101
|
Varga ZV, Ferdinandy P, Liaudet L, Pacher P. Drug-induced mitochondrial dysfunction and cardiotoxicity. Am J Physiol Heart Circ Physiol 2015; 309:H1453-H1467. [PMID: 26386112 PMCID: PMC4666974 DOI: 10.1152/ajpheart.00554.2015] [Citation(s) in RCA: 352] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 09/15/2015] [Indexed: 12/14/2022]
Abstract
Mitochondria has an essential role in myocardial tissue homeostasis; thus deterioration in mitochondrial function eventually leads to cardiomyocyte and endothelial cell death and consequent cardiovascular dysfunction. Several chemical compounds and drugs have been known to directly or indirectly modulate cardiac mitochondrial function, which can account both for the toxicological and pharmacological properties of these substances. In many cases, toxicity problems appear only in the presence of additional cardiovascular disease conditions or develop months/years following the exposure, making the diagnosis difficult. Cardiotoxic agents affecting mitochondria include several widely used anticancer drugs [anthracyclines (Doxorubicin/Adriamycin), cisplatin, trastuzumab (Herceptin), arsenic trioxide (Trisenox), mitoxantrone (Novantrone), imatinib (Gleevec), bevacizumab (Avastin), sunitinib (Sutent), and sorafenib (Nevaxar)], antiviral compound azidothymidine (AZT, Zidovudine) and several oral antidiabetics [e.g., rosiglitazone (Avandia)]. Illicit drugs such as alcohol, cocaine, methamphetamine, ecstasy, and synthetic cannabinoids (spice, K2) may also induce mitochondria-related cardiotoxicity. Mitochondrial toxicity develops due to various mechanisms involving interference with the mitochondrial respiratory chain (e.g., uncoupling) or inhibition of the important mitochondrial enzymes (oxidative phosphorylation, Szent-Györgyi-Krebs cycle, mitochondrial DNA replication, ADP/ATP translocator). The final phase of mitochondrial dysfunction induces loss of mitochondrial membrane potential and an increase in mitochondrial oxidative/nitrative stress, eventually culminating into cell death. This review aims to discuss the mechanisms of mitochondrion-mediated cardiotoxicity of commonly used drugs and some potential cardioprotective strategies to prevent these toxicities.
Collapse
Affiliation(s)
- Zoltán V Varga
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health/National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland; Cardiometabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Peter Ferdinandy
- Cardiometabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary; and
| | - Lucas Liaudet
- Department of Intensive Care Medicine BH 08-621-University Hospital Medical Center, Lausanne, Switzerland
| | - Pál Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health/National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland;
| |
Collapse
|
102
|
Redox mechanism, spectrophotometrical characterisation and voltammetric determination in serum samples of kinases inhibitor and anticancer drug dasatinib. J Electroanal Chem (Lausanne) 2015. [DOI: 10.1016/j.jelechem.2015.06.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
103
|
Huyer LD, Montgomery M, Zhao Y, Xiao Y, Conant G, Korolj A, Radisic M. Biomaterial based cardiac tissue engineering and its applications. Biomed Mater 2015; 10:034004. [PMID: 25989939 PMCID: PMC4464787 DOI: 10.1088/1748-6041/10/3/034004] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cardiovascular disease is a leading cause of death worldwide, necessitating the development of effective treatment strategies. A myocardial infarction involves the blockage of a coronary artery leading to depletion of nutrient and oxygen supply to cardiomyocytes and massive cell death in a region of the myocardium. Cardiac tissue engineering is the growth of functional cardiac tissue in vitro on biomaterial scaffolds for regenerative medicine application. This strategy relies on the optimization of the complex relationship between cell networks and biomaterial properties. In this review, we discuss important biomaterial properties for cardiac tissue engineering applications, such as elasticity, degradation, and induced host response, and their relationship to engineered cardiac cell environments. With these properties in mind, we also emphasize in vitro use of cardiac tissues for high-throughput drug screening and disease modelling.
Collapse
Affiliation(s)
- Locke Davenport Huyer
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Miles Montgomery
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Yimu Zhao
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Yun Xiao
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Genevieve Conant
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Anastasia Korolj
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
| | - Milica Radisic
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Toronto General Research Institute, University Health Network and IBBME, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
104
|
Alameddine RS, Yakan AS, Skouri H, Mukherji D, Temraz S, Shamseddine A. Cardiac and vascular toxicities of angiogenesis inhibitors: The other side of the coin. Crit Rev Oncol Hematol 2015; 96:195-205. [PMID: 26037841 DOI: 10.1016/j.critrevonc.2015.05.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 04/02/2015] [Accepted: 05/05/2015] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis is one of the best-described tumor hallmarks. Targeting angiogenesis is becoming a successful strategy to suppress cancer growth. Vascular endothelial growth factor (VEGF), the fulcrum of angiogenesis, contributes to vascular and cardiac homeostasis. Angiogenesis inhibitors classically associated with vascular side effects are increasingly recognized for cardiac adverse effects as reflected by several meta-analyses. A global approach to these findings is a pressing need, and future strategies involving collaboration among different medical specialties are highly encouraged.
Collapse
Affiliation(s)
- Raafat S Alameddine
- Division of Hematology and Oncology, American University of Beirut, Beirut, Lebanon
| | | | - Hadi Skouri
- Division of Cardiology, American University of Beirut, Beirut, Lebanon
| | - Deborah Mukherji
- Division of Hematology and Oncology, American University of Beirut, Beirut, Lebanon
| | - Sally Temraz
- Division of Hematology and Oncology, American University of Beirut, Beirut, Lebanon
| | - Ali Shamseddine
- Division of Hematology and Oncology, American University of Beirut, Beirut, Lebanon.
| |
Collapse
|
105
|
Maayah ZH, Ghebeh H, Alhaider AA, El-Kadi AO, Soshilov AA, Denison MS, Ansari MA, Korashy HM. Metformin inhibits 7,12-dimethylbenz[a]anthracene-induced breast carcinogenesis and adduct formation in human breast cells by inhibiting the cytochrome P4501A1/aryl hydrocarbon receptor signaling pathway. Toxicol Appl Pharmacol 2015; 284:217-26. [DOI: 10.1016/j.taap.2015.02.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Revised: 02/05/2015] [Accepted: 02/06/2015] [Indexed: 10/24/2022]
|
106
|
Tesori V, Piscaglia AC, Samengo D, Barba M, Bernardini C, Scatena R, Pontoglio A, Castellini L, Spelbrink JN, Maulucci G, Puglisi MA, Pani G, Gasbarrini A. The multikinase inhibitor Sorafenib enhances glycolysis and synergizes with glycolysis blockade for cancer cell killing. Sci Rep 2015; 5:9149. [PMID: 25779766 PMCID: PMC4361992 DOI: 10.1038/srep09149] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 02/20/2015] [Indexed: 12/20/2022] Open
Abstract
Although the only effective drug against primary hepatocarcinoma, the multikinase inhibitor Sorafenib (SFB) usually fails to eradicate liver cancer. Since SFB targets mitochondria, cell metabolic reprogramming may underlie intrinsic tumor resistance. To characterize cancer cell metabolic response to SFB, we measured oxygen consumption, generation of reactive oxygen species (ROS) and ATP content in rat LCSC (Liver Cancer Stem Cells) -2 cells exposed to the drug. Genome wide analysis of gene expression was performed by Affymetrix technology. SFB cytotoxicity was evaluated by multiple assays in the presence or absence of metabolic inhibitors, or in cells genetically depleted of mitochondria. We found that low concentrations (2.5-5 μM) of SFB had a relatively modest effect on LCSC-2 or 293 T cell growth, but damaged mitochondria and increased intracellular ROS. Gene expression profiling of SFB-treated cells was consistent with a shift toward aerobic glycolysis and, accordingly, SFB cytotoxicity was dramatically increased by glucose withdrawal or the glycolytic inhibitor 2-DG. Under metabolic stress, activation of the AMP dependent Protein Kinase (AMPK), but not ROS blockade, protected cells from death. We conclude that mitochondrial damage and ROS drive cell killing by SFB, while glycolytic cell reprogramming may represent a resistance strategy potentially targetable by combination therapies.
