exposed to them. Anthracyclines cause irreversible cardiomyocyte loss, leading to chronic, progressive heart failure. The risk of developing cardiotoxicity has been known to increase with the treatment-free interval and total cumulative dose. However, because of individual variations in anthracycline metabolism, it has recently been shown that there is no risk-free dose. Moreover, studies have shown that diagnosing anthracycline-induced cardiomyopathy in the symptomatic phase is associated with poor treatment response and prognosis. Thus, early and systematic evaluation of these patients is crucial to allow optimal therapeutic intervention. Although currently echocardiographic assessment of left ventricle ejection fraction and cardiac biomarker evaluation are being used for cardiac function monitoring in oncologic patients, there is no established follow-up and treatment protocol for these patients, and these methods are neither specific nor sensitive for identifying early cardiac dysfunction. All things considered, the need for ongoing research in the field of pediatric cardiooncology is crucial to offer these patients a chance at a good quality of life as adults.
Collapse
|
18
|
Minotti G, Menna P, Camilli M, Salvatorelli E, Reggiardo G. Predictors of Early or Delayed Diastolic Dysfunction After Anthracycline-Based or Nonanthracycline Chemotherapy: A Pharmacological Appraisal. J Pharmacol Exp Ther 2020; 376:231-239. [DOI: 10.1124/jpet.120.000323] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/04/2020] [Indexed: 12/22/2022] Open
|
19
|
Lothstein L, Soberman J, Parke D, Gandhi J, Sweatman T, Seagroves T. Pivarubicin Is More Effective Than Doxorubicin Against Triple-Negative Breast Cancer In Vivo. Oncol Res 2020; 28:451-465. [PMID: 32430093 PMCID: PMC7751225 DOI: 10.3727/096504020x15898794315356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is unresponsive to antiestrogen and anti-HER2 therapies, requiring the use of cytotoxic drug combinations of anthracyclines, taxanes, cyclophosphamide, and platinum compounds. Multidrug therapies achieve pathological cure rates of only 20–40%, a consequence of drug resistance and cumulative dose limitations necessitated by the reversible cardiotoxic effects of drug therapy. Safer and more effective treatments for TNBC are required to achieve durable therapeutic responses. This study describes the mechanistic analyses of the novel anthracycline, pivarubicin, and its in vivo efficacy against human primary TNBC. Pivarubicin directly activates PKCd, triggers rapid mitochondrial-dependent apoptosis, and circumvents resistance conferred by overexpression of P-glycoprotein, Bcl-2, Bcl-XL, and Bcr-Abl. As a consequence, pivarubicin is more cytotoxic than doxorubicin against MDA-MB-231, and SUM159 TNBC cell lines grown in both monolayer culture and tumorspheres. Comparative in vivo efficacy of pivarubicin and doxorubicin was performed in an orthotopic NSG mouse model implanted with MDA-MB-231 human TNBC cells and treated with the maximum tolerated doses (MTDs) of pivarubicin and doxorubicin. Tumor growth was monitored by digital caliper measurements and determination of endpoint tumor weight and volume. Endpoint cardiotoxicity was assessed histologically by identifying microvacuolization in ventricular cardiomyocytes. Primary tumors treated with multiple rounds of doxorubicin at MTD failed to inhibit tumor growth compared with vehicle-treated tumors. However, administration of a single MTD of pivarubicin produced significant inhibition of tumor growth and tumor regression relative to tumor volume prior to initiation of treatment. Histological analysis of hearts excised from drug- and vehicle-treated mice revealed that pivarubicin produced no evidence of myocardial damage at a therapeutic dose. These results support the development of pivarubicin as a safer and more effective replacement for doxorubicin against TNBC as well as other malignancies for which doxorubicin therapy is indicated.
Collapse
Affiliation(s)
- Leonard Lothstein
- Department of Pathology and Laboratory Medicine, The University of Tennessee Health Science CenterMemphis, TNUSA
| | - Judith Soberman
- Department of Medicine, The University of Tennessee Health Science CenterMemphis, TNUSA
| | - Deanna Parke
- Department of Pathology and Laboratory Medicine, The University of Tennessee Health Science CenterMemphis, TNUSA
| | - Jatin Gandhi
- Department of Pathology and Laboratory Medicine, The University of Tennessee Health Science CenterMemphis, TNUSA
| | - Trevor Sweatman
- Department of Pharmacology, The University of Tennessee Health Science CenterMemphis, TNUSA
| | - Tiffany Seagroves
- Department of Pathology and Laboratory Medicine, The University of Tennessee Health Science CenterMemphis, TNUSA
| |
Collapse
|
20
|
Bai T, Zhu B, Shao D, Lian Z, Liu P, Shi J, Kong J. Blocking ACAT-1 Activity for Tumor Therapy with Fluorescent Hyperstar Polymer-Encapsulated Avasimible. Macromol Biosci 2020; 20:e1900438. [PMID: 32406183 DOI: 10.1002/mabi.201900438] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 04/02/2020] [Indexed: 12/14/2022]
Abstract
Targeting the distinct cholesterol metabolism of tumor cells is proposed as a novel way to treat tumors. Blocking acyl-CoA cholesterol acyltransferase-1 (ACAT-1) by the inhibitor avasimible (Ava), which elevates intracellular free cholesterol levels, is shown to effectively induce apoptosis. However, Ava faces disadvantages of poor water solubility, a short half-life, and no capability for fluorescence detection, which have greatly limited its application. Herein, a fluorescent hyperstar polymer (FHSP) is developed to encapsulate Ava to improve its ability to inhibit HeLa cells and K562 cells. The results of this study show that the obtained Ava-FHSP micelles possess a high drug loading capacity of 22.7% and bright green fluorescence. Ava and Ava-FHSP are cytotoxic to both HeLa and K562 cells and cause reductions in cell size, nuclear lysis, and chromatin condensation and hindered proliferation of both cell types by causing S phase cell cycle arrest. Further mechanistic analysis indicates that Ava-FHSP reduces the protein and messenger RNA expression of ACAT-1 and significantly increases intracellular free cholesterol levels, which can increase endoplasmic reticulum stress and finally cause cell apoptosis. All these results suggest that this fluorescent hyperstar polymer represents a potential therapeutic tumor strategy by changing the cholesterol metabolism of tumor cells.
Collapse
Affiliation(s)
- Ting Bai
- Shaanxi Key Laboratory of Macromolecular Science and Technology, School of Science, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Bobo Zhu
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Dongyan Shao
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Ziyang Lian
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Pei Liu
- Shaanxi Key Laboratory of Macromolecular Science and Technology, School of Science, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Junling Shi
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Jie Kong
- Shaanxi Key Laboratory of Macromolecular Science and Technology, School of Science, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| |
Collapse
|
21
|
Improved melanoma suppression with target-delivered TRAIL and Paclitaxel by a multifunctional nanocarrier. J Control Release 2020; 325:10-24. [PMID: 32251770 DOI: 10.1016/j.jconrel.2020.03.049] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/21/2020] [Accepted: 03/31/2020] [Indexed: 12/27/2022]
Abstract
Malignant melanoma, a highly dangerous type of skin cancer, is usually resistant to pro-apoptosis agents such as tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) due to low death receptor expression levels. After verifying combination of chemotherapy drug paclitaxel (PTX) and TRAIL could significantly enhance their anti-melanoma effects, we developed a liposomal melanoma target-delivery system with tumor microenvironment responsiveness (TRAIL-[Lip-PTX]C18-TR) to co-deliver TRAIL and PTX. TRAIL is attached to negatively-charged liposome surface while PTX is encapsulated inside, with final surface modification of a stearyl chain (C18) fused pH-sensitive cell-penetrating peptide (TR). Here, C18-TR could specifically binds to melanoma-rich integrin receptors αvβ3 for melanoma targeting, help release TRAIL in low pH microenvironment by reversing the liposomal charge, and facilitate consequent liposome internalization. TRAIL-[Lip-PTX]C18-TR displayed significantly better in vitro half-maximal inhibitory concentration (IC50) than other formulations, and an in vivo tumor inhibition rate of 93.8%. Mechanistic study revealed that this synergistic effect is associated with the upregulation of death receptors DR4/5 by PTX. This co-delivery system significantly improved TRAIL-based therapy against melanoma, and provided a simple platform to co-deliver other drugs/agents for melanoma treatment.
Collapse
|
22
|
Effects of grapeseed extract on doxorubicin-induced cardiotoxicity in rats. Herz 2020; 46:103-108. [PMID: 31970462 DOI: 10.1007/s00059-019-04888-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 09/23/2019] [Accepted: 12/12/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Doxorubicin (DX) is used for the treatment of many types of cancer; however, a side effect of this agent is cardiotoxicity, which may lead to cardiomyopathy or cardiac failure. Oxidative stress is thought to play a major role in the development of cardiotoxic effects. Proanthocyanidins found in grapeseed (GS) extract may inhibit chemically induced lipid peroxidation and apoptosis caused by oxidative stress. We aimed to investigate the cardioprotective effects of GS extract against DX-induced cardiotoxicity. METHODS A total of 28 male Sprague Dawley rats were grouped to receive: (a) standard nutrition (n = 7); (b) standard nutrition with an additional dose of 10 mg/kg DX (n = 7); (c) standard nutrition plus 100 mg/kg/day of GS (n = 7); (d) standard nutrition with 100 mg/kg/day of GS plus a single dose of 10 mg/kg DX. After 35 days the rats were decapitated and blood samples were taken for biochemical testing. Cardiac tissue samples were prepared for microscopy and histopathological evaluation. RESULTS Rats in the DX group exhibited significant elevations in biomarkers such as troponin and NT-proBNP as well as in oxidative stress markers compared with all other groups. Histopathological examination corroborated these findings by demonstrating significant and severe structural injury in the cardiac tissue of DX rates. Moreover, rats in the DX + GS group had significantly lower cardiac injury than rats in the DX group according to both biochemical (troponin and NT-proBNP) and histopathological analyses. Serum malondialdehyde levels (a marker of oxidative stress) in the DX + GS rats were significantly lower than in the DX rats. CONCLUSION Our findings suggest that GS may reduce the severity of DX-induced cardiotoxicity and thus has the potential to prevent cardiac injury in this setting.
Collapse
|
23
|
Facile preparation of pH-responsive PEGylated prodrugs for activated intracellular drug delivery. CHINESE CHEM LETT 2019. [DOI: 10.1016/j.cclet.2019.04.052] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
24
|
Broughton KM. Prevalence of comorbidities in heart failure patients and those treated with cellular therapeutics. Expert Rev Cardiovasc Ther 2019; 17:597-604. [PMID: 31382789 DOI: 10.1080/14779072.2019.1653185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: Changes in our daily living, particularly in work routines, diet, and physical exercise, have influenced a worldwide crisis for life-threatening comorbidities and the likelihood of cardiovascular disease diagnosis. Cardiovascular regenerative medicine researchers continue to investigate new therapeutic approaches and reexamine completed clinical trials to design more effective future studies. As the frequency of cardiovascular disease diagnosis continues to rise, investigations of how to repair and regenerate the failing myocardium remains an essential mission for human health. Areas covered: This review first examines the aging process, the rising rate of comorbidities, and the likelihood of developing heart disease. In the ongoing efforts to recapitulate human health needs in clinical trials, a review of clinical trials involving cellular therapeutics for cardiac repair, with a focus on the patient population and patients' complex medical records, is presented. Expert opinion: The expert opinion first draws attention to the changing demographics of patients diagnosed with diseases that lead to heart failure and focusing on obesity as a primary driver for increased cardiovascular disease. The opinion focuses on the importance of designing preclinical models and experimentation that better mimic the patient population and clinical situations to evaluate the effectiveness of potential future therapeutic interventions.
Collapse
Affiliation(s)
- Kathleen M Broughton
- SDSU Heart Institute and Department of Biology, San Diego State University , San Diego , CA , USA
| |
Collapse
|
25
|
Pannunzio NR, Lieber MR. Constitutively active Artemis nuclease recognizes structures containing single-stranded DNA configurations. DNA Repair (Amst) 2019; 83:102676. [PMID: 31377101 DOI: 10.1016/j.dnarep.2019.102676] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 07/24/2019] [Accepted: 07/25/2019] [Indexed: 01/03/2023]
Abstract
The Artemis nuclease recognizes and endonucleolytically cleaves at single-stranded to double-stranded DNA (ss/dsDNA) boundaries. It is also a key enzyme in the non-homologous end joining (NHEJ) DNA double-strand break repair pathway. Previously, a truncated form, Artemis-413, was developed that is constitutively active both in vitro and in vivo. Here, we use this constitutively active form of Artemis to detect DNA structures with ss/dsDNA boundaries that arise under topological stress. Topoisomerases prevent abnormal levels of torsional stress through modulation of positive and negative supercoiling. We show that overexpression of Artemis-413 in yeast cells carrying genetic mutations that ablate topoisomerase activity have an increased frequency of DNA double-strand breaks (DSBs). Based on the biochemical activity of Artemis, this suggests an increase in ss/dsDNA-containing structures upon increased torsional stress, with DSBs arising due to Artemis cutting at these ss/dsDNA structures. Camptothecin targets topoisomerase IB (Top1), and cells treated with camptothecin show increased DSBs. We find that expression of Artemis-413 in camptothecin-treated cells leads to a reduction in DSBs, the opposite of what we find with topoisomerase genetic mutations. This contrast between outcomes not only confirms that topoisomerase mutation and topoisomerase poisoning have distinct effects on cells, but also demonstrates the usefulness of Artemis-413 to study changes in DNA structure.
Collapse
Affiliation(s)
- Nicholas R Pannunzio
- Department of Pathology, Keck School of Medicine of University of Southern California, Los Angeles, CA, 90089, USA; Norris Comprehensive Cancer Center, Keck School of Medicine of University of Southern California, Los Angeles, CA, 90089, USA.
| | - Michael R Lieber
- Department of Pathology, Keck School of Medicine of University of Southern California, Los Angeles, CA, 90089, USA; Norris Comprehensive Cancer Center, Keck School of Medicine of University of Southern California, Los Angeles, CA, 90089, USA; Department of Biological Sciences, Molecular and Computational Biology Section, University of Southern California, Los Angeles, CA, 90089, USA.
| |
Collapse
|
26
|
Fetah K, Tebon P, Goudie MJ, Eichenbaum J, Ren L, Barros N, Nasiri R, Ahadian S, Ashammakhi N, Dokmeci MR, Khademhosseini A. The emergence of 3D bioprinting in organ-on-chip systems. ACTA ACUST UNITED AC 2019. [DOI: 10.1088/2516-1091/ab23df] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
27
|
Tocchetti CG, Cadeddu C, Di Lisi D, Femminò S, Madonna R, Mele D, Monte I, Novo G, Penna C, Pepe A, Spallarossa P, Varricchi G, Zito C, Pagliaro P, Mercuro G. From Molecular Mechanisms to Clinical Management of Antineoplastic Drug-Induced Cardiovascular Toxicity: A Translational Overview. Antioxid Redox Signal 2019; 30:2110-2153. [PMID: 28398124 PMCID: PMC6529857 DOI: 10.1089/ars.2016.6930] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Significance: Antineoplastic therapies have significantly improved the prognosis of oncology patients. However, these treatments can bring to a higher incidence of side-effects, including the worrying cardiovascular toxicity (CTX). Recent Advances: Substantial evidence indicates multiple mechanisms of CTX, with redox mechanisms playing a key role. Recent data singled out mitochondria as key targets for antineoplastic drug-induced CTX; understanding the underlying mechanisms is, therefore, crucial for effective cardioprotection, without compromising the efficacy of anti-cancer treatments. Critical Issues: CTX can occur within a few days or many years after treatment. Type I CTX is associated with irreversible cardiac cell injury, and it is typically caused by anthracyclines and traditional chemotherapeutics. Type II CTX is generally caused by novel biologics and more targeted drugs, and it is associated with reversible myocardial dysfunction. Therefore, patients undergoing anti-cancer treatments should be closely monitored, and patients at risk of CTX should be identified before beginning treatment to reduce CTX-related morbidity. Future Directions: Genetic profiling of clinical risk factors and an integrated approach using molecular, imaging, and clinical data may allow the recognition of patients who are at a high risk of developing chemotherapy-related CTX, and it may suggest methodologies to limit damage in a wider range of patients. The involvement of redox mechanisms in cancer biology and anticancer treatments is a very active field of research. Further investigations will be necessary to uncover the hallmarks of cancer from a redox perspective and to develop more efficacious antineoplastic therapies that also spare the cardiovascular system.
Collapse
Affiliation(s)
| | - Christian Cadeddu
- 2 Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Daniela Di Lisi
- 3 Biomedical Department of Internal Medicine, University of Palermo, Palermo, Italy
| | - Saveria Femminò
- 4 Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Rosalinda Madonna
- 5 Center of Aging Sciences and Translational Medicine - CESI-MeT, "G. d'Annunzio" University, Chieti, Italy.,6 Department of Internal Medicine, The Texas Heart Institute and Center for Cardiovascular Biology and Atherosclerosis Research, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Donato Mele
- 7 Cardiology Unit, Emergency Department, University Hospital of Ferrara, Ferrara, Italy
| | - Ines Monte
- 8 Department of General Surgery and Medical-Surgery Specialities, University of Catania, Catania, Italy
| | - Giuseppina Novo
- 3 Biomedical Department of Internal Medicine, University of Palermo, Palermo, Italy
| | - Claudia Penna
- 4 Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Alessia Pepe
- 9 U.O.C. Magnetic Resonance Imaging, Fondazione Toscana G. Monasterio C.N.R., Pisa, Italy
| | - Paolo Spallarossa
- 10 Clinic of Cardiovascular Diseases, IRCCS San Martino IST, Genova, Italy
| | - Gilda Varricchi
- 1 Department of Translational Medical Sciences, Federico II University, Naples, Italy.,11 Center for Basic and Clinical Immunology Research (CISI) - Federico II University, Naples, Italy
| | - Concetta Zito
- 12 Division of Cardiology, Clinical and Experimental Department of Medicine and Pharmacology, Policlinico "G. Martino" University of Messina, Messina, Italy
| | - Pasquale Pagliaro
- 4 Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Giuseppe Mercuro
- 2 Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| |
Collapse
|
28
|
Minotti G, Menna P, Calabrese V, Greco C, Armento G, Annibali O, Marchesi F, Salvatorelli E, Reggiardo G. Pharmacology of Ranolazine versus Common Cardiovascular Drugs in Patients with Early Diastolic Dysfunction Induced by Anthracyclines or Nonanthracycline Chemotherapeutics: A Phase 2b Minitrial. J Pharmacol Exp Ther 2019; 370:197-205. [PMID: 31101682 DOI: 10.1124/jpet.119.258178] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 05/15/2019] [Indexed: 12/12/2022] Open
Abstract
We have reported that anthracyclines and nonanthracycline chemotherapeutics caused diastolic dysfunction in cancer patients without cardiovascular risk factors. Diastolic dysfunction occurred as early as 1 week after the last chemotherapy cycle and manifested as impaired myocardial relaxation at echocardiography or persistent elevations of B-type natriuretic peptide (BNP) or troponin. The antianginal drug ranolazine shows cardiac relaxant effects that we considered of value to treat early diastolic dysfunction induced by cancer drugs; therefore, 24 low-risk patients with post-chemotherapy diastolic dysfunction were randomized (1:1) to ranolazine or the investigator's choice of common cardiovascular drugs, such as β-blockers and/or angiotensin-converting enzyme inhibitors or loop diuretics (best standard therapy, BST). After 5 weeks, 12 of 12 patients on ranolazine recovered from diastolic dysfunction, whereas 3 of 12 patients on BST did not improve; however, adverse events (not serious) were apparently more frequent for ranolazine than for BST (4/12 vs. 1/12). Ranolazine did not lower blood pressure, whereas BST reduced systolic pressure and caused a trend toward a reduced diastolic pressure. Most patients at randomization showed tachycardia resulting from chemotherapy-related anemia. Hemoglobin recovery contributed to normalizing heart rate in these patients; however, some patients in the ranolazine arm developed tachycardia through chronotropic effects of high BNP levels and returned to a normal heart rate through the effects of ranolazine on decreasing BNP levels. This minitrial describes the potential effects of ranolazine on relieving chemotherapy-related diastolic dysfunction; however, clinical implications of these findings need to be characterized by studies with an adequate sample size. SIGNIFICANCE STATEMENT: The antianginal drug ranolazine causes cardiac relaxant effects that might relieve diastolic dysfunction. In a clinical pharmacology study, 24 patients were randomized (1:1) to receive ranolazine or common cardiovascular drugs to treat early diastolic dysfunction induced by anthracycline-based or nonanthracycline chemotherapy. Ranolazine relieved diastolic dysfunction in these patients. The safety profile of ranolazine in cancer patients is similar to that of the general population. Compared with common cardiovascular drugs, ranolazine relieved diastolic dysfunction without lowering blood pressure. The sample size of this study was nonetheless too small to permit considerations about the potential clinical value of ranolazine for oncologic patients with early diastolic dysfunction induced by anthracyclines or nonanthracycline chemotherapeutics. This information should be obtained by studies with an adequate sample size.