Collapse
Affiliation(s)
- Valentina Tesori
- Institute of Internal Medicine and Gastroenterology, Catholic University of the Sacred Heart School of Medicine
| | - Anna Chiara Piscaglia
- Institute of Internal Medicine and Gastroenterology, Catholic University of the Sacred Heart School of Medicine
| | - Daniela Samengo
- Institute of General Pathology, Laboratory of Cell Signaling, Catholic University of the Sacred Heart School of Medicine
| | - Marta Barba
- Institute of Human Anatomy and Cell Biology, Catholic University of the Sacred Heart School of Medicine
| | - Camilla Bernardini
- Institute of Human Anatomy and Cell Biology, Catholic University of the Sacred Heart School of Medicine
| | - Roberto Scatena
- Institute of Biochemistry and Clinical Biochemistry, Catholic University of the sacred Heart School of Medicine
| | - Alessandro Pontoglio
- Institute of Biochemistry and Clinical Biochemistry, Catholic University of the sacred Heart School of Medicine
| | - Laura Castellini
- Department of Radiation Oncology, Center for Clinical Sciences Research, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Johannes N Spelbrink
- Department of Pediatrics, Nijmegen Centre for Mitochondrial Disorders, Radboud University Medical Centre, Geert Grooteplein 10, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands; Institute of Biomedical Technology &Tampere University Hospital, Pirkanmaa Hospital District, University of Tampere, FI-33014, Finland
| | - Giuseppe Maulucci
- Institute of Physics, Catholic University of the Sacred Heart School of Medicine
| | - Maria Ausiliatrice Puglisi
- Institute of Internal Medicine and Gastroenterology, Catholic University of the Sacred Heart School of Medicine
| | - Giovambattista Pani
- Institute of General Pathology, Laboratory of Cell Signaling, Catholic University of the Sacred Heart School of Medicine
| | - Antonio Gasbarrini
- Institute of Internal Medicine and Gastroenterology, Catholic University of the Sacred Heart School of Medicine
| |
Collapse
|
107
|
Weng Z, Luo Y, Yang X, Greenhaw JJ, Li H, Xie L, Mattes WB, Shi Q. Regorafenib impairs mitochondrial functions, activates AMP-activated protein kinase, induces autophagy, and causes rat hepatocyte necrosis. Toxicology 2015; 327:10-21. [DOI: 10.1016/j.tox.2014.11.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 10/22/2014] [Accepted: 11/03/2014] [Indexed: 12/16/2022]
|
108
|
Gillani TB, Rawling T, Murray M. Cytochrome P450-Mediated Biotransformation of Sorafenib and Its N-Oxide Metabolite: Implications for Cell Viability and Human Toxicity. Chem Res Toxicol 2014; 28:92-102. [DOI: 10.1021/tx500373g] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Tina B. Gillani
- Pharmacogenomics
and Drug Development Group, Discipline of Pharmacology, University of Sydney, Sydney, NSW 2006, Australia
| | - Tristan Rawling
- School
of Pharmacy, Graduate School of Health, University of Technology, Sydney, PO
Box 123, Broadway, Ultimo, NSW 2007, Australia
| | - Michael Murray
- Pharmacogenomics
and Drug Development Group, Discipline of Pharmacology, University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
109
|
Martinez Marignac VL, Smith S, Toban N, Bazile M, Aloyz R. Resistance to Dasatinib in primary chronic lymphocytic leukemia lymphocytes involves AMPK-mediated energetic re-programming. Oncotarget 2014; 4:2550-66. [PMID: 24334291 PMCID: PMC3926848 DOI: 10.18632/oncotarget.1508] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) is the most common leukemia in adults in the western world. Although promising new therapies for this incurable disease are being tested in clinical trials, the therapeutic relevance of metabolic rewiring in chronic lymphocytic leukemia (CLL) is poorly understood. The aim of this study was to identify targetable metabolic differences in primary CLL lymphocytes by the use of Dasatinib. Dasatinib is a multi-tyrosine kinase inhibitor used to treat chronic myelogenous leukemia (CML) and is being tested in clinical trials for several cancers including CLL. This drug has been shown to be beneficial to CML patients suffering from diabetes by reducing their glucose plasma levels. In keeping with this previous observation, we report that Dasatinib induced glucose use while reducing lactate production, suggesting that this tyrosine kinase inhibitor decreases aerobic glycolysis and shifts glucose use in primary CLL lymphocytes. Our results suggest that primary CLL lymphocytes (independently of traditional prognostic factors) can be stratified in two subsets by their sensitivity to Dasatinib in vitro. Increased glucose use induced by Dasatinib or by inhibition of mitochondrial respiration was not sufficient to sustain survival and ATP levels in CLL samples sensitive to Dasatinib. The two subsets of primary CLL lymphocytes are characterized as well by a differential dependency on mitochondrial respiration and the use of anabolic or catabolic processes to cope with induced metabolic/energetic stress. Differential metabolic reprogramming between subsets is supported by the contrasting effect on the survival of Dasatinib treated CLL lymphocytes with pharmacological inhibition of two master metabolic regulators (mTorc1 and AMPK) as well as induced autophagy. Alternative metabolic organization between subsets is further supported by the differential basal expression (freshly purified lymphocytes) of active AMPK, regulators of glucose metabolism and modulators of AKT signaling. The contrasting metabolic features revealed by our strategy could be used to metabolically target CLL lymphocyte subsets creating new therapeutic windows for this disease for mTORC1 or AMPK inhibitors. Indeed, we report that Metformin, a drug used to treat diabetes was selectively cytotoxic to Dasatinib sensitive samples. Ultimately, we suggest that a similar strategy could be applied to other cancer types by using Dasatinib and/or relevant tyrosine kinase inhibitors.
Collapse
|
110
|
ZhiQing Z, XinXing W, Jingbo G, Rui Z, Xiujie G, Yun Z, Lei W, Xue L, LingJia Q. Effects of HIP in protection of HSP70 for stress-induced cardiomyocytes injury and its glucorticoid receptor pathway. Cell Stress Chaperones 2014; 19:865-75. [PMID: 24789270 PMCID: PMC4389846 DOI: 10.1007/s12192-014-0510-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 03/26/2014] [Accepted: 03/27/2014] [Indexed: 01/16/2023] Open
Abstract
Moderate levels of stress can be beneficial to health, while stress overload can cause injury or contribute to diseases. Despite a number of studies of adaptation or stress damage, the mechanisms of adaptation and stress damage remain far from clear. The effect and mechanisms of adaptation on cardiomyocytes damage caused by stress overload are discussed in this study. Data showed that mild repeated stress mitigated stress overload-induced cardiomyocyte injury both in an animal model of restraint stress and in H9C2 cells with GC (glucocorticoid) treatment. HSP70, HIP expression and interaction between HSP70 and HIP increased during adaptation induced by mild stress both in animals and H9C2 cells. Overexpression or inhibition of HSP70 in H9C2 cells with pCDNA-3.1-Hsp70 or KNK437 (HSP70 inhibitor) showed that HSP70 can protect H9C2 cells from GC-induced cell damage. A luciferase assay showed that Hsp70 plays its protective role through inhibition of GR transcription activity dependent on the interaction with HIP. These results indicated that HSP70 may promote adaptation with its interacting protein HIP, and increased levels of HSP70 and its interacting protein HIP during adaptation may play a protective role on stress-overload-induced cardiomyocyte injury.
Collapse
MESH Headings
- Adaptation, Physiological
- Animals
- Benzhydryl Compounds/pharmacology
- Cell Line
- Disease Models, Animal
- Glucocorticoids/toxicity
- HSP70 Heat-Shock Proteins/antagonists & inhibitors
- HSP70 Heat-Shock Proteins/genetics
- HSP70 Heat-Shock Proteins/metabolism
- Heart Diseases/genetics
- Heart Diseases/metabolism
- Heart Diseases/pathology
- Heart Diseases/prevention & control
- Hydrocortisone/blood
- Male
- Molecular Chaperones/genetics
- Molecular Chaperones/metabolism
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Pyrrolidinones/pharmacology
- RNA Interference
- Rats, Wistar
- Receptors, Glucocorticoid/agonists
- Receptors, Glucocorticoid/genetics
- Receptors, Glucocorticoid/metabolism
- Restraint, Physical
- Signal Transduction/drug effects
- Stress, Psychological/complications
- Stress, Psychological/genetics
- Stress, Psychological/metabolism
- Stress, Psychological/pathology
- Time Factors
- Transfection
Collapse
Affiliation(s)
- Zhang ZhiQing
- />Institute of Health & Environmental Medicine, Tianjin, 300050 People’s Republic of China
| | - Wang XinXing
- />Institute of Basic Medical Sciences, Beijing, 100850 People’s Republic of China
| | - Gong Jingbo
- />Institute of Basic Medical Sciences, Beijing, 100850 People’s Republic of China
| | - Zhan Rui
- />Institute of Basic Medical Sciences, Beijing, 100850 People’s Republic of China
| | - Gao Xiujie
- />Institute of Health & Environmental Medicine, Tianjin, 300050 People’s Republic of China
| | - Zhao Yun
- />Institute of Basic Medical Sciences, Beijing, 100850 People’s Republic of China
| | - Wu Lei
- />Institute of Health & Environmental Medicine, Tianjin, 300050 People’s Republic of China
| | - Leng Xue
- />Institute of Medical Equipment, Tianjin, 300161 People’s Republic of China
| | - Qian LingJia
- />Institute of Basic Medical Sciences, Beijing, 100850 People’s Republic of China
| |
Collapse
|
111
|
Barr LA, Makarewich CA, Berretta RM, Gao H, Troupes CD, Woitek F, Recchia F, Kubo H, Force T, Houser SR. Imatinib activates pathological hypertrophy by altering myocyte calcium regulation. Clin Transl Sci 2014; 7:360-7. [PMID: 24931551 DOI: 10.1111/cts.12173] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Imatinib mesylate is a selective tyrosine-kinase inhibitor used in the treatment of multiple cancers, most notably chronic myelogenous leukemia. There is evidence that imatinib can induce cardiotoxicity in cancer patients. Our hypothesis is that imatinib alters calcium regulatory mechanisms and can contribute to development of pathological cardiac hypertrophy. METHODS AND RESULTS Neonatal rat ventricular myocytes (NRVMs) were treated with clinical doses (low: 2 μM; high: 5 μM) of imatinib and assessed for molecular changes. Imatinib increased peak systolic Ca(2+) and Ca(2+) transient decay rates and Western analysis revealed significant increases in phosphorylation of phospholamban (Thr-17) and the ryanodine receptor (Ser-2814), signifying activation of calcium/calmodulin-dependent kinase II (CaMKII). Imatinib significantly increased NRVM volume as assessed by Coulter counter, myocyte surface area, and atrial natriuretic peptide abundance seen by Western. Imatinib induced cell death, but did not activate the classical apoptotic program as assessed by caspase-3 cleavage, indicating a necrotic mechanism of death in myocytes. We expressed AdNFATc3-green fluorescent protein in NRVMs and showed imatinib treatment significantly increased nuclear factor of activated T cells translocation that was inhibited by the calcineurin inhibitor FK506 or CaMKII inhibitors. CONCLUSION These data show that imatinib can activate pathological hypertrophic signaling pathways by altering intracellular Ca(2+) dynamics. This is likely a contributing mechanism for the adverse cardiac effects of imatinib.
Collapse
Affiliation(s)
- Larry A Barr
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
112
|
Papadopoulou LC, Kyriazou AV, Bonovolias ID, Tsiftsoglou AS. Imatinib inhibits the expression of SCO2 and FRATAXIN genes that encode mitochondrial proteins in human Bcr-Abl⁺ leukemia cells. Blood Cells Mol Dis 2014; 53:84-90. [PMID: 24726617 DOI: 10.1016/j.bcmd.2014.03.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 03/06/2014] [Accepted: 03/06/2014] [Indexed: 12/19/2022]
Abstract
Imatinib mesylate (IM/Gleevec®), a selective inhibitor of chimeric Bcr-Abl tyrosine kinase, was developed as a first line drug to treat CML and ALL Ph(+) patients. Earlier studies have shown that hemin counteracts the IM-induced cell killing in human K-562 CML cells. In this study, we investigated whether IM disrupts the heme-dependent Cytochrome c Oxidase (COX) Biosynthesis and Assembly Pathway (HDCBAP) in Bcr-Abl(+) and Bcr-Abl(-) cells by affecting the expression of key-genes. Cells were exposed to IM and evaluated at time intervals for cell growth, cell death, expression of various genes by RT-PCR analysis as well as Sco2 mature protein levels by western blot analysis and COX enzymatic activity. IM at 1 μM induced extensive cell growth inhibition and cell death as well as marked suppression of the expression of SCO2 and FRATAXIN (FXN) genes in human K-562 and KU-812 Bcr-Abl(+) CML cells. IM also reduced the protein level of mature Sco2 mitochondrial protein as well as COX activity in these cell lines. However, treatment of human MOLT-4 Bcr-Abl(-) cells with 1μM and even with higher concentrations (4×10(-5)M) of IM neither reduced the expression of SCO2 and FXN genes nor suppressed the protein level of mature Sco2 protein and COX activity. Our findings indicate that SCO2 and FXN genes, involved in HDCBAP, are repressed by IM in human Bcr-Abl(+) CML cells and may represent novel target sites in leukemia therapy.