Collapse
Affiliation(s)
- Giorgio Minotti
- Clinical Pharmacology Unit (G.M., P.M.) and Cardio Center (V.C.), Campus Bio-Medico University Hospital, Rome; Units of Drug Sciences (G.M., E.S.), Radiation Oncology (C.G.), Oncology (G.A.), and Hematology (O.A.), Department of Medicine and Center for Integrated Research, University Campus Bio-Medico, Rome; Hematology and Stem Cell Transplant Unit, IRCCS Regina Elena National Cancer Institute, Rome (F.M.); and Mediservice S.r.l., Agrate Brianza (Monza) (G.R.), Italy
| | - Pierantonio Menna
- Clinical Pharmacology Unit (G.M., P.M.) and Cardio Center (V.C.), Campus Bio-Medico University Hospital, Rome; Units of Drug Sciences (G.M., E.S.), Radiation Oncology (C.G.), Oncology (G.A.), and Hematology (O.A.), Department of Medicine and Center for Integrated Research, University Campus Bio-Medico, Rome; Hematology and Stem Cell Transplant Unit, IRCCS Regina Elena National Cancer Institute, Rome (F.M.); and Mediservice S.r.l., Agrate Brianza (Monza) (G.R.), Italy
| | - Vito Calabrese
- Clinical Pharmacology Unit (G.M., P.M.) and Cardio Center (V.C.), Campus Bio-Medico University Hospital, Rome; Units of Drug Sciences (G.M., E.S.), Radiation Oncology (C.G.), Oncology (G.A.), and Hematology (O.A.), Department of Medicine and Center for Integrated Research, University Campus Bio-Medico, Rome; Hematology and Stem Cell Transplant Unit, IRCCS Regina Elena National Cancer Institute, Rome (F.M.); and Mediservice S.r.l., Agrate Brianza (Monza) (G.R.), Italy
| | - Carlo Greco
- Clinical Pharmacology Unit (G.M., P.M.) and Cardio Center (V.C.), Campus Bio-Medico University Hospital, Rome; Units of Drug Sciences (G.M., E.S.), Radiation Oncology (C.G.), Oncology (G.A.), and Hematology (O.A.), Department of Medicine and Center for Integrated Research, University Campus Bio-Medico, Rome; Hematology and Stem Cell Transplant Unit, IRCCS Regina Elena National Cancer Institute, Rome (F.M.); and Mediservice S.r.l., Agrate Brianza (Monza) (G.R.), Italy
| | - Grazia Armento
- Clinical Pharmacology Unit (G.M., P.M.) and Cardio Center (V.C.), Campus Bio-Medico University Hospital, Rome; Units of Drug Sciences (G.M., E.S.), Radiation Oncology (C.G.), Oncology (G.A.), and Hematology (O.A.), Department of Medicine and Center for Integrated Research, University Campus Bio-Medico, Rome; Hematology and Stem Cell Transplant Unit, IRCCS Regina Elena National Cancer Institute, Rome (F.M.); and Mediservice S.r.l., Agrate Brianza (Monza) (G.R.), Italy
| | - Ombretta Annibali
- Clinical Pharmacology Unit (G.M., P.M.) and Cardio Center (V.C.), Campus Bio-Medico University Hospital, Rome; Units of Drug Sciences (G.M., E.S.), Radiation Oncology (C.G.), Oncology (G.A.), and Hematology (O.A.), Department of Medicine and Center for Integrated Research, University Campus Bio-Medico, Rome; Hematology and Stem Cell Transplant Unit, IRCCS Regina Elena National Cancer Institute, Rome (F.M.); and Mediservice S.r.l., Agrate Brianza (Monza) (G.R.), Italy
| | - Francesco Marchesi
- Clinical Pharmacology Unit (G.M., P.M.) and Cardio Center (V.C.), Campus Bio-Medico University Hospital, Rome; Units of Drug Sciences (G.M., E.S.), Radiation Oncology (C.G.), Oncology (G.A.), and Hematology (O.A.), Department of Medicine and Center for Integrated Research, University Campus Bio-Medico, Rome; Hematology and Stem Cell Transplant Unit, IRCCS Regina Elena National Cancer Institute, Rome (F.M.); and Mediservice S.r.l., Agrate Brianza (Monza) (G.R.), Italy
| | - Emanuela Salvatorelli
- Clinical Pharmacology Unit (G.M., P.M.) and Cardio Center (V.C.), Campus Bio-Medico University Hospital, Rome; Units of Drug Sciences (G.M., E.S.), Radiation Oncology (C.G.), Oncology (G.A.), and Hematology (O.A.), Department of Medicine and Center for Integrated Research, University Campus Bio-Medico, Rome; Hematology and Stem Cell Transplant Unit, IRCCS Regina Elena National Cancer Institute, Rome (F.M.); and Mediservice S.r.l., Agrate Brianza (Monza) (G.R.), Italy
| | - Giorgio Reggiardo
- Clinical Pharmacology Unit (G.M., P.M.) and Cardio Center (V.C.), Campus Bio-Medico University Hospital, Rome; Units of Drug Sciences (G.M., E.S.), Radiation Oncology (C.G.), Oncology (G.A.), and Hematology (O.A.), Department of Medicine and Center for Integrated Research, University Campus Bio-Medico, Rome; Hematology and Stem Cell Transplant Unit, IRCCS Regina Elena National Cancer Institute, Rome (F.M.); and Mediservice S.r.l., Agrate Brianza (Monza) (G.R.), Italy
| |
Collapse
|
29
|
Chronic effects of platinum(IV) complex and its diamine ligand on rat heart function: comparison with cisplatin. Mol Cell Biochem 2019; 458:89-98. [DOI: 10.1007/s11010-019-03533-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 04/10/2019] [Indexed: 12/15/2022]
|
30
|
Cardiotoxicity of Targeted Cancer Drugs: Concerns, "The Cart Before the Horse," and Lessons from Trastuzumab. Curr Cardiol Rep 2019; 21:33. [PMID: 30887161 DOI: 10.1007/s11886-019-1121-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
PURPOSE OF REVIEW Modern oncology is witnessing a renaissance of its pharmacologic armamentarium. Old generation drugs, such as anthracyclines and other cytotoxic or cytostatic drugs, were plagued with a lack of specificity and with the possible occurrence of untoward effects in the cardiovascular system and other healthy tissues. The old drugs are now combined with, or replaced by, new agents that are more specific in attacking some unique moieties and vital functions of cancer cells, causing less noxious effects in healthy tissues. Regrettably, however, the new "targeted" drugs still cause varying levels of cardiac or vascular toxicity. Here, we describe the case of trastuzumab, a monoclonal antibody that dramatically improved the life expectancy of women with Erbb2-overexpressing breast tumor, while also raising concerns about a possible incidence of cardiac dysfunction. RECENT FINDINGS The scientific community counts experts that label trastuzumab as a "cardiotoxic agent" and other experts that maintain a more benign assessment. We describe the biologic foundations and clinical evidence for such controversy. We show that trastuzumab cardiotoxicity is probably overrated, leading some experts to raise unjustified overconcerns about the cardiotoxicity of trastuzumab as a single agent or in combination with anthracyclines or other old and new drugs. We analyze the biases that caused trastuzumab cardiotoxicity to be overrated. Trastuzumab is a life-saving agent showing a moderate and clinically manageable cardiac dysfunction, and yet, it is portrayed as cardiotoxic. We take the trastuzumab lesson to reaffirm that cardio-oncologists should provide cancer patients with the best therapeutic opportunity, as is the case for trastuzumab, while also devising the necessary strategies of risk assessment and mitigation.
Collapse
|
31
|
Talib WH, Al-Hadid SA, Ali MBW, Al-Yasari IH, Ali MRA. Role of curcumin in regulating p53 in breast cancer: an overview of the mechanism of action. BREAST CANCER (DOVE MEDICAL PRESS) 2018; 10:207-217. [PMID: 30568488 PMCID: PMC6276637 DOI: 10.2147/bctt.s167812] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
p53 is a tumor suppressor gene involved in various cellular mechanisms including DNA repair, apoptosis, and cell cycle arrest. More than 50% of human cancers have a mutated nonfunctional p53. Breast cancer (BC) is one of the main causes of cancer-related deaths among females. p53 mutations in BC are associated with low survival rates and more resistance to the conventional therapies. Thus, targeting p53 activity was suggested as an important strategy in cancer therapy. During the past decades, cancer research was focused on the development of monotargeted anticancer therapies. However, the development of drug resistance by modulation of genes, proteins, and pathways was the main hindrance to the success of such therapies. Curcumin is a natural product, extracted from the roots of Curcuma longa, and possesses various biological effects including anticancer activity. Previous studies proved the ability of curcumin to modulate several signaling pathways and biomolecules in cancer. Safety and cost-effectiveness are additional inevitable advantages of curcumin. This review summarizes the effects of curcumin as a regulator of p53 in BC and the key molecular mechanisms of this regulation.
Collapse
Affiliation(s)
- Wamidh H Talib
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman, Jordan,
| | - Sonia A Al-Hadid
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman, Jordan,
| | - Mai B Wild Ali
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman, Jordan,
| | - Intisar Hadi Al-Yasari
- Food Technology Department, Faculty of Food Science, AL-Qasim Green University, Babylon, Iraq
| | | |
Collapse
|
32
|
Corremans R, Adão R, De Keulenaer GW, Leite-Moreira AF, Brás-Silva C. Update on pathophysiology and preventive strategies of anthracycline-induced cardiotoxicity. Clin Exp Pharmacol Physiol 2018; 46:204-215. [DOI: 10.1111/1440-1681.13036] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/31/2018] [Accepted: 09/13/2018] [Indexed: 12/13/2022]
Affiliation(s)
| | - Rui Adão
- Department of Surgery and Physiology; UnIC-Cardiovascular Research Centre; Faculty of Medicine; University of Porto; Porto Portugal
| | | | - Adelino F. Leite-Moreira
- Department of Surgery and Physiology; UnIC-Cardiovascular Research Centre; Faculty of Medicine; University of Porto; Porto Portugal
| | - Carmen Brás-Silva
- Department of Surgery and Physiology; UnIC-Cardiovascular Research Centre; Faculty of Medicine; University of Porto; Porto Portugal
- Faculty of Nutrition and Food Sciences; University of Porto; Porto Portugal
| |
Collapse
|
33
|
Hevener K, Verstak TA, Lutat KE, Riggsbee DL, Mooney JW. Recent developments in topoisomerase-targeted cancer chemotherapy. Acta Pharm Sin B 2018; 8:844-861. [PMID: 30505655 PMCID: PMC6251812 DOI: 10.1016/j.apsb.2018.07.008] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 07/18/2018] [Accepted: 07/18/2018] [Indexed: 12/17/2022] Open
Abstract
The DNA topoisomerase enzymes are essential to cell function and are found ubiquitously in all domains of life. The various topoisomerase enzymes perform a wide range of functions related to the maintenance of DNA topology during DNA replication, and transcription are the targets of a wide range of antimicrobial and cancer chemotherapeutic agents. Natural product-derived agents, such as the camptothecin, anthracycline, and podophyllotoxin drugs, have seen broad use in the treatment of many types of cancer. Selective targeting of the topoisomerase enzymes for cancer treatment continues to be a highly active area of basic and clinical research. The focus of this review will be to summarize the current state of the art with respect to clinically used topoisomerase inhibitors for targeted cancer treatment and to discuss the pharmacology and chemistry of promising new topoisomerase inhibitors in clinical and pre-clinical development.
Collapse
Affiliation(s)
- KirkE. Hevener
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | | | | | | | | |
Collapse
|
34
|
Menna P, Salvatorelli E, Armento G, Annibali O, Greco C, Marchesi F, Calabrese V, Reggiardo G, Minotti G. The Endogenous Lusitropic and Chronotropic Agent, B-Type Natriuretic Peptide, Limits Cardiac Troponin Release in Cancer Patients with an Early Impairment of Myocardial Relaxation Induced by Anthracyclines. J Pharmacol Exp Ther 2018; 367:518-527. [PMID: 30275150 DOI: 10.1124/jpet.118.253104] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 09/27/2018] [Indexed: 11/22/2022] Open
Abstract
We have reported that cancer patients treated with anthracycline-based or nonanthracycline chemotherapy developed an early impairment of myocardial relaxation at echocardiography or persistent elevations of the cardiac hormone B-type natriuretic peptide (BNP). Post-hoc pharmacologic analyses showed that BNP elevations were induced by impaired relaxation and caused positive lusitropic effects that maintained normal relaxation. High BNP levels and impaired relaxation were therefore characterized as mutually exclusive manifestations of diastolic dysfunction, but high BNP levels resulted in positive chronotropism and inappropriate tachycardia. Some patients developed increased circulating levels of cardiac troponin I isoform (cTnI), a marker of cardiomyocyte necrosis. Here we have characterized whether cTnI elevations correlated with diastolic dysfunction that manifested as impaired relaxation or a high level of BNP. The effects of high BNP levels on cTnI elevations were also characterized. We show that impaired relaxation or high BNP levels were significantly more frequent in patients with cTnI elevations. High BNP levels diminished the plasma peak and area under the curve of cTnI, but this result was accompanied by inappropriate tachycardia. cTnI elevations occurred only in patients treated with anthracyclines; moreover, the association of impaired relaxation or high BNP levels with cTnI elevations was significantly more frequent in doxorubicin-treated patients compared with patients treated with its analog, epirubicin. These findings describe cause-and-effect relations between impaired relaxation and cardiomyocyte necrosis, illuminate the role of anthracycline analogs, denote that the beneficial effects of BNP in relieving impaired relaxation and cardiomyocyte necrosis are counterbalanced by inappropriate tachycardia. Patients showing troponin elevations and impaired relaxation or high BNP levels should be treated with lusitropic drugs that lack a positive chronotropism.
Collapse
Affiliation(s)
- Pierantonio Menna
- Units of Drug Sciences (P.M., E.S., G.M.), Oncology (G.A.), Hematology (O.A.), Radiation Oncology (C.G.), and Cardiovascular Sciences (V.C.), Department of Medicine and Center for Integrated Research, University Campus Bio-Medico, Rome; Hematology and Stem Cell Transplant Unit, IRCCS Regina Elena National Cancer Institute, Rome (F.M.); and Mediservice S.r.l., Agrate Brianza (Monza), Italy (G.R.)
| | - Emanuela Salvatorelli
- Units of Drug Sciences (P.M., E.S., G.M.), Oncology (G.A.), Hematology (O.A.), Radiation Oncology (C.G.), and Cardiovascular Sciences (V.C.), Department of Medicine and Center for Integrated Research, University Campus Bio-Medico, Rome; Hematology and Stem Cell Transplant Unit, IRCCS Regina Elena National Cancer Institute, Rome (F.M.); and Mediservice S.r.l., Agrate Brianza (Monza), Italy (G.R.)
| | - Grazia Armento
- Units of Drug Sciences (P.M., E.S., G.M.), Oncology (G.A.), Hematology (O.A.), Radiation Oncology (C.G.), and Cardiovascular Sciences (V.C.), Department of Medicine and Center for Integrated Research, University Campus Bio-Medico, Rome; Hematology and Stem Cell Transplant Unit, IRCCS Regina Elena National Cancer Institute, Rome (F.M.); and Mediservice S.r.l., Agrate Brianza (Monza), Italy (G.R.)
| | - Ombretta Annibali
- Units of Drug Sciences (P.M., E.S., G.M.), Oncology (G.A.), Hematology (O.A.), Radiation Oncology (C.G.), and Cardiovascular Sciences (V.C.), Department of Medicine and Center for Integrated Research, University Campus Bio-Medico, Rome; Hematology and Stem Cell Transplant Unit, IRCCS Regina Elena National Cancer Institute, Rome (F.M.); and Mediservice S.r.l., Agrate Brianza (Monza), Italy (G.R.)
| | - Carlo Greco
- Units of Drug Sciences (P.M., E.S., G.M.), Oncology (G.A.), Hematology (O.A.), Radiation Oncology (C.G.), and Cardiovascular Sciences (V.C.), Department of Medicine and Center for Integrated Research, University Campus Bio-Medico, Rome; Hematology and Stem Cell Transplant Unit, IRCCS Regina Elena National Cancer Institute, Rome (F.M.); and Mediservice S.r.l., Agrate Brianza (Monza), Italy (G.R.)
| | - Francesco Marchesi
- Units of Drug Sciences (P.M., E.S., G.M.), Oncology (G.A.), Hematology (O.A.), Radiation Oncology (C.G.), and Cardiovascular Sciences (V.C.), Department of Medicine and Center for Integrated Research, University Campus Bio-Medico, Rome; Hematology and Stem Cell Transplant Unit, IRCCS Regina Elena National Cancer Institute, Rome (F.M.); and Mediservice S.r.l., Agrate Brianza (Monza), Italy (G.R.)
| | - Vito Calabrese
- Units of Drug Sciences (P.M., E.S., G.M.), Oncology (G.A.), Hematology (O.A.), Radiation Oncology (C.G.), and Cardiovascular Sciences (V.C.), Department of Medicine and Center for Integrated Research, University Campus Bio-Medico, Rome; Hematology and Stem Cell Transplant Unit, IRCCS Regina Elena National Cancer Institute, Rome (F.M.); and Mediservice S.r.l., Agrate Brianza (Monza), Italy (G.R.)
| | - Giorgio Reggiardo
- Units of Drug Sciences (P.M., E.S., G.M.), Oncology (G.A.), Hematology (O.A.), Radiation Oncology (C.G.), and Cardiovascular Sciences (V.C.), Department of Medicine and Center for Integrated Research, University Campus Bio-Medico, Rome; Hematology and Stem Cell Transplant Unit, IRCCS Regina Elena National Cancer Institute, Rome (F.M.); and Mediservice S.r.l., Agrate Brianza (Monza), Italy (G.R.)
| | - Giorgio Minotti
- Units of Drug Sciences (P.M., E.S., G.M.), Oncology (G.A.), Hematology (O.A.), Radiation Oncology (C.G.), and Cardiovascular Sciences (V.C.), Department of Medicine and Center for Integrated Research, University Campus Bio-Medico, Rome; Hematology and Stem Cell Transplant Unit, IRCCS Regina Elena National Cancer Institute, Rome (F.M.); and Mediservice S.r.l., Agrate Brianza (Monza), Italy (G.R.)