Collapse
Affiliation(s)
- Lefkothea C Papadopoulou
- Laboratory of Pharmacology, Department of Pharmaceutical Sciences, Aristotle University of Thessaloniki (A.U.TH.), GR-54124 Thessaloniki, Macedonia, Greece
| | - Angeliki V Kyriazou
- Laboratory of Pharmacology, Department of Pharmaceutical Sciences, Aristotle University of Thessaloniki (A.U.TH.), GR-54124 Thessaloniki, Macedonia, Greece
| | - Ioannis D Bonovolias
- Laboratory of Pharmacology, Department of Pharmaceutical Sciences, Aristotle University of Thessaloniki (A.U.TH.), GR-54124 Thessaloniki, Macedonia, Greece
| | - Asterios S Tsiftsoglou
- Laboratory of Pharmacology, Department of Pharmaceutical Sciences, Aristotle University of Thessaloniki (A.U.TH.), GR-54124 Thessaloniki, Macedonia, Greece.
| |
Collapse
|
113
|
Li J, Cousin C, Tinkler H, Touhami J, Petit V, Thomas N. Profiling of Nutrient Transporter Expression in Human Stem Cell–Derived Cardiomyocytes Exposed to Tyrosine Kinase Inhibitor Anticancer Drugs Using RBD Ligands. ACTA ACUST UNITED AC 2014; 19:1185-92. [DOI: 10.1177/1087057114533724] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 04/08/2014] [Indexed: 02/03/2023]
Abstract
We applied a novel profiling approach using receptor binding domain (RBD) ligands to cell surface domains of a panel of nutrient transporters to characterize the impact of a number of tyrosine kinase inhibitor anticancer drugs on human stem cell–derived cardiomyocytes. High-content screening and flow cytometry analysis showed diagnostic changes in nutrient transporter expression correlating with glycolysis and oxidative phosphorylation–based cell metabolism in glucose and galactose media. Cluster analysis of RBD binding signatures of drug-treated cells cultured in glucose medium showed good correlation with sensitization of mitochondrial toxicity in cells undergoing oxidative phosphorylation in galactose medium. These data demonstrate the potential for RBD ligands as profiling tools to improve the clinical predictivity of in vitro cell assays for drug toxicity.
Collapse
Affiliation(s)
- Jianliang Li
- GE Healthcare, The Maynard Centre, Forest Farm, Whitchurch, Cardiff, UK
| | | | - Hayley Tinkler
- GE Healthcare, The Maynard Centre, Forest Farm, Whitchurch, Cardiff, UK
| | - Jawida Touhami
- METAFORA Biosystems, Pépinière Genopole Entreprises, Evry, France
| | - Vincent Petit
- METAFORA Biosystems, Pépinière Genopole Entreprises, Evry, France
| | - Nick Thomas
- GE Healthcare, The Maynard Centre, Forest Farm, Whitchurch, Cardiff, UK
| |
Collapse
|
114
|
Canonical and new generation anticancer drugs also target energy metabolism. Arch Toxicol 2014; 88:1327-50. [PMID: 24792321 DOI: 10.1007/s00204-014-1246-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 04/15/2014] [Indexed: 01/05/2023]
Abstract
Significant efforts have been made for the development of new anticancer drugs (protein kinase or proteasome inhibitors, monoclonal humanized antibodies) with presumably low or negligible side effects and high specificity. However, an in-depth analysis of the side effects of several currently used canonical (platin-based drugs, taxanes, anthracyclines, etoposides, antimetabolites) and new generation anticancer drugs as the first line of clinical treatment reveals significant perturbation of glycolysis and oxidative phosphorylation. Canonical and new generation drug side effects include decreased (1) intracellular ATP levels, (2) glycolytic/mitochondrial enzyme/transporter activities and/or (3) mitochondrial electrical membrane potentials. Furthermore, the anti-proliferative effects of these drugs are markedly attenuated in tumor rho (0) cells, in which functional mitochondria are absent; in addition, several anticancer drugs directly interact with isolated mitochondria affecting their functions. Therefore, several anticancer drugs also target the energy metabolism, and hence, the documented inhibitory effect of anticancer drugs on cancer growth should also be linked to the blocking of ATP supply pathways. These often overlooked effects of canonical and new generation anticancer drugs emphasize the role of energy metabolism in maintaining cancer cells viable and its targeting as a complementary and successful strategy for cancer treatment.
Collapse
|
115
|
Nisr RB, Affourtit C. Insulin acutely improves mitochondrial function of rat and human skeletal muscle by increasing coupling efficiency of oxidative phosphorylation. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1837:270-6. [PMID: 24212054 PMCID: PMC4331040 DOI: 10.1016/j.bbabio.2013.10.012] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 10/24/2013] [Accepted: 10/29/2013] [Indexed: 12/24/2022]
Abstract
Insulin is essential for the regulation of fuel metabolism and triggers the uptake of glucose by skeletal muscle. The imported glucose is either stored or broken down, as insulin stimulates glycogenesis and ATP synthesis. The mechanism by which ATP production is increased is incompletely understood at present and, generally, relatively little functional information is available on the effect of insulin on mitochondrial function. In this paper we have exploited extracellular flux technology to investigate insulin effects on the bioenergetics of rat (L6) and human skeletal muscle myoblasts and myotubes. We demonstrate that a 20-min insulin exposure significantly increases (i) the cell respiratory control ratio, (ii) the coupling efficiency of oxidative phosphorylation, and (iii) the glucose sensitivity of anaerobic glycolysis. The improvement of mitochondrial function is explained by an insulin-induced immediate decrease of mitochondrial proton leak. Palmitate exposure annuls the beneficial mitochondrial effects of insulin. Our data improve the mechanistic understanding of insulin-stimulated ATP synthesis, and reveal a hitherto undisclosed insulin sensitivity of cellular bioenergetics that suggests a novel way of detecting insulin responsiveness of cells.
Collapse
Affiliation(s)
- Raid B Nisr
- School of Biomedical & Healthcare Sciences, Plymouth University, Drake Circus, Plymouth, UK.
| | - Charles Affourtit
- School of Biomedical & Healthcare Sciences, Plymouth University, Drake Circus, Plymouth, UK.
| |
Collapse
|
116
|
Finsterer J, Ohnsorge P. Influence of mitochondrion-toxic agents on the cardiovascular system. Regul Toxicol Pharmacol 2013; 67:434-45. [DOI: 10.1016/j.yrtph.2013.09.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Revised: 09/01/2013] [Accepted: 09/02/2013] [Indexed: 10/26/2022]
|
117
|
Heyen JR, Hu W, Jamieson J, Thibault S, Batugo M, Loi CM, Burns-Naas LA, McHarg AD, Jessen B. Cardiovascular differentiation of imatinib and bosutinib in the rat. Int J Hematol 2013; 98:597-607. [DOI: 10.1007/s12185-013-1453-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 08/13/2013] [Accepted: 10/06/2013] [Indexed: 12/23/2022]
|
118
|
Doherty KR, Wappel RL, Talbert DR, Trusk PB, Moran DM, Kramer JW, Brown AM, Shell SA, Bacus S. Multi-parameter in vitro toxicity testing of crizotinib, sunitinib, erlotinib, and nilotinib in human cardiomyocytes. Toxicol Appl Pharmacol 2013; 272:245-55. [PMID: 23707608 DOI: 10.1016/j.taap.2013.04.027] [Citation(s) in RCA: 133] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 04/24/2013] [Accepted: 04/28/2013] [Indexed: 01/28/2023]
|
119
|
Preservation of cardiomyocytes from the adult heart. J Mol Cell Cardiol 2013; 64:108-19. [PMID: 24051370 DOI: 10.1016/j.yjmcc.2013.09.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 08/08/2013] [Accepted: 09/05/2013] [Indexed: 11/21/2022]
Abstract
Cardiomyocytes represent one of the most useful models to conduct cardiac research. A single adult heart yields millions of cardiomyocytes, but these cells do not survive for long after isolation. We aimed to determine whether inhibition of myosin II ATPase that is essential for muscle contraction may preserve fully differentiated adult cardiomyocytes. Using inhibitors of the myosin II ATPase, blebbistatin and N-benzyl-p-toluene sulphonamide (BTS), we preserved freshly isolated fully differentiated adult primary cardiomyocytes that were stored at a refrigerated temperature. Specifically, preserved cardiomyocytes stayed viable for a 2-week period with a stable expression of cardiac genes and retained the expression of key markers characteristic of cardiomyocytes. Furthermore, voltage-clamp, action potential, calcium transient and contractility studies confirmed that the preserved cardiomyocytes are comparable to freshly isolated cells. Long-term exposure of preserved cardiomyocytes to four tyrosine kinase inhibitors, sunitinib malate, dasatinib, sorafenib tosylate and imatinib mesylate, revealed their potential to induce cardiac toxicity that was manifested with a decrease in contractility and induction of cell death, but this toxicity was not observed in acute experiments conducted over the time course amenable to freshly prepared cardiomyocytes. This study introduces the concept that the inhibition of myosin II ATPase safeguards the structure and function of fully differentiated adult cardiomyocytes. The fact that these preserved cardiomyocytes can be used for numerous days after preparation makes them a robust and versatile tool in cardiac research and allows the investigation of long-term exposure to novel drugs on cardiomyocyte function.