| |
Collapse
|
35
|
Goswami U, Kandimalla R, Kalita S, Chattopadhyay A, Ghosh SS. Polyethylene Glycol-Encapsulated Histone Deacetylase Inhibitor Drug-Composite Nanoparticles for Combination Therapy with Artesunate. ACS OMEGA 2018; 3:11504-11516. [PMID: 30320264 PMCID: PMC6173507 DOI: 10.1021/acsomega.8b02105] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 09/06/2018] [Indexed: 06/08/2023]
Abstract
Combination drug therapy has become an effective clinical practice for cancer treatment because of low cytotoxicity by the synergistic effect of each medicine. Luminescent Au nanoclusters (Au NCs) were formulated into spherical polyethylene glycol (PEG)-Au NC-encapsulated drug-sodium butyrate (NaB) composite nanoparticles (PEG-Au NC-NaB-NPs) in the presence of PEG and NaB. Their effect on cancer cells was investigated using bio imaging, unravelling the mechanism of the endocytosis pathway and combination therapeutic interventions with a plant-based antimalarial drug artesunate (ART). PEG-Au NC-NaB-NPs showed bright red luminescence in the lysosomal compartment of the cells upon uptake predominantly through a caveolae-mediated pathway. Combination of PEG-Au NC-NaB-NPs with ART displayed enhanced therapeutic activity at a reduced dose compared to its individual doses and revealed heightened synergistic activity as identified from the combination index. The mechanism of synergism revealed elevated generation of reactive oxygen species with both NaB and ART, which disrupts mitochondrial membrane potential as evident from JC-1 staining. Remarkably, the histone deacetylase (HDAC) assay and terminal deoxynucleotidyl transferase dUTP nick end labeling assay enlightened the role of NaB and ART in HDAC inhibition and DNA fragmentation, respectively. Thus, induction of apoptosis with the synergistic effect of both NaB and ART with its meticulous mechanism makes it a promising tool for combinational cancer therapy. In vivo activity of the NPs was evaluated on Daltons lymphoma ascites bearing mice, which exhibited significant reduction of tumor volume and viable tumor cells with a prolonged life span.
Collapse
Affiliation(s)
- Upashi Goswami
- Centre
for Nanotechnology, Department of Chemistry, and Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | - Raghuram Kandimalla
- Drug
Discovery Lab, Institute of Advanced Study
in Science and Technology, Guwahati 781035, Assam, India
| | - Sanjeeb Kalita
- Drug
Discovery Lab, Institute of Advanced Study
in Science and Technology, Guwahati 781035, Assam, India
| | - Arun Chattopadhyay
- Centre
for Nanotechnology, Department of Chemistry, and Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | - Siddhartha Sankar Ghosh
- Centre
for Nanotechnology, Department of Chemistry, and Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| |
Collapse
|
36
|
Antoniak S, Tatsumi K, Schmedes CM, Grover SP, Pawlinski R, Mackman N. Protease-activated receptor 1 activation enhances doxorubicin-induced cardiotoxicity. J Mol Cell Cardiol 2018; 122:80-87. [PMID: 30098988 DOI: 10.1016/j.yjmcc.2018.08.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 08/07/2018] [Accepted: 08/09/2018] [Indexed: 12/21/2022]
Abstract
OBJECTIVE The anti-cancer anthracycline drug Doxorubicin (Dox) causes cardiotoxicity. We investigated the role of protease-activated receptor 1 (PAR-1) in Dox-induced cardiotoxicity. METHODS AND RESULTS In vitro experiments revealed that PAR-1 enhanced Dox-induced mitochondrial dysfunction, reactive oxygen species and cell death of cardiac myocytes and cardiac fibroblasts. The contribution of PAR-1 to Dox-induced cardiotoxicity was investigated by subjecting PAR-1-/- mice and PAR-1+/+ mice to acute and chronic exposure to Dox. Heart function was measured by echocardiography. PAR-1-/- mice exhibited significant less cardiac injury and dysfunction compared to PAR-1+/+ mice after acute and chronic Dox administration. PAR-1-/- mice had reduced levels of nitrotyrosine, apoptosis and inflammation in their heart compared to PAR-1+/+ mice. Furthermore, inhibition of PAR-1 in wild-type mice with vorapaxar significantly reduced the acute Dox-induced cardiotoxicity. CONCLUSION Our results indicate that activation of PAR-1 contributes to Dox-induced cardiotoxicity. Inhibition of PAR-1 may be a new approach to reduce Dox-induced cardiotoxicity in cancer patients.
Collapse
Affiliation(s)
- Silvio Antoniak
- Department of Medicine, Thrombosis and Hemostasis Program, Division of Hematology and Oncology, UNC McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, United States; Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.
| | - Kohei Tatsumi
- Department of Medicine, Thrombosis and Hemostasis Program, Division of Hematology and Oncology, UNC McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, United States; Department of Physiology and Regenerative Medicine, Kindai University, Faculty of Medicine, Osaka-sayama, Osaka, Japan
| | - Clare M Schmedes
- Department of Medicine, Thrombosis and Hemostasis Program, Division of Hematology and Oncology, UNC McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, United States
| | - Steven P Grover
- Department of Medicine, Thrombosis and Hemostasis Program, Division of Hematology and Oncology, UNC McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, United States
| | - Rafal Pawlinski
- Department of Medicine, Thrombosis and Hemostasis Program, Division of Hematology and Oncology, UNC McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, United States
| | - Nigel Mackman
- Department of Medicine, Thrombosis and Hemostasis Program, Division of Hematology and Oncology, UNC McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, United States
| |
Collapse
|
37
|
Increased Plasma Nitrite and von Willebrand Factor Indicates Early Diagnosis of Vascular Diseases in Chemotherapy Treated Cancer Patients. Cardiovasc Toxicol 2018; 19:36-47. [DOI: 10.1007/s12012-018-9471-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
38
|
Menna P, Calabrese V, Armento G, Annibali O, Greco C, Salvatorelli E, Marchesi F, Reggiardo G, Minotti G. Pharmacology of Cardio-Oncology: Chronotropic and Lusitropic Effects of B-Type Natriuretic Peptide in Cancer Patients with Early Diastolic Dysfunction Induced by Anthracycline or Nonanthracycline Chemotherapy. J Pharmacol Exp Ther 2018; 366:158-168. [DOI: 10.1124/jpet.118.249235] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 04/30/2018] [Indexed: 12/31/2022] Open
|
39
|
Varricchi G, Ameri P, Cadeddu C, Ghigo A, Madonna R, Marone G, Mercurio V, Monte I, Novo G, Parrella P, Pirozzi F, Pecoraro A, Spallarossa P, Zito C, Mercuro G, Pagliaro P, Tocchetti CG. Antineoplastic Drug-Induced Cardiotoxicity: A Redox Perspective. Front Physiol 2018; 9:167. [PMID: 29563880 PMCID: PMC5846016 DOI: 10.3389/fphys.2018.00167] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 02/20/2018] [Indexed: 12/28/2022] Open
Abstract
Antineoplastic drugs can be associated with several side effects, including cardiovascular toxicity (CTX). Biochemical studies have identified multiple mechanisms of CTX. Chemoterapeutic agents can alter redox homeostasis by increasing the production of reactive oxygen species (ROS) and reactive nitrogen species RNS. Cellular sources of ROS/RNS are cardiomyocytes, endothelial cells, stromal and inflammatory cells in the heart. Mitochondria, peroxisomes and other subcellular components are central hubs that control redox homeostasis. Mitochondria are central targets for antineoplastic drug-induced CTX. Understanding the mechanisms of CTX is fundamental for effective cardioprotection, without compromising the efficacy of anticancer treatments. Type 1 CTX is associated with irreversible cardiac cell injury and is typically caused by anthracyclines and conventional chemotherapeutic agents. Type 2 CTX, associated with reversible myocardial dysfunction, is generally caused by biologicals and targeted drugs. Although oxidative/nitrosative reactions play a central role in CTX caused by different antineoplastic drugs, additional mechanisms involving directly and indirectly cardiomyocytes and inflammatory cells play a role in cardiovascular toxicities. Identification of cardiologic risk factors and an integrated approach using molecular, imaging, and clinical data may allow the selection of patients at risk of developing chemotherapy-related CTX. Although the last decade has witnessed intense research related to the molecular and biochemical mechanisms of CTX of antineoplastic drugs, experimental and clinical studies are urgently needed to balance safety and efficacy of novel cancer therapies.
Collapse
Affiliation(s)
- Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
- Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research, University of Naples Federico II, Naples, Italy
| | - Pietro Ameri
- Clinic of Cardiovascular Diseases, IRCCS San Martino IST, Genova, Italy
| | - Christian Cadeddu
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Rosalinda Madonna
- Institute of Cardiology, Center of Excellence on Aging, Università degli Studi “G. d'Annunzio” Chieti – Pescara, Chieti, Italy
- Department of Internal Medicine, Texas Heart Institute and Center for Cardiovascular Biology and Atherosclerosis Research, University of Texas Health Science Center, Houston, TX, United States
| | - Giancarlo Marone
- Section of Hygiene, Department of Public Health, University of Naples Federico II, Naples, Italy
- Monaldi Hospital Pharmacy, Naples, Italy
| | - Valentina Mercurio
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Ines Monte
- Department of General Surgery and Medical-Surgery Specialities, University of Catania, Catania, Italy
| | - Giuseppina Novo
- U.O.C. Magnetic Resonance Imaging, Fondazione Toscana G. Monasterio C.N.R., Pisa, Italy
| | - Paolo Parrella
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Flora Pirozzi
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Antonio Pecoraro
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Paolo Spallarossa
- Clinic of Cardiovascular Diseases, IRCCS San Martino IST, Genova, Italy
| | - Concetta Zito
- Division of Clinical and Experimental Cardiology, Department of Medicine and Pharmacology, Policlinico “G. Martino” University of Messina, Messina, Italy
| | - Giuseppe Mercuro
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Carlo G. Tocchetti
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| |
Collapse
|
40
|
Pecoraro M, Ciccarelli M, Fiordelisi A, Iaccarino G, Pinto A, Popolo A. Diazoxide Improves Mitochondrial Connexin 43 Expression in a Mouse Model of Doxorubicin-Induced Cardiotoxicity. Int J Mol Sci 2018. [PMID: 29518932 PMCID: PMC5877618 DOI: 10.3390/ijms19030757] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Doxorubicin (DOXO) administration induces alterations in Connexin 43 (Cx43) expression and localization, thus, inducing alterations in chemical and electrical signal transmission between cardiomyocytes and in intracellular calcium homeostasis even evident after a single administration. This study was designed to evaluate if Diazoxide (DZX), a specific opener of mitochondrial KATP channels widely used for its cardioprotective effects, can fight DOXO-induced cardiotoxicity in a short-time mouse model. DZX (20 mg/kg i.p.) was administered 30 min before DOXO (10 mg/kg i.p.) in C57BL/6j female mice for 1–3 or seven days once every other day. A recovery of cardiac parameters, evaluated by Echocardiography, were observed in DZX+DOXO co-treated mice. Western blot analysis performed on heart lysates showed an increase in sarco/endoplasmic reticulum Ca2+-ATPase (SERCAII) and a reduction in phospholamban (PLB) amounts in DZX+DOXO co-treated mice. A contemporary recovery of intracellular Ca2+-signal, detected spectrofluorometrically by means of FURA-2AM, was observed in these mice. Cx43 expression and localization, analyzed by Western blot and confirmed by immunofluorescence analysis, showed that DZX co-treatement increases Cx43 amount both on sarcoplasmic membrane and on mitochondria. In conclusion, our data demonstrate that, in a short-time mouse model of DOXO-induced cardiotoxicity, DZX exerts its cardioprotective effects also by enhancing the amount Cx43.
Collapse
Affiliation(s)
- Michela Pecoraro
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 84084 Fisciano, Italy.
| | - Michele Ciccarelli
- Department of Medicine and Surgery, University of Salerno, 84084 Baronissi, Italy.
| | - Antonella Fiordelisi
- Department of Advanced Biomedical Sciences, Federico II University, 80138 Naples, Italy.
| | - Guido Iaccarino
- Department of Medicine and Surgery, University of Salerno, 84084 Baronissi, Italy.
| | - Aldo Pinto
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 84084 Fisciano, Italy.
| | - Ada Popolo
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 84084 Fisciano, Italy.
| |
Collapse
|
41
|
Calabrese V, Menna P, Annibali O, Armento G, Carpino A, Cerchiara E, Greco C, Marchesi F, Spallarossa P, Toglia G, Reggiardo G, Minotti G. Early Diastolic Dysfunction after Cancer Chemotherapy: Primary Endpoint Results of a Multicenter Cardio-Oncology Study. Chemotherapy 2018; 63:55-63. [PMID: 29428939 DOI: 10.1159/000486761] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 01/11/2018] [Indexed: 01/14/2023]
Abstract
Asymptomatic diastolic dysfunction (DD) with preserved left ventricular ejection fraction (LVEF) is suspected to precede late cardiac events in cancer survivors treated by chemotherapy. We conducted the first multicenter study of early DD induced by chemotherapy. Patients who were candidates for standard dose chemotherapy were screened for the absence of cardiovascular risk factors, LVEF ≥50%, normal-for-age diastolic function at echocardiography (E/A ratio, E wave deceleration time; DT), normal levels of potential DD biomarkers like Nt-proBNP (≤125 pg/mL), and cardiac troponin I (cTnI, ≤0.05 ng/mL). Mitral Doppler (E/E') was left at the investigator's discretion. Chemotherapy-induced DD with preserved LVEF was diagnosed for patients showing LVEF ≥50% and any of the following: Nt-proBNP > 125 pg/mL, cTnI > 0.05 ng/mL, and out-of-range E/A and DT. Eighty patients (68 females, 12 males, median age 49 years) were evaluated at 1 week after chemotherapy (T1) [corrected]. Thirty-three protocol-defined diastolic events were observed (15 Nt-proBNP > 125 pg/mL, 14 grade I DD by E/A and DT, 4 cTnI > 0.05 ng/mL). The events occurred in 29 asymptomatic patients with LVEF ≥50% (36% incidence of DD with preserved LVEF). Interactions occurred between biomarkers and grade I DD. E/E' abnormalities were not observed. Both anthracycline-based and nonanthracycline regimens induced DD. These findings show that biomarkers and echocardiography intercept early DD in otherwise asymptomatic low-risk cancer patients treated by standard dose chemotherapy. These findings therefore call for the adequate cardiac management of cancer patients.
Collapse
Affiliation(s)
- Vito Calabrese
- Units of Cardiology, Hematology, Oncology, Radiation Therapy and Clinical Pharmacology, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Pierantonio Menna
- Units of Cardiology, Hematology, Oncology, Radiation Therapy and Clinical Pharmacology, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Ombretta Annibali
- Units of Cardiology, Hematology, Oncology, Radiation Therapy and Clinical Pharmacology, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Grazia Armento
- Units of Cardiology, Hematology, Oncology, Radiation Therapy and Clinical Pharmacology, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Armando Carpino
- Department of Cardiology, National Cancer Institute "Regina Elena", Rome, Italy
| | - Elisabetta Cerchiara
- Units of Cardiology, Hematology, Oncology, Radiation Therapy and Clinical Pharmacology, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Carlo Greco
- Units of Cardiology, Hematology, Oncology, Radiation Therapy and Clinical Pharmacology, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Francesco Marchesi
- Department of Cardiology, National Cancer Institute "Regina Elena", Rome, Italy
| | - Paolo Spallarossa
- Department of Cardiology, San Martino Hospital and IRCCS, Genoa, Italy
| | - Giuseppe Toglia
- Department of Cardiology, National Cancer Institute "Regina Elena", Rome, Italy
| | | | - Giorgio Minotti
- Units of Cardiology, Hematology, Oncology, Radiation Therapy and Clinical Pharmacology, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | | |
Collapse
|
42
|
Silva dos Santos D, Brasil GV, Ramos IPR, Mesquita FCP, Kasai-Brunswick TH, Christie MLA, Cahli GM, Barbosa RAQ, da Cunha ST, Pereira JX, Medei E, Campos de Carvalho AC, Carvalho AB, Goldenberg RCDS. Embryonic stem cell-derived cardiomyocytes for the treatment of doxorubicin-induced cardiomyopathy. Stem Cell Res Ther 2018; 9:30. [PMID: 29402309 PMCID: PMC5799903 DOI: 10.1186/s13287-018-0788-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 01/19/2018] [Accepted: 01/23/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Doxorubicin (Dox) is a chemotherapy drug with limited application due to cardiotoxicity that may progress to heart failure. This study aims to evaluate the role of cardiomyocytes derived from mouse embryonic stem cells (CM-mESCs) in the treatment of Dox-induced cardiomyopathy (DIC) in mice. METHODS The mouse embryonic stem cell (mESC) line E14TG2A was characterized by karyotype analysis, gene expression using RT-PCR and immunofluorescence. Cells were transduced with luciferase 2 and submitted to cardiac differentiation. Total conditioned medium (TCM) from the CM-mESCs was collected for proteomic analysis. To establish DIC in CD1 mice, Dox (7.5 mg/kg) was administered once a week for 3 weeks, resulting in a cumulative Dox dose of 22.5 mg/kg. At the fourth week, a group of animals was injected intramyocardially with CM-mESCs (8 × 105 cells). Cells were tracked by a bioluminescence assay, and the body weight, echocardiogram, electrocardiogram and number of apoptotic cardiomyocytes were evaluated. RESULTS mESCs exhibited a normal karyotype and expressed pluripotent markers. Proteomic analysis of TCM showed proteins related to the negative regulation of cell death. CM-mESCs presented ventricular action potential characteristics. Mice that received Dox developed heart failure and showed significant differences in body weight, ejection fraction (EF), end-systolic volume (ESV), stroke volume (SV), heart rate and QT and corrected QT (QTc) intervals when compared to the control group. After cell or placebo injection, the Dox + CM-mESC group showed significant increases in EF and SV when compared to the Dox + placebo group. Reduction in ESV and QT and QTc intervals in Dox + CM-mESC-treated mice was observed at 5 or 30 days after cell treatment. Cells were detected up to 11 days after injection. The Dox + CM-mESC group showed a significant reduction in the percentage of apoptotic cardiomyocytes in the hearts of mice when compared to the Dox + placebo group. CONCLUSIONS CM-mESC transplantation improves cardiac function in mice with DIC.