Collapse
|
120
|
Fumarola C, Caffarra C, La Monica S, Galetti M, Alfieri RR, Cavazzoni A, Galvani E, Generali D, Petronini PG, Bonelli MA. Effects of sorafenib on energy metabolism in breast cancer cells: role of AMPK-mTORC1 signaling. Breast Cancer Res Treat 2013; 141:67-78. [PMID: 23963659 DOI: 10.1007/s10549-013-2668-x] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 08/09/2013] [Indexed: 12/18/2022]
Abstract
In this study, we investigated the effects and the underlying molecular mechanisms of the multi-kinase inhibitor sorafenib in a panel of breast cancer cell lines. Sorafenib inhibited cell proliferation and induced apoptosis through the mitochondrial pathway. These effects were neither correlated with modulation of MAPK and AKT pathways nor dependent on the ERα status. Sorafenib promoted an early perturbation of mitochondrial function, inducing a deep depolarization of mitochondrial membrane, associated with drop of intracellular ATP levels and increase of ROS generation. As a response to this stress condition, the energy sensor AMPK was rapidly activated in all the cell lines analyzed. In MCF-7 and SKBR3 cells, AMPK enhanced glucose uptake by up-regulating the expression of GLUT-1 glucose transporter, as also demonstrated by AMPKα1 RNA interference, and stimulated aerobic glycolysis thus increasing lactate production. Moreover, the GLUT-1 inhibitor fasentin blocked sorafenib-induced glucose uptake and potentiated its cytotoxic activity in SKBR3 cells. Persistent activation of AMPK by sorafenib finally led to the impairment of glucose metabolism both in MCF-7 and SKBR3 cells as well as in the highly glycolytic MDA-MB-231 cells, resulting in cell death. This previously unrecognized long-term effect of sorafenib was mediated by AMPK-dependent inhibition of the mTORC1 pathway. Suppression of mTORC1 activity was sufficient for sorafenib to hinder glucose utilization in breast cancer cells, as demonstrated by the observation that the mTORC1 inhibitor rapamycin induced a comparable down-regulation of GLUT-1 expression and glucose uptake. The key role of AMPK-dependent inhibition of mTORC1 in sorafenib mechanisms of action was confirmed by AMPKα1 silencing, which restored mTORC1 activity conferring a significant protection from cell death. This study provides insights into the molecular mechanisms driving sorafenib anti-tumoral activity in breast cancer, and supports the need for going on with clinical trials aimed at proving the efficacy of sorafenib for breast cancer treatment.
Collapse
Affiliation(s)
- Claudia Fumarola
- Department of Clinical and Experimental Medicine, University of Parma, Via Volturno, 39, Parma 43125, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
121
|
Dy GK, Adjei AA. Understanding, recognizing, and managing toxicities of targeted anticancer therapies. CA Cancer J Clin 2013; 63:249-79. [PMID: 23716430 DOI: 10.3322/caac.21184] [Citation(s) in RCA: 235] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 02/21/2013] [Accepted: 02/25/2013] [Indexed: 12/11/2022] Open
Abstract
Answer questions and earn CME/CNE Advances in genomics and molecular biology have identified aberrant proteins in cancer cells that are attractive targets for cancer therapy. Because these proteins are overexpressed or dysregulated in cancer cells compared with normal cells, it was assumed that their inhibitors will be narrowly targeted and relatively nontoxic. However, this hope has not been achieved. Current targeted agents exhibit the same frequency and severity of toxicities as traditional cytotoxic agents, with the main difference being the nature of the toxic effects. Thus, the classical chemotherapy toxicities of alopecia, myelosuppression, mucositis, nausea, and vomiting have been generally replaced by vascular, dermatologic, endocrine, coagulation, immunologic, ocular, and pulmonary toxicities. These toxicities need to be recognized, prevented, and optimally managed.
Collapse
Affiliation(s)
- Grace K Dy
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York, USA
| | | |
Collapse
|
122
|
Porceddu M, Buron N, Roussel C, Labbe G, Fromenty B, Borgne-Sanchez A. Prediction of liver injury induced by chemicals in human with a multiparametric assay on isolated mouse liver mitochondria. Toxicol Sci 2013; 129:332-45. [PMID: 22987451 PMCID: PMC3446843 DOI: 10.1093/toxsci/kfs197] [Citation(s) in RCA: 127] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Drug-induced liver injury (DILI) in humans is difficult to predict using classical
in vitro cytotoxicity screening and regulatory animal studies. This
explains why numerous compounds are stopped during clinical trials or withdrawn from the
market due to hepatotoxicity. Thus, it is important to improve early prediction of DILI in
human. In this study, we hypothesized that this goal could be achieved by investigating
drug-induced mitochondrial dysfunction as this toxic effect is a major mechanism of DILI.
To this end, we developed a high-throughput screening platform using isolated mouse liver
mitochondria. Our broad spectrum multiparametric assay was designed to detect the global
mitochondrial membrane permeabilization (swelling), inner membrane permeabilization
(transmembrane potential), outer membrane permeabilization (cytochrome c
release), and alteration of mitochondrial respiration driven by succinate or
malate/glutamate. A pool of 124 chemicals (mainly drugs) was selected, including 87 with
documented DILI and 37 without reported clinical hepatotoxicity. Our screening assay
revealed an excellent sensitivity for clinical outcome of DILI (94 or 92% depending on
cutoff) and a high positive predictive value (89 or 82%). A highly significant
relationship between drug-induced mitochondrial toxicity and DILI occurrence in patients
was calculated (p < 0.001). Moreover, this multiparametric assay
allowed identifying several compounds for which mitochondrial toxicity had never been
described before and even helped to clarify mechanisms with some drugs already known to be
mitochondriotoxic. Investigation of drug-induced loss of mitochondrial integrity and
function with this multiparametric assay should be considered for integration into basic
screening processes at early stage to select drug candidates with lower risk of DILI in
human. This assay is also a valuable tool for assessing the mitochondrial toxicity profile
and investigating the mechanism of action of new compounds and marketed compounds.
Collapse
|
123
|
Pointon A, Abi-Gerges N, Cross MJ, Sidaway JE. Phenotypic Profiling of Structural Cardiotoxins In Vitro Reveals Dependency on Multiple Mechanisms of Toxicity. Toxicol Sci 2013; 132:317-26. [DOI: 10.1093/toxsci/kft005] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
124
|
Tsiper MV, Sturgis J, Avramova LV, Parakh S, Fatig R, Juan-García A, Li N, Rajwa B, Narayanan P, Qualls CW, Robinson JP, Davisson VJ. Differential mitochondrial toxicity screening and multi-parametric data analysis. PLoS One 2012; 7:e45226. [PMID: 23077490 PMCID: PMC3471932 DOI: 10.1371/journal.pone.0045226] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Accepted: 08/20/2012] [Indexed: 11/18/2022] Open
Abstract
Early evaluation of new drug entities for their potential to cause mitochondrial dysfunction is becoming an important task for drug development. Multi-parametric high-content screening (mp-HCS) of mitochondrial toxicity holds promise as a lead in-vitro strategy for drug testing and safety evaluations. In this study, we have developed a mp-HCS and multi-parametric data analysis scheme for assessing cell responses to induced mitochondrial perturbation. The mp-HCS measurements are shown to be robust enough to allow for quantitative comparison of biological systems with different metabolic pathways simulated by alteration of growth media. Substitution of medium glucose for galactose sensitized cells to drug action and revealed novel response parameters. Each compound was quantitatively characterized according to induced phenotypic changes of cell morphology and functionality measured by fluorescent biomarkers for mitochondrial activity, plasma membrane permeability, and nuclear morphology. Descriptors of drug effects were established by generation of a SCRIT (Specialized-Cell-Response-to-Induced-Toxicity) vector, consisting of normalized statistical measures of each parameter at each dose and growth condition. The dimensionality of SCRIT vectors depends on the number of parameters chosen, which in turn depends on the hypothesis being tested. Specifically, incorporation of three parameters of response into SCRIT vectors enabled clustering of 84 training compounds with known pharmacological and toxicological activities according to the degree of toxicity and mitochondrial involvement. Inclusion of 6 parameters enabled the resolution of more subtle differences between compounds within a common therapeutic class; scoring enabled a ranking of statins in direct agreement with clinical outcomes. Comparison of drug-induced changes required variations in glucose for separation of mitochondrial dysfunction from other types of cytotoxicity. These results also demonstrate that the number of drugs in a training set, the choice of parameters used in analysis, and statistical measures are fundamental for specific hypothesis testing and assessment of quantitative phenotypic differences.
Collapse
Affiliation(s)
- Maria V. Tsiper
- Bindley Bioscience Center at Purdue University Discovery Park, West Lafayette, Indiana, United States of America
| | - Jennifer Sturgis
- Department of Basic Medical Sciences, Purdue University, West Lafayette, Indiana, United States of America
| | - Larisa V. Avramova
- Bindley Bioscience Center at Purdue University Discovery Park, West Lafayette, Indiana, United States of America
| | - Shilpa Parakh
- Bindley Bioscience Center at Purdue University Discovery Park, West Lafayette, Indiana, United States of America
| | - Raymond Fatig
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana, United States of America
| | - Ana Juan-García
- Department of Basic Medical Sciences, Purdue University, West Lafayette, Indiana, United States of America
| | - Nianyu Li
- Comparative Biology and Safety Sciences, Amgen Inc, Seattle, Washington, United States of America
| | - Bartek Rajwa
- Bindley Bioscience Center at Purdue University Discovery Park, West Lafayette, Indiana, United States of America
| | - Padma Narayanan
- Comparative Biology and Safety Sciences, Amgen Inc, Seattle, Washington, United States of America
| | - C. W. Qualls
- Comparative Biology and Safety Sciences, Amgen Inc, Seattle, Washington, United States of America
| | - J. Paul Robinson
- Department of Basic Medical Sciences, Purdue University, West Lafayette, Indiana, United States of America
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, United States of America
| | - V. Jo Davisson
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana, United States of America
| |
Collapse
|
125
|
Hu W, Hirakawa B, Jessen B, Lee M, Aguirre S. A tyrosine kinase inhibitor-induced myocardial degeneration in rats through off-target phosphodiesterase inhibition. J Appl Toxicol 2012; 32:1008-20. [PMID: 22936366 DOI: 10.1002/jat.2801] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Revised: 06/26/2012] [Accepted: 06/27/2012] [Indexed: 11/08/2022]
Abstract
PF-04254644 is a selective kinase inhibitor of mesenchymal epithelial transition factor/hepatocyte growth factor receptor with known off-target inhibitory activity against the phosphodiesterase (PDE) family. Rats given repeated oral doses of PF-04254644 developed a mild to moderate myocardial degeneration accompanied by sustained increase in heart rate and contractility. Investigative studies were conducted to delineate the mechanisms of toxicity. Microarray analysis of Sprague-Dawley rat hearts in a 6 day repeat dose study with PF-04254644 or milrinone, a selective PDE3 inhibitor, revealed similar perturbation of the cyclic adenosine monophosphate (c-AMP) pathway. PDE inhibition and activation of c-AMP were further substantiated using PDE3B immunofluorescence staining and through a c-AMP response element reporter gene assay. The intracellular calcium and oxidative stress signaling pathways were more perturbed by treatment with PF-04254644 than milrinone. The rat cardiomyocytes calcium assay found a dose-dependent increase in intracellular calcium with PF-04254644 treatment. These data suggest that cardiotoxicity of PF-04254644 was probably due to activation of c-AMP signaling, and possibly subsequent disruption of intracellular calcium and oxidative stress signaling pathways. The greater response with PF-04254644 as compared with milrinone in gene expression and micro- and ultrastructural changes is probably due to the broader panel of PDEs inhibition.
Collapse
Affiliation(s)
- Wenyue Hu
- Pfizer Inc, Drug Safety Research and Development, La Jolla Laboratories, 10646 Science Center Drive, San Diego, CA 92121, USA.
| | | | | | | | | |
Collapse
|
126
|
Rana P, Anson B, Engle S, Will Y. Characterization of human-induced pluripotent stem cell-derived cardiomyocytes: bioenergetics and utilization in safety screening. Toxicol Sci 2012; 130:117-31. [PMID: 22843568 DOI: 10.1093/toxsci/kfs233] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Cardiotoxicity remains the number one reason for drug withdrawal from the market, and Food and Drug Administration issued black box warnings, thus demonstrating the need for more predictive preclinical safety screening, especially early in the drug discovery process when much chemical substrate is available. Whereas human-ether-a-go-go related gene screening has become routine to mitigate proarrhythmic risk, the development of in vitro assays predicting additional on- and off-target biochemical toxicities will benefit from cellular models exhibiting true cardiomyocyte characteristics such as native tissue-like mitochondrial activity. Human stem cell-derived tissue cells may provide such a model. This hypothesis was tested using a combination of flux analysis, gene and protein expression, and toxicity-profiling techniques to characterize mitochondrial function in induced pluripotent stem cell (iPSC) derived human cardiomyocytes in the presence of differing carbon sources over extended periods in cell culture. Functional analyses demonstrate that iPSC-derived cardiomyocytes are (1) capable of utilizing anaerobic or aerobic respiration depending upon the available carbon substrate and (2) bioenergetically closest to adult heart tissue cells when cultured in galactose or galactose supplemented with fatty acids. We utilized this model to test a variety of kinase inhibitors with known clinical cardiac liabilities for their potential toxicity toward these cells. We found that the kinase inhibitors showed a dose-dependent toxicity to iPSC cardiomyocytes grown in galactose and that oxygen consumption rates were significantly more affected than adenosine triphosphate production. Sorafenib was found to have the most effect, followed by sunitinib, dasatinib, imatinib, lapatinib, and nioltinib.
Collapse
Affiliation(s)
- Payal Rana
- Compound Safety Prediction, Pfizer Global R&D, Groton, Connecticut, USA
| | | | | | | |
Collapse
|
127
|
Hu W, Lu S, McAlpine I, Jamieson JD, Lee DU, Marroquin LD, Heyen JR, Jessen BA. Mechanistic investigation of imatinib-induced cardiac toxicity and the involvement of c-Abl kinase. Toxicol Sci 2012; 129:188-99. [PMID: 22641616 DOI: 10.1093/toxsci/kfs192] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The Bcr-abl tyrosine kinase inhibitor imatinib mesylate is the frontline therapy for chronic myeloid leukemia. Imatinib has been reported to cause congestive heart failure and left ventricular contractile dysfunction in patients and cardiomyopathy in rodents, findings proposed to be associated with its pharmacological activity. To investigate the specific role of Abelson oncogene 1 (c-Abl) in imatinib-induced cardiac toxicity, we performed targeted gene inhibition of c-Abl by RNA interference in neonatal cardiomyocytes (NCMs). Suppression of c-Abl did not lead to cytotoxicity or induction of endoplasmic reticulum (ER) stress. To further dis associate c-Abl from imatinib-induced cardiac toxicity, we designed imatinib structural analogs that do not have appreciable c-Abl inhibition in NCMs. The c-Abl inactive analogs induced cytotoxicity and ER stress, at similar or greater potencies and magnitudes as imatinib. Furthermore, combining c-Abl gene silencing with imatinib and analogs treatment did not significantly shift the cytotoxicity dose response curves. Imatinib and analogs were shown to accumulate in lysosomes, likely due to their physicochemical properties, and disrupt autophagy. The toxicity induced by imatinib and analogs can be rescued by bafilomycin A pretreatment, demonstrating the involvement of lysosomal accumulation in cardiac toxicity. The results from our studies strongly suggest that imatinib induces cardiomyocyte dysfunction through disruption of autophagy and induction of ER stress, independent of c-Abl inhibition.
Collapse
Affiliation(s)
- Wenyue Hu
- Drug Safety Research and Development, La Jolla Laboratories, Pfizer Inc., San Diego, California 92121, USA.
| | | | | | | | | | | | | | | |
Collapse
|
128
|
Elevation of pulmonary artery pressure as a complication of nilotinib therapy for chronic myeloid leukemia. Int J Hematol 2012; 96:132-5. [DOI: 10.1007/s12185-012-1103-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Revised: 05/08/2012] [Accepted: 05/11/2012] [Indexed: 10/28/2022]
|
129
|
Dasanu CA, Padmanabhan P, Clark BA, Do C. Cardiovascular toxicity associated with small molecule tyrosine kinase inhibitors currently in clinical use. Expert Opin Drug Saf 2012; 11:445-57. [DOI: 10.1517/14740338.2012.672971] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
130
|
Greineder CF, Kohnstamm S, Ky B. Heart failure associated with sunitinib: lessons learned from animal models. Curr Hypertens Rep 2012; 13:436-41. [PMID: 21826469 DOI: 10.1007/s11906-011-0225-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Sunitinib is a highly potent, multitargeted anticancer agent. However, there is growing clinical evidence that sunitinib induces cardiac dysfunction. Disruption of multiple signaling pathways, which are important in the maintenance of adult cardiac function, is likely to result in cardiovascular toxicity. Basic and translational evidence implicates a potential role for specific growth factor signaling pathways. This review discusses the relevant translational data from animal models of heart failure, focusing on three key pathways that are inhibited by sunitinib: AMP-activated protein kinase (AMPK), platelet-derived growth factor receptors (PDGFRs), and the vascular endothelial growth factor receptors (VEGFRs) 1, 2, and 3. We hypothesize that disruption of these pathways by sunitinib results in cardiotoxicity, and present direct and indirect evidence to support the notion that sunitinib-induced cardiac dysfunction likely involves a variety of molecular mechanisms that are critical for cardiac homeostasis.
Collapse
Affiliation(s)
- Colin F Greineder
- Department of Pharmacology, Institute for Translational Medicine and Therapeutics, Department of Emergency Medicine, University of Pennsylvania School of Medicine, 3400 Spruce Street, Philadelphia, PA 19104, USA.
| | | | | |
Collapse
|
131
|
Montani D, Bergot E, Günther S, Savale L, Bergeron A, Bourdin A, Bouvaist H, Canuet M, Pison C, Macro M, Poubeau P, Girerd B, Natali D, Guignabert C, Perros F, O'Callaghan DS, Jaïs X, Tubert-Bitter P, Zalcman G, Sitbon O, Simonneau G, Humbert M. Pulmonary arterial hypertension in patients treated by dasatinib. Circulation 2012; 125:2128-37. [PMID: 22451584 DOI: 10.1161/circulationaha.111.079921] [Citation(s) in RCA: 426] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND The French pulmonary hypertension (PH) registry allows the survey of epidemiological trends. Isolated cases of precapillary PH have been reported in patients who have chronic myelogenous leukemia treated with the tyrosine kinase inhibitor dasatinib. METHODS AND RESULTS This study was designed to describe incident cases of dasatinib-associated PH reported in the French PH registry. From the approval of dasatinib (November 2006) to September 30, 2010, 9 incident cases treated by dasatinib at the time of PH diagnosis were identified. At diagnosis, patients had moderate to severe precapillary PH with functional and hemodynamic impairment. No other incident PH cases were exposed to other tyrosine kinase inhibitors at the time of PH diagnosis. Clinical, functional, or hemodynamic improvements were observed within 4 months of dasatinib discontinuation in all but 1 patient. Three patients required PH treatment with endothelin receptor antagonist (n=2) or calcium channel blocker (n=1). After a median follow-up of 9 months (min-max 3-36), the majority of patients did not demonstrate complete clinical and hemodynamic recovery, and no patients reached a normal value of mean pulmonary artery pressure (≤20 mm Hg). Two patients (22%) died at follow-up (1 of unexplained sudden death and 1 of cardiac failure in the context of septicemia, respectively, 8 and 12 months after dasatinib withdrawal). The lowest estimate of incident PH occurring in patients exposed to dasatinib in France was 0.45%. CONCLUSIONS Dasatinib may induce severe precapillary PH fulfilling the criteria of pulmonary arterial hypertension, thus suggesting a direct and specific effect of dasatinib on pulmonary vessels. Improvement is usually observed after withdrawal of dasatinib.
Collapse
Affiliation(s)
- David Montani
- Univ. Paris-Sud, Faculté de Médecine, Le Kremlin-Bicêtre, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
132
|
Bull VH, Rajalingam K, Thiede B. Sorafenib-induced mitochondrial complex I inactivation and cell death in human neuroblastoma cells. J Proteome Res 2012; 11:1609-20. [PMID: 22268697 DOI: 10.1021/pr200790e] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Sorafenib is a multikinase inhibitor that is approved for use against renal cell and hepatocellular carcinoma. We found that sorafenib potently induced cell death in human neuroblastoma cells. To understand the molecular basis of sorafenib-mediated cell death in human SH-SY5Y cells, we performed a temporal quantitative proteome analysis. The results showed significant quantitative changes of 193 unique proteins. Bioinformatics-assisted pathway analysis of the regulated proteins revealed that mitochondrial proteins, especially components of the electron transport chain and the mitochondrial ribosomes, were significantly affected upon exposure to sorafenib. The observed down-regulation of the respiratory chain complex I (NADH dehydrogenase) was accompanied with loss of mitochondrial transmembrane potential (Δψm) and complete impairment of complex I enzyme activity. The destabilization of complex I subunits was consistent, rapid, and independent of caspase activation as well as Bcl-2 overexpression. This study provides an overview of the molecular machinery driving sorafenib-mediated cell death in neuroblastoma cells and suggests that sorafenib could be a potential therapeutic drug for the treatment of neuroblastoma.