Collapse
Affiliation(s)
- Danúbia Silva dos Santos
- 0000 0001 2294 473Xgrid.8536.8Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
| | - Guilherme Visconde Brasil
- 0000 0001 2294 473Xgrid.8536.8Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
| | - Isalira Peroba Rezende Ramos
- 0000 0001 2294 473Xgrid.8536.8Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
- 0000 0001 2294 473Xgrid.8536.8Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Bloco M, Rio de Janeiro, RJ 21941-902 Brazil
| | - Fernanda Cristina Paccola Mesquita
- 0000 0001 2294 473Xgrid.8536.8Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
| | - Tais Hanae Kasai-Brunswick
- 0000 0001 2294 473Xgrid.8536.8Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
- 0000 0001 2294 473Xgrid.8536.8Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Bloco M, Rio de Janeiro, RJ 21941-902 Brazil
| | - Michelle Lopes Araújo Christie
- 0000 0001 2294 473Xgrid.8536.8Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
| | - Gustavo Monnerat Cahli
- 0000 0001 2294 473Xgrid.8536.8Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
| | - Raiana Andrade Quintanilha Barbosa
- 0000 0001 2294 473Xgrid.8536.8Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
| | - Sandro Torrentes da Cunha
- 0000 0001 2294 473Xgrid.8536.8Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
| | - Jonathas Xavier Pereira
- 0000 0001 2294 473Xgrid.8536.8Departamento de Patologia—Faculdade de Medicina, Hospital Universitário Clementino Fraga Filho, Universiade Federal do Rio de Janeiro, Av. Rodolpho Paulo Rocco, 255, Sub-solo, SAP, Rio de Janeiro, RJ 21910-590 Brazil
| | - Emiliano Medei
- 0000 0001 2294 473Xgrid.8536.8Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
- 0000 0001 2294 473Xgrid.8536.8Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Bloco M, Rio de Janeiro, RJ 21941-902 Brazil
- Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Av. Carlos Chagas Filho 373, Rio de Janeiro, RJ 21941-902 Brazil
| | - Antonio Carlos Campos de Carvalho
- 0000 0001 2294 473Xgrid.8536.8Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
- 0000 0001 2294 473Xgrid.8536.8Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Bloco M, Rio de Janeiro, RJ 21941-902 Brazil
- Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Av. Carlos Chagas Filho 373, Rio de Janeiro, RJ 21941-902 Brazil
| | - Adriana Bastos Carvalho
- 0000 0001 2294 473Xgrid.8536.8Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
- 0000 0001 2294 473Xgrid.8536.8Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Bloco M, Rio de Janeiro, RJ 21941-902 Brazil
- Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Av. Carlos Chagas Filho 373, Rio de Janeiro, RJ 21941-902 Brazil
| | - Regina Coeli dos Santos Goldenberg
- 0000 0001 2294 473Xgrid.8536.8Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373 Bloco G—Sala G2-053, Rio de Janeiro, RJ 21941-902 Brazil
- 0000 0001 2294 473Xgrid.8536.8Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Bloco M, Rio de Janeiro, RJ 21941-902 Brazil
- Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Av. Carlos Chagas Filho 373, Rio de Janeiro, RJ 21941-902 Brazil
| |
Collapse
|
43
|
Xu M, Sheng L, Zhu X, Zeng S, Chi D, Zhang GJ. Protective Effect of Tetrandrine on Doxorubicin-Induced Cardiotoxicity in Rats. TUMORI JOURNAL 2018; 96:460-4. [DOI: 10.1177/030089161009600314] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Aims and background Doxorubicin (Dox) is effective in curative and adjuvant chemotherapy of malignant tumors. Cardiotoxicity is the chief toxic effect that limits the clinical use of Dox. We studied the effects of tetrandrine (Tet) on doxorubicin-induced cardiotoxicity in rats and its protective activity. Materials and methods Sprague-Dawley rats were randomly divided into the following 4 groups: a control group (received only saline), Dox group (received only Dox), Tet/Dox group (received Tet plus Dox), and Tet group (received only Tet). Rats were injected intravenously with 2 mg/kg Dox once a week for 7 weeks and 50 mg/kg Tet was administered intraperitoneally weekly for 7 weeks. Measurements of cardiac contractile parameters including LSVP, +dP/dt max and -dP/dt max, and assessment of electrocardiograms were carried out. Mitochondrial oxidation and phosphorylation state 3 (S3) and state 4 (S4) respiration were measured. Respiration control rate (RCR) and the ADP/O ratio were calculated. Cardiac ultrastructure was examined by electron microscopy. Results Dox induced significant cardiotoxicity in this rat model. The values of LSVP, +dP/dt max, and -dP/dt max in the Tet/Dox group increased as compared to the Dox group (P <0.05). The cardiac contraction and relaxation improved on Tet administration. Tet inhibited the prolonged QT interval on the electrocardiogram in Dox-treated rats. Compared to the Dox group, the values of S3, RCR, and ADP/O increased by more than 28%, 48%, and 27%, respectively, in the Tet/Dox group. Significant cardiac morphological protection was observed in the Tet/Dox-treated rats. Conclusion Tet can improve the reduced cardiac function caused by Dox treatment and prevent Dox-induced mitochondrial impairment in rat cardiotoxicity.
Collapse
Affiliation(s)
- Meng Xu
- Department of Oncology, The First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Lianghe Sheng
- Department of Oncology, The First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Xinhai Zhu
- Department of Oncology, The First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Shibin Zeng
- Department of Oncology, The First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Dexiang Chi
- Department of Oncology, The First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Guo-jun Zhang
- Department of Oncology, The First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| |
Collapse
|
44
|
Delgado JL, Hsieh CM, Chan NL, Hiasa H. Topoisomerases as anticancer targets. Biochem J 2018; 475:373-398. [PMID: 29363591 PMCID: PMC6110615 DOI: 10.1042/bcj20160583] [Citation(s) in RCA: 289] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 12/14/2017] [Accepted: 12/21/2017] [Indexed: 12/15/2022]
Abstract
Many cancer type-specific anticancer agents have been developed and significant advances have been made toward precision medicine in cancer treatment. However, traditional or nonspecific anticancer drugs are still important for the treatment of many cancer patients whose cancers either do not respond to or have developed resistance to cancer-specific anticancer agents. DNA topoisomerases, especially type IIA topoisomerases, are proved therapeutic targets of anticancer and antibacterial drugs. Clinically successful topoisomerase-targeting anticancer drugs act through topoisomerase poisoning, which leads to replication fork arrest and double-strand break formation. Unfortunately, this unique mode of action is associated with the development of secondary cancers and cardiotoxicity. Structures of topoisomerase-drug-DNA ternary complexes have revealed the exact binding sites and mechanisms of topoisomerase poisons. Recent advances in the field have suggested a possibility of designing isoform-specific human topoisomerase II poisons, which may be developed as safer anticancer drugs. It may also be possible to design catalytic inhibitors of topoisomerases by targeting certain inactive conformations of these enzymes. Furthermore, identification of various new bacterial topoisomerase inhibitors and regulatory proteins may inspire the discovery of novel human topoisomerase inhibitors. Thus, topoisomerases remain as important therapeutic targets of anticancer agents.
Collapse
Affiliation(s)
- Justine L Delgado
- Division of Medicinal and Natural Products Chemistry, Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, 115 S Grand Ave., S321 Pharmacy Building, Iowa City, IA 52242, U.S.A
| | - Chao-Ming Hsieh
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei City 100, Taiwan
| | - Nei-Li Chan
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei City 100, Taiwan
| | - Hiroshi Hiasa
- Department of Pharmacology, University of Minnesota Medical School, 6-120 Jackson Hall, 321 Church Street SE, Minneapolis, MN 55455, U.S.A.
| |
Collapse
|
45
|
Cardiotoxic Effects of Short-Term Doxorubicin Administration: Involvement of Connexin 43 in Calcium Impairment. Int J Mol Sci 2017; 18:ijms18102121. [PMID: 29019935 PMCID: PMC5666803 DOI: 10.3390/ijms18102121] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 09/29/2017] [Accepted: 10/09/2017] [Indexed: 12/12/2022] Open
Abstract
The use of Doxorubicin (DOXO), a potent antineoplastic agent, is limited by the development of cardiotoxicity. DOXO-induced cardiotoxicity is multifactorial, although alterations in calcium homeostasis, seem to be involved. Since even the Connexin43 (Cx43) plays a pivotal role in these two phenomena, in this study we have analyzed the effects of DOXO on Cx43 expression and localization. Damage caused by anthracyclines on cardiomyocytes is immediate after each injection, in the present study we used a short-term model of DOXO-induced cardiomyopathy. C57BL/6j female mice were randomly divided in groups and injected with DOXO (2 or 10 mg/kg i.p.) for 1–3 or 7 days once every other day. Cardiac function was assessed by Echocardiography. Sarco/endoplasmic reticulum Ca2+-ATPase (SERCAII) and phospholamban (PLB) expression were assessed by Western blot analysis, intracellular [Ca2+] were detected spectrofluorometrically by means of Fura-2 pentakis (acetoxymethyl) ester (FURA-2AM), and Cx43 and pCx43 expression and localization was analyzed by Western blot and confirmed by immunofluorescence analysis. DOXO induces impairment in Ca2+ homeostasis, already evident after a single administration, and affects Cx43 expression and localization. Our data suggest that DOXO-induced alterations in Ca2+ homeostasis causes in the cells the induction of compensatory mechanisms until a certain threshold, above which cardiac injury is triggered.
Collapse
|
46
|
Evaluation of the Cardiotoxicity of Evodiamine In Vitro and In Vivo. Molecules 2017; 22:molecules22060943. [PMID: 28598372 PMCID: PMC6152646 DOI: 10.3390/molecules22060943] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 06/01/2017] [Accepted: 06/02/2017] [Indexed: 11/27/2022] Open
Abstract
Evodiamine is a bioactive alkaloid that is specified as a biomarker for the quality assessment of Evodia rutaecarpa (E. rutaecarpa) and for traditional Chinese medicines containing this plant. We previously reported that quantitative structure–activity modeling indicated that evodiamine may cause cardiotoxicity. However, previous investigations have indicated that evodiamine has beneficial effects in patients with cardiovascular diseases and there are no previous in vitro or in vivo reports of evodiamine-induced cardiotoxicity. The present study investigated the effects of evodiamine on primary cultured neonatal rat cardiomyocytes in vitro, and on zebrafish in vivo. Cell viability was reduced in vitro, where evodiamine had a 24 h 50% inhibitory concentration of 28.44 µg/mL. Cells exposed to evodiamine also showed increased lactate dehydrogenase release and maleic dialdehyde levels, and reduced superoxide dismutase activity. In vivo, evodiamine had a 10% lethal concentration of 354 ng/mL and induced cardiac malfunction, as evidenced by changes in heart rate and circulation, and pericardial malformations. This study indicated that evodiamine could cause cardiovascular side effects involving oxidative stress. These findings suggest that cardiac function should be monitored in patients receiving preparations containing evodiamine.
Collapse
|
47
|
Salvatorelli E, Menna P, Chello M, Covino E, Minotti G. Modeling Human Myocardium Exposure to Doxorubicin Defines the Risk of Heart Failure from Low-Dose Doxorubicin. J Pharmacol Exp Ther 2017; 362:263-270. [PMID: 28559479 DOI: 10.1124/jpet.117.242388] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 05/26/2017] [Indexed: 11/22/2022] Open
Abstract
The antitumor anthracycline, doxorubicin (DOX), can cause heart failure (HF) upon cumulative administration. Lowering the cumulative dose of DOX proved useful to minimize HF risk, and, yet, there is a growing concern that HF might occur after doses that were thought to be safe. Clinical trials that prospectively address such concerns are lacking. Because HF risk correlates with cardiac exposure to DOX, cumulative doses associated with HF risk were re-explored by modeling the accumulation of anthracycline pools in human myocardium. Ex vivo myocardial samples were used in vitro to simulate DOX rapid infusions. The accumulation of anthracycline pools was measured and incorporated into equations from which a risk versus dose curve was obtained. The experimental curve identified a 5% risk dose that was congruent with a previously reported clinical value (380 versus 400 mg/m2, respectively); however, 1-2% risk occurred after lower doses than reported. Simulations of gain-of-function polymorphism of carbonyl reductase 3, which converts DOX to its poorly diffusible alcohol metabolite, doxorubicinol (DOXOL), expanded anthracycline pools and caused 5% or 1-2% risk doses to decrease to 330 or 180-230 mg DOX/m2, respectively. These data show there is no safe dose of DOX. Diminishing cardiac exposure to circulating DOX may represent a cardioprotective strategy. We show that DOX slow infusions or liposomal DOX, which reduce cardiac exposure to DOX, caused formation of smaller anthracycline pools, did not generate DOXOL, increased the 5% risk dose to 750-800 mg/m2, and prevented HF risk aggravation by carbonyl reductase polymorphism.
Collapse
Affiliation(s)
- Emanuela Salvatorelli
- Drug Sciences (E.S., P.M., G.M.) and Cardiac Surgery (M.C., E.C.), Department of Medicine and Center for Integrated Research, University Campus Bio-Medico, Rome, Italy
| | - Pierantonio Menna
- Drug Sciences (E.S., P.M., G.M.) and Cardiac Surgery (M.C., E.C.), Department of Medicine and Center for Integrated Research, University Campus Bio-Medico, Rome, Italy
| | - Massimo Chello
- Drug Sciences (E.S., P.M., G.M.) and Cardiac Surgery (M.C., E.C.), Department of Medicine and Center for Integrated Research, University Campus Bio-Medico, Rome, Italy
| | - Elvio Covino
- Drug Sciences (E.S., P.M., G.M.) and Cardiac Surgery (M.C., E.C.), Department of Medicine and Center for Integrated Research, University Campus Bio-Medico, Rome, Italy
| | - Giorgio Minotti
- Drug Sciences (E.S., P.M., G.M.) and Cardiac Surgery (M.C., E.C.), Department of Medicine and Center for Integrated Research, University Campus Bio-Medico, Rome, Italy
| |
Collapse
|
48
|
Zhang YS, Aleman J, Shin SR, Kilic T, Kim D, Mousavi Shaegh SA, Massa S, Riahi R, Chae S, Hu N, Avci H, Zhang W, Silvestri A, Sanati Nezhad A, Manbohi A, De Ferrari F, Polini A, Calzone G, Shaikh N, Alerasool P, Budina E, Kang J, Bhise N, Ribas J, Pourmand A, Skardal A, Shupe T, Bishop CE, Dokmeci MR, Atala A, Khademhosseini A. Multisensor-integrated organs-on-chips platform for automated and continual in situ monitoring of organoid behaviors. Proc Natl Acad Sci U S A 2017; 114:E2293-E2302. [PMID: 28265064 PMCID: PMC5373350 DOI: 10.1073/pnas.1612906114] [Citation(s) in RCA: 467] [Impact Index Per Article: 58.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Organ-on-a-chip systems are miniaturized microfluidic 3D human tissue and organ models designed to recapitulate the important biological and physiological parameters of their in vivo counterparts. They have recently emerged as a viable platform for personalized medicine and drug screening. These in vitro models, featuring biomimetic compositions, architectures, and functions, are expected to replace the conventional planar, static cell cultures and bridge the gap between the currently used preclinical animal models and the human body. Multiple organoid models may be further connected together through the microfluidics in a similar manner in which they are arranged in vivo, providing the capability to analyze multiorgan interactions. Although a wide variety of human organ-on-a-chip models have been created, there are limited efforts on the integration of multisensor systems. However, in situ continual measuring is critical in precise assessment of the microenvironment parameters and the dynamic responses of the organs to pharmaceutical compounds over extended periods of time. In addition, automated and noninvasive capability is strongly desired for long-term monitoring. Here, we report a fully integrated modular physical, biochemical, and optical sensing platform through a fluidics-routing breadboard, which operates organ-on-a-chip units in a continual, dynamic, and automated manner. We believe that this platform technology has paved a potential avenue to promote the performance of current organ-on-a-chip models in drug screening by integrating a multitude of real-time sensors to achieve automated in situ monitoring of biophysical and biochemical parameters.
Collapse
Affiliation(s)
- Yu Shrike Zhang
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02139;
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Cambridge, MA 02139
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
| | - Julio Aleman
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02139
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Cambridge, MA 02139
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Su Ryon Shin
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02139
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Cambridge, MA 02139
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
| | - Tugba Kilic
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02139
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Cambridge, MA 02139
- Department of Biomedical Engineering, Faculty of Engineering and Architecture, Izmir Katip Celebi University, Izmir 35620, Turkey
| | - Duckjin Kim
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02139
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Cambridge, MA 02139
| | - Seyed Ali Mousavi Shaegh
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02139
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Cambridge, MA 02139
- Orthopaedic Research Center, Mashhad University of Medical Sciences, Mashhad 9176699199, Iran
| | - Solange Massa
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02139
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Cambridge, MA 02139
- Graduate School Program in Biomedicine, Universidad de los Andes, Santiago 7620001, Chile
| | - Reza Riahi
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02139
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Cambridge, MA 02139
| | - Sukyoung Chae
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02139
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Cambridge, MA 02139
| | - Ning Hu
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02139
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Cambridge, MA 02139
- Biosensor National Special Laboratory, Key Laboratory of Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou 310027, People's Republic of China
| | - Huseyin Avci
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02139
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Cambridge, MA 02139
- Metallurgical and Materials Engineering Department, Faculty of Engineering and Architecture, Eskisehir Osmangazi University, Eskisehir 26030, Turkey
| | - Weijia Zhang
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02139
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Cambridge, MA 02139
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, People's Republic of China
| | - Antonia Silvestri
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02139
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Cambridge, MA 02139
- Department of Electronics and Telecommunications, Polytechnic University of Turin, Turin 10129, Italy
| | - Amir Sanati Nezhad
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02139
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Cambridge, MA 02139
- BioMEMS and Bioinspired Microfluidics Laboratory, Center for Bioengineering Research and Education, Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Ahmad Manbohi
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02139
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Cambridge, MA 02139
- Department of Marine Science, Iranian National Institute for Oceanography and Atmospheric Science, Tehran 1411813389, Iran
| | - Fabio De Ferrari
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02139
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Cambridge, MA 02139
- Department of Electronics and Telecommunications, Polytechnic University of Turin, Turin 10129, Italy
| | - Alessandro Polini
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02139
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Cambridge, MA 02139
| | - Giovanni Calzone
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02139
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Cambridge, MA 02139
| | - Noor Shaikh
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02139
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Cambridge, MA 02139
- Division of Engineering Science, Faculty of Applied Science and Engineering, University of Toronto, Toronto, ON, Canada M5S 1A4
| | - Parissa Alerasool
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02139
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Cambridge, MA 02139
| | - Erica Budina
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02139
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Cambridge, MA 02139
| | - Jian Kang
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02139
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Cambridge, MA 02139
| | - Nupura Bhise
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02139
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Cambridge, MA 02139
| | - João Ribas
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02139
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Cambridge, MA 02139
- Doctoral Program in Experimental Biology and Biomedicine, Center for Neuroscience and Cell Biology, Institute for Interdisciplinary Research, University of Coimbra, Coimbra 3030-789, Portugal
| | - Adel Pourmand
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02139
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Cambridge, MA 02139
- Department of Electrical Engineering, Sahand University of Technology, Tabriz 5331711111, Iran
| | - Aleksander Skardal
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Thomas Shupe
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Colin E Bishop
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Mehmet Remzi Dokmeci
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02139
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Cambridge, MA 02139
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Ali Khademhosseini
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02139;
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Cambridge, MA 02139
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
- Department of Bioindustrial Technologies, College of Animal Bioscience and Technology, Konkuk University, Seoul 143-701, Republic of Korea
- Center for Nanotechnology, King Abdulaziz University, Jeddah 21569, Saudi Arabia
| |
Collapse
|
49
|
Menna P, Salvatorelli E. Primary Prevention Strategies for Anthracycline Cardiotoxicity: A Brief Overview. Chemotherapy 2017; 62:159-168. [DOI: 10.1159/000455823] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 01/07/2017] [Indexed: 11/19/2022]
Abstract
The clinical use of doxorubicin and other antitumor anthracyclines is limited by a dose-related risk of cardiomyopathy and heart failure which may occur “on treatment” or any time, from months to years, after completing chemotherapy. Dose reductions diminish the incidence of cardiac events attributable to anthracyclines, but heart failure still occurs in some patients exposed to low or moderate anthracycline doses. Because anthracyclines improve the life expectancy of patients with, for example, breast cancer or lymphomas, preventing or diminishing the risk of early or delayed cardiotoxicity is of obvious clinical importance. Here, we briefly review some potential strategies of primary prevention that are based on what we know about the molecular mechanisms of cardiotoxicity, and what can be done, or might be done, to interfere with the pharmacokinetic, pharmacodynamic, and genetic determinants of cardiotoxicity.
Collapse
|
50
|
Mitoxantrone-Surfactant Interactions: A Physicochemical Overview. Molecules 2016; 21:molecules21101356. [PMID: 27754390 PMCID: PMC6273455 DOI: 10.3390/molecules21101356] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 09/29/2016] [Accepted: 10/01/2016] [Indexed: 11/17/2022] Open
Abstract
Mitoxantrone is a synthetic anticancer drug used clinically in the treatment of different types of cancer. It was developed as a doxorubicin analogue in a program to find drugs with improved antitumor activity and decreased cardiotoxicity compared with the anthracyclines. As the cell membrane is the first barrier encountered by anticancer drugs before reaching the DNA sites inside the cells and as surfactant micelles are known as simple model systems for biological membranes, the drugs-surfactant interaction has been the subject of great research interest. Further, quantitative understanding of the interactions of drugs with biomimicking structures like surfactant micelles may provide helpful information for the control of physicochemical properties and bioactivities of encapsulated drugs in order to design better delivery systems with possible biomedical applications. The present review describes the physicochemical aspects of the interactions between the anticancer drug mitoxantrone and different surfactants. Mitoxantrone-micelle binding constants, partitions coefficient of the drug between aqueous and micellar phases and the corresponding Gibbs free energy for the above processes, and the probable location of drug molecules in the micelles are discussed.
Collapse
|