Collapse
Affiliation(s)
- Vibeke Hervik Bull
- The Biotechnology Centre of Oslo, University of Oslo , P.O. Box 1125 Blindern, 0317 Oslo, Norway
| | | | | |
Collapse
|
133
|
Chiusa M, Hool SL, Truetsch P, Djafarzadeh S, Jakob SM, Seifriz F, Scherer SJ, Suter TM, Zuppinger C, Zbinden S. Cancer therapy modulates VEGF signaling and viability in adult rat cardiac microvascular endothelial cells and cardiomyocytes. J Mol Cell Cardiol 2012; 52:1164-75. [PMID: 22326847 DOI: 10.1016/j.yjmcc.2012.01.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Revised: 01/10/2012] [Accepted: 01/26/2012] [Indexed: 12/11/2022]
Abstract
This work was motivated by the incomplete characterization of the role of vascular endothelial growth factor-A (VEGF-A) in the stressed heart in consideration of upcoming cancer treatment options challenging the natural VEGF balance in the myocardium. We tested, if the cytotoxic cancer therapy doxorubicin (Doxo) or the anti-angiogenic therapy sunitinib alters viability and VEGF signaling in primary cardiac microvascular endothelial cells (CMEC) and adult rat ventricular myocytes (ARVM). ARVM were isolated and cultured in serum-free medium. CMEC were isolated from the left ventricle and used in the second passage. Viability was measured by LDH-release and by MTT-assay, cellular respiration by high-resolution oxymetry. VEGF-A release was measured using a rat specific VEGF-A ELISA-kit. CMEC were characterized by marker proteins including CD31, von Willebrand factor, smooth muscle actin and desmin. Both Doxo and sunitinib led to a dose-dependent reduction of cell viability. Sunitinib treatment caused a significant reduction of complex I and II-dependent respiration in cardiomyocytes and the loss of mitochondrial membrane potential in CMEC. Endothelial cells up-regulated VEGF-A release after peroxide or Doxo treatment. Doxo induced HIF-1α stabilization and upregulation at clinically relevant concentrations of the cancer therapy. VEGF-A release was abrogated by the inhibition of the Erk1/2 or the MAPKp38 pathway. ARVM did not answer to Doxo-induced stress conditions by the release of VEGF-A as observed in CMEC. VEGF receptor 2 amounts were reduced by Doxo and by sunitinib in a dose-dependent manner in both CMEC and ARVM. In conclusion, these data suggest that cancer therapy with anthracyclines modulates VEGF-A release and its cellular receptors in CMEC and ARVM, and therefore alters paracrine signaling in the myocardium.
Collapse
Affiliation(s)
- Manuel Chiusa
- Bern University Hospital, Cardiology, CH-3010 Bern, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
134
|
Aparicio-Gallego G, Blanco M, Figueroa A, García-Campelo R, Valladares-Ayerbes M, Grande-Pulido E, Antón-Aparicio L. New insights into molecular mechanisms of sunitinib-associated side effects. Mol Cancer Ther 2011; 10:2215-23. [PMID: 22161785 DOI: 10.1158/1535-7163.mct-10-1124] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The introduction of targeted therapy represents a major advance in the treatment of tumor progression. Targeted agents are a novel therapeutic approach developed to disrupt different cellular signaling pathways. The tyrosine kinase inhibitor sunitinib specifically blocks multiple tyrosine kinase receptors that are involved in the progression of many tumors. Sunitinib is the current standard of care in first-line treatment of advanced renal cell carcinoma, and it is approved in imatinib-intolerant and imatinib-refractory gastrointestinal stromal tumors. However, it is increasingly evident that sunitinib may display collateral effects on other proteins beyond its main target receptors, eliciting undesirable and unexpected adverse events. A better understanding of the molecular mechanisms underlying these undesirable sunitinib-associated side effects will help physicians to maximize efficacy of sunitinib and minimize adverse events. Here, we focus on new insights into molecular mechanisms that may mediate sunitinib-associated adverse events.
Collapse
Affiliation(s)
- Guadalupe Aparicio-Gallego
- Clinical Oncology Department and Biomedical Research Institute (INIBIC), A Coruña University Hospital, A Coruña, Spain
| | | | | | | | | | | | | |
Collapse
|
135
|
Hedhli N, Russell KS. Cardiotoxicity of molecularly targeted agents. Curr Cardiol Rev 2011; 7:221-33. [PMID: 22758623 PMCID: PMC3322440 DOI: 10.2174/157340311799960636] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Revised: 06/08/2011] [Accepted: 12/07/2011] [Indexed: 02/06/2023] Open
Abstract
Cardiac toxicity of molecularly targeted cancer agents is increasingly recognized as a significant side effect of chemotherapy. These new potent therapies may not only affect the survival of cancer cells, but have the potential to adversely impact normal cardiac and vascular function. Unraveling the mechanisms by which these therapies affect the heart and vasculature is crucial for improving drug design and finding alternative therapies to protect patients predisposed to cardiovascular disease. In this review, we summarize the classification and side effects of currently approved molecularly targeted chemotherapeutics.
Collapse
Affiliation(s)
- Nadia Hedhli
- Department of Internal Medicine-Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Kerry S Russell
- Department of Internal Medicine-Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
136
|
Herman EH, Knapton A, Rosen E, Thompson K, Rosenzweig B, Estis J, Agee S, Lu QA, Todd JA, Lipshultz S, Hasinoff B, Zhang J. A multifaceted evaluation of imatinib-induced cardiotoxicity in the rat. Toxicol Pathol 2011; 39:1091-106. [PMID: 21937741 DOI: 10.1177/0192623311419524] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Cardiotoxicity was an unanticipated side effect elicited by the clinical use of imatinib (Imb). This toxicity has been examined in only a limited number of experimental studies. The present study sought, by a variety of approaches, to identify important characteristics of Imb-induced cardiac alterations. Male spontaneously hypertensive rats (SHRs) received oral doses of 10, 30, or 50 mg/kg Imb or water daily for 10 d. Cardiac lesions, detected at all doses, were characterized by cytoplasmic vacuolization and myofibrillar loss. In a second experiment, cardiac lesions were found in Sprague Dawley (SD) and SHR rats given 50 or 100 mg/kg Imb for 14 d. Mean cardiac lesion scores and serum levels of cardiac troponin I were higher in SHRs than in SD rats. Imb induced myocyte death by necrosis, autophagy, and apoptosis. Dose-related increases in cardiac expression were observed for several genes associated with endoplasmic reticulum stress response, protein folding, and vascular development and remodeling. Imb caused alterations in isolated myocytes (myofibrillar loss, highly disrupted and disorganized sarcomeric α-actinin, apoptosis, and increased lactate dehydrogenase release) at low concentrations (5 mM). The authors conclude that Imb exerts cardiotoxic effects that are manifest through a complex pattern of cellular alterations, the severity of which can be influenced by arterial blood pressure.
Collapse
Affiliation(s)
- Eugene H Herman
- Food and Drug Administration, Division of Applied Pharmacology Research, Silver Spring, Maryland, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
137
|
Abstract
Assessing mitochondrial dysfunction requires definition of the dysfunction to be investigated. Usually, it is the ability of the mitochondria to make ATP appropriately in response to energy demands. Where other functions are of interest, tailored solutions are required. Dysfunction can be assessed in isolated mitochondria, in cells or in vivo, with different balances between precise experimental control and physiological relevance. There are many methods to measure mitochondrial function and dysfunction in these systems. Generally, measurements of fluxes give more information about the ability to make ATP than do measurements of intermediates and potentials. For isolated mitochondria, the best assay is mitochondrial respiratory control: the increase in respiration rate in response to ADP. For intact cells, the best assay is the equivalent measurement of cell respiratory control, which reports the rate of ATP production, the proton leak rate, the coupling efficiency, the maximum respiratory rate, the respiratory control ratio and the spare respiratory capacity. Measurements of membrane potential provide useful additional information. Measurement of both respiration and potential during appropriate titrations enables the identification of the primary sites of effectors and the distribution of control, allowing deeper quantitative analyses. Many other measurements in current use can be more problematic, as discussed in the present review.
Collapse
|
138
|
Fiume L, Manerba M, Vettraino M, Di Stefano G. Effect of sorafenib on the energy metabolism of hepatocellular carcinoma cells. Eur J Pharmacol 2011; 670:39-43. [PMID: 21924262 DOI: 10.1016/j.ejphar.2011.08.038] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Revised: 08/12/2011] [Accepted: 08/27/2011] [Indexed: 01/09/2023]
Abstract
Recent data demonstrated that sorafenib impaired the oxidative phosphorylation of a rat myogenic cell line and suggested that this biochemical lesion can contribute to the cardiac toxicity caused by the drug. With the experiments reported here, we verified whether sorafenib inhibits oxidative phosphorylation also in cells from human hepatocellular carcinomas (HCCs), which are treated with this drug. By using the HCC cell lines PLC/PRF/5 and SNU-449 we studied the effects of the drug on ATP cellular levels, oxygen consumption and aerobic glycolysis, a metabolic pathway generally used by neoplastic cells to meet their energy demand. The effect of sorafenib on ATP cellular levels was also studied in cells grown in a glucose-free medium, which only derive their energy from oxidative phosphorylation. We found that at clinically relevant concentrations sorafenib hindered oxidative phosphorylation, whereas at the same time stimulated aerobic glycolysis in glucose-grown cells, thus attenuating the cellular ATP depletion. These results support the impairment of oxidative phosphorylation as a mechanism contributing to the antineoplastic activity of sorafenib in the treatment of HCCs.
Collapse
Affiliation(s)
- Luigi Fiume
- Department of Experimental Pathology, University of Bologna, Italy
| | | | | | | |
Collapse
|
139
|
Blasi E, Heyen J, Patyna S, Hemkens M, Ramirez D, John-Baptiste A, Steidl-Nichols J, McHarg A. Sunitinib, a receptor tyrosine kinase inhibitor, increases blood pressure in rats without associated changes in cardiac structure and function. Cardiovasc Ther 2011; 30:287-94. [PMID: 21884012 DOI: 10.1111/j.1755-5922.2011.00278.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Sunitinib, a multi-tyrosine kinase inhibitor has demonstrated clinical activity in advanced renal cell carcinoma and imatinib-resistant/intolerant gastrointestinal stromal tumor. It has been associated with manageable hypertension and other unique toxicities. AIMS Two nonclinical studies were conducted to determine if sunitinib has direct/indirect effects on cardiac structure/function that may be related to hypertension at clinically relevant exposures. MATERIALS & METHODS Rats received once-daily vehicle or sunitinib 1 or 10 mg/kg/day (n = 10/group) orally for 4 weeks, followed by 2 weeks off treatment then a 2-week rechallenge. Blood pressure (BP) and heart rate (HR) were continuously acquired and echocardiograms were obtained weekly. Effects of sunitinib and its metabolite (0.003-0.3 μM) were also evaluated in guinea pig isolated Langendorff-perfused hearts (n = 4-6 hearts/group). RESULTS Sunitinib 10 mg/kg/day produced significant (P < 0.05) hemodynamic changes: 24 h average BP increased during initial dosing/rechallenge, with rebound hypotension during the off-treatment period; 24 h average HR increased during the off-treatment period, and decreased during rechallenge; no changes in cardiac structure/function were observed. In guinea pig isolated hearts, neither sunitinib nor its metabolite had direct effects on contractility, HR or left ventricular pressure. DISCUSSION & CONCLUSION These studies demonstrate that sunitinib/metabolite had no direct effects on cardiac function ex vivo, and that therapeutically relevant concentrations of sunitinib dosed on a "clinical schedule" increased BP in rats without adverse changes in cardiac structure/function.
Collapse
Affiliation(s)
- E Blasi
- Safety Pharmacology-Pfizer Global Research and Development, La Jolla, CA, USA.
| | | | | | | | | | | | | | | |
Collapse
|
140
|
Tolcher AW, Appleman LJ, Shapiro GI, Mita AC, Cihon F, Mazzu A, Sundaresan PR. A phase I open-label study evaluating the cardiovascular safety of sorafenib in patients with advanced cancer. Cancer Chemother Pharmacol 2011; 67:751-64. [PMID: 20521052 PMCID: PMC3064895 DOI: 10.1007/s00280-010-1372-3] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2010] [Accepted: 05/14/2010] [Indexed: 11/30/2022]
Abstract
PURPOSE To characterize the cardiovascular profile of sorafenib, a multitargeted kinase inhibitor, in patients with advanced cancer. METHODS Fifty-three patients with advanced cancer received oral sorafenib 400 mg bid in continuous 28-day cycles in this open-label study. Left ventricular ejection fraction (LVEF) was evaluated using multigated acquisition scanning at baseline and after 2 and 4 cycles of sorafenib. QT/QTc interval on the electrocardiograph (ECG) was measured in triplicate with a Holter 12-lead ECG at baseline and after 1 cycle of sorafenib. Heart rate (HR) and blood pressure (BP) were obtained in duplicate at baseline and after 1 and 4 cycles of sorafenib. Plasma pharmacokinetic data were obtained for sorafenib and its 3 main metabolites after 1 and 4 cycles of sorafenib. RESULTS LVEF (SD) mean change from baseline was -0.8 (±8.6) LVEF(%) after 2 cycles (n = 31) and -1.2 (±7.8) LVEF(%) after 4 cycles of sorafenib (n = 24). The QT/QTc mean changes from baseline observed at maximum sorafenib concentrations (t(max)) after 1 cycle (n = 31) were small (QTcB: 4.2 ms; QTcF: 9.0 ms). Mean changes observed after 1 cycle in BP (n = 31) and HR (n = 30) at maximum sorafenib concentrations (t(max)) were moderate (up to 11.7 mm Hg and -6.6 bpm, respectively). No correlation was found between the AUC and C(max) of sorafenib and its main metabolites and any cardiovascular parameters. CONCLUSIONS The effects of sorafenib on changes in QT/QTc interval on the ECG, LVEF, BP, and HR were modest and unlikely to be of clinical significance in the setting of advanced cancer treatment.
Collapse
Affiliation(s)
- Anthony W Tolcher
- START (South Texas Accelerated Research Therapeutics), 4319 Medical Drive, Suite 205, San Antonio, TX 78229, USA.
| | | | | | | | | | | | | |
Collapse
|
141
|
Cardiotoxicity of kinase inhibitors: the prediction and translation of preclinical models to clinical outcomes. Nat Rev Drug Discov 2011; 10:111-26. [PMID: 21283106 DOI: 10.1038/nrd3252] [Citation(s) in RCA: 275] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
142
|
Targeting non-malignant disorders with tyrosine kinase inhibitors. Nat Rev Drug Discov 2011; 9:956-70. [PMID: 21119733 DOI: 10.1038/nrd3297] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Receptor and non-receptor tyrosine kinases are involved in multiple proliferative signalling pathways. Imatinib, one of the first tyrosine kinase inhibitors (TKIs) to be approved, revolutionized the treatment of chronic myelogenous leukaemia, and other TKIs with different spectra of kinase inhibition are used to treat renal cell carcinoma, non-small-cell lung cancer and colon cancer. Studies also support the potential use of TKIs as anti-proliferative agents in non-malignant disorders such as cardiac hypertrophy, and in benign-proliferative disorders including pulmonary hypertension, lung fibrosis, rheumatoid disorders, atherosclerosis, in-stent restenosis and glomerulonephritis. In this Review, we provide an overview of the most recent developments--both experimental as well as clinical--regarding the therapeutic potential of TKIs in non-malignant disorders.
Collapse
|
143
|
Mellor HR, Bell AR, Valentin JP, Roberts RRA. Cardiotoxicity Associated with Targeting Kinase Pathways in Cancer. Toxicol Sci 2010; 120:14-32. [DOI: 10.1093/toxsci/kfq378] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
144
|
Nowis D, Maczewski M, Mackiewicz U, Kujawa M, Ratajska A, Wieckowski MR, Wilczyński GM, Malinowska M, Bil J, Salwa P, Bugajski M, Wójcik C, Siński M, Abramczyk P, Winiarska M, Dabrowska-Iwanicka A, Duszyński J, Jakóbisiak M, Golab J. Cardiotoxicity of the anticancer therapeutic agent bortezomib. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:2658-68. [PMID: 20519734 DOI: 10.2353/ajpath.2010.090690] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Recent case reports provided alarming signals that treatment with bortezomib might be associated with cardiac events. In all reported cases, patients experiencing cardiac problems were previously or concomitantly treated with other chemotherapeutics including cardiotoxic anthracyclines. Therefore, it is difficult to distinguish which components of the therapeutic regimens contribute to cardiotoxicity. Here, we addressed the influence of bortezomib on cardiac function in rats that were not treated with other drugs. Rats were treated with bortezomib at a dose of 0.2 mg/kg thrice weekly. Echocardiography, histopathology, and electron microscopy were used to evaluate cardiac function and structural changes. Respiration of the rat heart mitochondria was measured polarographically. Cell culture experiments were used to determine the influence of bortezomib on cardiomyocyte survival, contractility, Ca(2+) fluxes, induction of endoplasmic reticulum stress, and autophagy. Our findings indicate that bortezomib treatment leads to left ventricular contractile dysfunction manifested by a significant drop in left ventricle ejection fraction. Dramatic ultrastructural abnormalities of cardiomyocytes, especially within mitochondria, were accompanied by decreased ATP synthesis and decreased cardiomyocyte contractility. Monitoring of cardiac function in bortezomib-treated patients should be implemented to evaluate how frequently cardiotoxicity develops especially in patients with pre-existing cardiac conditions, as well as when using additional cardiotoxic drugs.
Collapse
Affiliation(s)
- Dominika Nowis
- Department of Immunology, Center of Biostructure Research, Medical University of Warsaw, 1A Banacha Str., F Building, 02-097 Warsaw, Poland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
145
|
Kappers MH, van Esch JH, Sluiter W, Sleijfer S, Danser AHJ, van den Meiracker AH. Hypertension Induced by the Tyrosine Kinase Inhibitor Sunitinib Is Associated With Increased Circulating Endothelin-1 Levels. Hypertension 2010; 56:675-81. [DOI: 10.1161/hypertensionaha.109.149690] [Citation(s) in RCA: 207] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Mariëtte H.W. Kappers
- From the Division of Pharmacology, Vascular and Metabolic Diseases, Department of Internal Medicine (M.H.W.K., J.H.M.v.E., A.H.J.D., A.H.v.d.M.) and Departments of Neurology (W.S.) and Medical Oncology (S.S.), Erasmus MC, Rotterdam, The Netherlands
| | - Joep H.M. van Esch
- From the Division of Pharmacology, Vascular and Metabolic Diseases, Department of Internal Medicine (M.H.W.K., J.H.M.v.E., A.H.J.D., A.H.v.d.M.) and Departments of Neurology (W.S.) and Medical Oncology (S.S.), Erasmus MC, Rotterdam, The Netherlands
| | - Wim Sluiter
- From the Division of Pharmacology, Vascular and Metabolic Diseases, Department of Internal Medicine (M.H.W.K., J.H.M.v.E., A.H.J.D., A.H.v.d.M.) and Departments of Neurology (W.S.) and Medical Oncology (S.S.), Erasmus MC, Rotterdam, The Netherlands
| | - Stefan Sleijfer
- From the Division of Pharmacology, Vascular and Metabolic Diseases, Department of Internal Medicine (M.H.W.K., J.H.M.v.E., A.H.J.D., A.H.v.d.M.) and Departments of Neurology (W.S.) and Medical Oncology (S.S.), Erasmus MC, Rotterdam, The Netherlands
| | - A. H. Jan Danser
- From the Division of Pharmacology, Vascular and Metabolic Diseases, Department of Internal Medicine (M.H.W.K., J.H.M.v.E., A.H.J.D., A.H.v.d.M.) and Departments of Neurology (W.S.) and Medical Oncology (S.S.), Erasmus MC, Rotterdam, The Netherlands
| | - Anton H. van den Meiracker
- From the Division of Pharmacology, Vascular and Metabolic Diseases, Department of Internal Medicine (M.H.W.K., J.H.M.v.E., A.H.J.D., A.H.v.d.M.) and Departments of Neurology (W.S.) and Medical Oncology (S.S.), Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
146
|
Kerkela R, Woulfe KC, Durand JB, Vagnozzi R, Kramer D, Chu TF, Beahm C, Chen MH, Force T. Sunitinib-induced cardiotoxicity is mediated by off-target inhibition of AMP-activated protein kinase. Clin Transl Sci 2010; 2:15-25. [PMID: 20376335 DOI: 10.1111/j.1752-8062.2008.00090.x] [Citation(s) in RCA: 196] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Tyrosine kinase inhibitors (TKIs) are transforming the treatment of patients with malignancies. One such agent, sunitinib (Sutent, Pfizer), has demonstrated activity against a variety of solid tumors. Sunitinib is "multi-targeted," inhibiting growth factor receptors that regulate both tumor angiogenesis and tumor cell survival. However cardiac dysfunction has been associated with its use. Identification of the target of sunitinib associated cardiac dysfunction could guide future drug design to reduce toxicity while preserving anti-cancer activity. Herein we identify severe mitochondrial structural abnormalities in the heart of a patient with sunitinib-induced heart failure. In cultured cardiomyocytes, sunitinib induces loss of mitochondrial membrane potential and energy rundown. Despite the latter, AMPK activity, which should be increased in the setting of energy compromise, is reduced in hearts of sunitinib-treated mice and cardiomyocytes in culture and this is due to direct inhibition of AMPK by sunitinib. Critically, we find that adenovirus-mediated gene transfer of an actived mutant of AMPK reduces sunitinib-induced cell death. Our findings suggest AMPK inhibition plays a central role in sunitinib cardiomyocyte toxicity, highlighting the potential of off-target effects of TKIs contributing to cardiotoxicity. While multi-targeting can enhance tumor cell killing, this must be balanced against the potential increased risk of cardiac dysfunction.
Collapse
Affiliation(s)
- Risto Kerkela
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
147
|
French KJ, Coatney RW, Renninger JP, Hu CX, Gales TL, Zhao S, Storck LM, Davis CB, McSurdy-Freed J, Chen E, Frazier KS. Differences in Effects on Myocardium and Mitochondria by Angiogenic Inhibitors Suggest Separate Mechanisms of Cardiotoxicity. Toxicol Pathol 2010; 38:691-702. [DOI: 10.1177/0192623310373775] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Several multikinase angiogenesis inhibitors demonstrate mitochondrial and/or cardiovascular toxicity, suggesting an on-target pharmacologic effect. To evaluate whether cardiotoxicity is directly related to vascular endothelial growth factor receptor inhibition, we investigated the effects of sunitinib, sorafenib, and pazopanib on myocardial function and structure. We used a rat model to assess myocardial effects of the inhibitors concurrently exposed to the cardiac stressor dobutamine. Echocardiographic abnormalities including premature ventricular contractions, decreases in heart rate, circumferential strain, and radial and circumferential strain rates were noted with sorafenib, but not with sunitinib or pazopanib. Ultrastructural analysis of ventricular cardiomyocytes by transmission electron microscopy revealed mitochondrial swelling, dense deposits, and matrix cavitation in rats given sunitinib and disrupted mitochondrial cristae in rats given sorafenib, but there were no effects with pazopanib. Effects on neonatal rat cardiomyocyte cultures were assessed, which identified decreases in mitochondrial membrane potential with sunitinib treatment, but not with sorafenib or pazopanib. Intracellular adenosine triphosphate depletion was observed with sunitinib and sorafenib, but not pazopanib. Our results show that cardiotoxicity is not necessarily related to a pharmacologic classwide effect of vascular endothelial growth factor receptor inhibition, and the rat myocardial structural and functional changes identified in this study may be instead a result of inhibition of other kinase pathways, the mechanism of which may be associated with mitochondrial toxicity.
Collapse
Affiliation(s)
- Kevin J. French
- Department of Safety Assessment, GlaxoSmithKline, King of Prussia, Pennsylvania, USA
| | - Robert W. Coatney
- Department of Laboratory Animal Sciences, GlaxoSmithKline, King of Prussia, Pennsylvania, USA
| | - Jon P. Renninger
- Department of Safety Assessment, GlaxoSmithKline, King of Prussia, Pennsylvania, USA
| | - Catherine X. Hu
- Department of Safety Assessment, GlaxoSmithKline, King of Prussia, Pennsylvania, USA
| | - Tracy L. Gales
- Department of Safety Assessment, GlaxoSmithKline, King of Prussia, Pennsylvania, USA
| | - Shufang Zhao
- Department of Laboratory Animal Sciences, GlaxoSmithKline, King of Prussia, Pennsylvania, USA
| | - Laura M. Storck
- Department of Safety Assessment, GlaxoSmithKline, King of Prussia, Pennsylvania, USA
| | - Charles B. Davis
- Department of Oncology Drug Discovery, GlaxoSmithKline, King of Prussia, Pennsylvania, USA
| | - Jeanelle McSurdy-Freed
- Department of Oncology Drug Discovery, GlaxoSmithKline, King of Prussia, Pennsylvania, USA
| | - Emile Chen
- Department of Drug Metabolism and Pharmacokinetics, GlaxoSmithKline, King of Prussia, Pennsylvania, USA
| | - Kendall S. Frazier
- Department of Safety Assessment, GlaxoSmithKline, King of Prussia, Pennsylvania, USA
| |
Collapse
|
148
|
Kamada P, Dudek AZ. Sorafenib therapy for metastatic renal carcinoma in patients with low cardiac ejection fraction: report of two cases and literature review. Cancer Invest 2010; 28:501-4. [PMID: 20014944 DOI: 10.3109/07357900903476794] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Targeted therapies used in the treatment of metastatic renal cell carcinoma (RCC) are known to have the potential for cardiotoxicity and should be used with caution in patients with cardiac comorbidities. A retrospective review identified two RCC cases treated with sorafenib in the context of preexisting cardiomyopathy. Sorafenib therapy resulted in disease stabilization of progressing RCC for both cases, without worsening of cardiac ejection fraction. Further evaluation of the cardiac safety of sorafenib in patients with cardiomyopathy is warranted.
Collapse
Affiliation(s)
- Pratima Kamada
- Department of Oncology, and Transplantation, University of Minnesota, Minneapolis, USA
| | | |
Collapse
|
149
|
Hasinoff BB, Patel D. Mechanisms of myocyte cytotoxicity induced by the multikinase inhibitor sorafenib. Cardiovasc Toxicol 2010; 10:1-8. [PMID: 19915982 DOI: 10.1007/s12012-009-9056-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The use of the anticancer multikinase inhibitor sorafenib is associated with cardiac ischemia or infarction and an increase in hypertension. We investigated various mechanisms that might be responsible for its cardiotoxicity in a neonatal rat myocyte model. As measured by lactate dehydrogenase release, sorafenib treatment of myocytes caused dose-dependent damage at therapeutically relevant concentrations. It had been hypothesized that inhibition of RAF1 and BRAF kinases may be responsible for sorafenib induced cardiotoxicity. However, because sorafenib treatment did not inhibit phosphorylation of ERK (extracellular signal-regulated kinase), it was concluded that sorafenib did not exert its damaging effects through RAF inhibition of the RAF/MEK/ERK kinase cascade. The clinically approved doxorubicin cardioprotective agent dexrazoxane did not protect myocytes from damage. At lower sorafenib concentrations, at least, these results are consistent with sorafenib not being able to induce significant oxidative damage. In conclusion, given the extreme lack of kinase selectivity that sorafenib exhibits, it is likely that inhibition of kinases other than RAF, or combinations of kinases, contributes to the cardiotoxic effects of sorafenib.
Collapse
|
150
|
Jourdain M, Bauchart JJ, Auffray JL, LeJemtel TH, Asseman P, Ennezat PV. Acute heart failure due to transient left ventricular dyssynchrony: case study. Am J Crit Care 2010; 19:e12-4. [PMID: 20436059 DOI: 10.4037/ajcc2010310] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
This case study describes an unusual cause of acute heart failure that resolved with early beta-blockade therapy. A 52-year-old woman who had acute heart failure with severe left ventricular systolic dysfunction and left bundle branch block was admitted to a university medical center. Contrast-enhanced magnetic resonance images of the heart did not show any evidence of myocardial infarction or myocarditis. Rate-related left bundle branch block and subsequent left ventricular dyssynchrony resulted in acute systolic dysfunction that resolved with beta-blockade therapy that allowed heart rate control and narrowing of the QRS complex. Of note, the use of inotropic agents would have dramatically worsened the cardiac condition.
Collapse
Affiliation(s)
- Matthieu Jourdain
- Matthieu Jourdain was a fellow in the intensive care unit, Jean Jacques Bauchart is a consultant in the intensive care unit and the cardiac catheterization laboratory, and Jean Luc Auffray, Philippe Asseman, and Pierre Vladimir Ennezat are consultants in the intensive care unit at the Cardiology Hospital of the Centre Hospitalier Régional et Universitaire in Lille, France. Thierry H. LeJemtel is a professor in the division of cardiology at Tulane University School of Medicine, New Orleans, Louisiana
| | - Jean Jacques Bauchart
- Matthieu Jourdain was a fellow in the intensive care unit, Jean Jacques Bauchart is a consultant in the intensive care unit and the cardiac catheterization laboratory, and Jean Luc Auffray, Philippe Asseman, and Pierre Vladimir Ennezat are consultants in the intensive care unit at the Cardiology Hospital of the Centre Hospitalier Régional et Universitaire in Lille, France. Thierry H. LeJemtel is a professor in the division of cardiology at Tulane University School of Medicine, New Orleans, Louisiana
| | - Jean Luc Auffray
- Matthieu Jourdain was a fellow in the intensive care unit, Jean Jacques Bauchart is a consultant in the intensive care unit and the cardiac catheterization laboratory, and Jean Luc Auffray, Philippe Asseman, and Pierre Vladimir Ennezat are consultants in the intensive care unit at the Cardiology Hospital of the Centre Hospitalier Régional et Universitaire in Lille, France. Thierry H. LeJemtel is a professor in the division of cardiology at Tulane University School of Medicine, New Orleans, Louisiana
| | - Thierry H. LeJemtel
- Matthieu Jourdain was a fellow in the intensive care unit, Jean Jacques Bauchart is a consultant in the intensive care unit and the cardiac catheterization laboratory, and Jean Luc Auffray, Philippe Asseman, and Pierre Vladimir Ennezat are consultants in the intensive care unit at the Cardiology Hospital of the Centre Hospitalier Régional et Universitaire in Lille, France. Thierry H. LeJemtel is a professor in the division of cardiology at Tulane University School of Medicine, New Orleans, Louisiana
| | - Philippe Asseman
- Matthieu Jourdain was a fellow in the intensive care unit, Jean Jacques Bauchart is a consultant in the intensive care unit and the cardiac catheterization laboratory, and Jean Luc Auffray, Philippe Asseman, and Pierre Vladimir Ennezat are consultants in the intensive care unit at the Cardiology Hospital of the Centre Hospitalier Régional et Universitaire in Lille, France. Thierry H. LeJemtel is a professor in the division of cardiology at Tulane University School of Medicine, New Orleans, Louisiana
| | - Pierre Vladimir Ennezat
- Matthieu Jourdain was a fellow in the intensive care unit, Jean Jacques Bauchart is a consultant in the intensive care unit and the cardiac catheterization laboratory, and Jean Luc Auffray, Philippe Asseman, and Pierre Vladimir Ennezat are consultants in the intensive care unit at the Cardiology Hospital of the Centre Hospitalier Régional et Universitaire in Lille, France. Thierry H. LeJemtel is a professor in the division of cardiology at Tulane University School of Medicine, New Orleans, Louisiana
| |
Collapse
|