101
|
Vávrová A, Popelová O, Stěrba M, Jirkovský E, Hašková P, Mertlíková-Kaiserová H, Geršl V, Simůnek T. In vivo and in vitro assessment of the role of glutathione antioxidant system in anthracycline-induced cardiotoxicity. Arch Toxicol 2010; 85:525-35. [PMID: 21046361 DOI: 10.1007/s00204-010-0615-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2010] [Accepted: 10/14/2010] [Indexed: 10/18/2022]
Abstract
The clinical usefulness of anthracycline antineoplastic drugs is limited by their cardiotoxicity. Its mechanisms have not been fully understood, although the induction of oxidative stress is widely believed to play the principal role. Glutathione is the dominant cellular antioxidant, while glutathione peroxidase (GPx) together with glutathione reductase (GR) constitutes the major enzymatic system protecting the cardiac cells from oxidative damage. Therefore, this study aimed to assess their roles in anthracycline cardiotoxicity. Ten-week intravenous administration of daunorubicin (DAU, 3 mg/kg weekly) to rabbits induced heart failure, which was evident from decreased left ventricular ejection fraction and release of cardiac troponins to circulation. However, no significant changes in either total or oxidized glutathione contents or GR activity were detected in left ventricular tissue of DAU-treated rabbits when compared with control animals. GPx activity in the cardiac tissue significantly increased. In H9c2 rat cardiac cells, 24-h DAU exposure (0.1-10 μM) induced significant dose-dependent toxicity. Cellular content of reduced glutathione was insignificantly decreased, oxidized glutathione and GR activity were unaffected, and GPx activity was significantly increased. Neither buthionine sulfoximine (BSO, glutathione biosynthesis inhibitor) nor 2-oxo-4-thiazolidine-carboxylic acid (OTC, glutathione biosynthetic precursor) had significant effects on DAU cytotoxicity. This contrasted with model oxidative injury induced by hydrogen peroxide, which cytotoxicity was increased by BSO and decreased by OTC. In conclusion, our results suggest that the dysfunction of glutathione antioxidant system does not play a causative role in anthracycline cardiotoxicity.
Collapse
Affiliation(s)
- Anna Vávrová
- Department of Biochemical Sciences, Charles University in Prague, Heyrovského, Hradec Králové, Czech Republic
| | | | | | | | | | | | | | | |
Collapse
|
102
|
Hasinoff BB, Patel D. The lack of target specificity of small molecule anticancer kinase inhibitors is correlated with their ability to damage myocytes in vitro. Toxicol Appl Pharmacol 2010; 249:132-9. [PMID: 20832415 DOI: 10.1016/j.taap.2010.08.026] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Revised: 08/24/2010] [Accepted: 08/26/2010] [Indexed: 10/19/2022]
Abstract
Many new targeted small molecule anticancer kinase inhibitors are actively being developed. However, the clinical use of some kinase inhibitors has been shown to result in cardiotoxicity. In most cases the mechanisms by which they exert their cardiotoxicity are not well understood. We have used large scale profiling data on 8 FDA-approved tyrosine kinase inhibitors and 10 other kinase inhibitors to a panel of 317 kinases in order to correlate binding constants and kinase inhibitor binding selectivity scores with kinase inhibitor-induced damage to neonatal rat cardiac myocytes. The 18 kinase inhibitors that were the subject of this study were: canertinib, dasatinib, dovitinib, erlotinib, flavopiridol, gefitinib, imatinib, lapatinib, midostaurin, motesanib, pazopanib, sorafenib, staurosporine, sunitinib, tandutinib, tozasertib, vandetanib and vatalanib. The combined tyrosine kinase and serine-threonine kinase selectivity scores were highly correlated with the myocyte-damaging effects of the kinase inhibitors. This result suggests that myocyte damage was due to a lack of target selectivity to binding of both tyrosine kinases and serine-threonine kinases, and was not due to binding to either group specifically. Finally, the strength of kinase inhibitor binding for 290 kinases was examined for correlations with myocyte damage. Kinase inhibitor binding was significantly correlated with myocyte damage for 12 kinases. Thus, myocyte damage may be multifactorial in nature with the inhibition of a number of kinases involved in producing kinase inhibitor-induced myocyte damage.
Collapse
Affiliation(s)
- Brian B Hasinoff
- Faculty of Pharmacy, Apotex Centre, University of Manitoba, 750 McDermot Avenue, Winnipeg, Manitoba R3E0T5, Canada.
| | | |
Collapse
|
103
|
Xiao L, Hu Z, Zhang W, Wu C, Yu H, Wang P. Evaluation of doxorubicin toxicity on cardiomyocytes using a dual functional extracellular biochip. Biosens Bioelectron 2010; 26:1493-9. [PMID: 20732805 DOI: 10.1016/j.bios.2010.07.093] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2010] [Revised: 07/16/2010] [Accepted: 07/23/2010] [Indexed: 11/20/2022]
Abstract
Nowadays, cardiotoxicity induced by clinical drugs presents a high prevalence and has aroused great attention onto the effective and reliable drug evaluation before clinical treatment. Doxorubicin (Adriamycin), as a type of anthracycline chemotherapy agent for cancer treatment, was restricted in the clinical use because of its cardiotoxicity. In the present work, a dual functional biochip ExCell integrated with microelectrode arrays and interdigitated electrodes was designed to study the electrophysiological function and physical state of cardiomyocytes under the treatment of doxorubicin. Extracellular field potentials and cell-substrate impedance were measured to respectively express these two functions simultaneously in the same culture. The result detected by ExCell presented a portrait of cardiotoxicity induced by doxorubicin. The amplitude of extracellular field potentials decreased to 93%, 82% and 50% at 50 min treatment of doxorubicin with concentrations of 20 μM, 100 μM and 200 μM, respectively. Successively, beating rate decrease, beat-to-beat variation and Ca(2+) flux manifested severe abnormality. The cell-substrate impedance declined continuously in the depressing process of electrophysiological function and cell death was induced in high concentration treatment. All these result indicate that the biochip ExCell has the potential to be a fast-response and subtle tool for high-throughput drug evaluation assays.
Collapse
Affiliation(s)
- Lidan Xiao
- Biosensor National Special Lab, Department of Biomedical Engineering, Zhejiang University, Hangzhou 310027, PR China
| | | | | | | | | | | |
Collapse
|
104
|
Fu Z, Guo J, Jing L, Li R, Zhang T, Peng S. Enhanced toxicity and ROS generation by doxorubicin in primary cultures of cardiomyocytes from neonatal metallothionein-I/II null mice. Toxicol In Vitro 2010; 24:1584-91. [PMID: 20600803 DOI: 10.1016/j.tiv.2010.06.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2010] [Revised: 06/02/2010] [Accepted: 06/17/2010] [Indexed: 11/30/2022]
Abstract
The clinical use of doxorubicin (Dox), a potent anticancer drug, is limited by its concurrent dose-dependent cardiotoxicity. We previously found that metallothionein-I/II (MT-I/II) null mice are more vulnerable to Dox-induced cardiomyopathy, but it is unknown whether depletion of MT would sensitize cardiomyocytes to Dox toxicity in vitro since the protective effect of MT still remains controversial. In the present study, a primary culture system of cardiomyocytes from neonatal MT-I/II null (MT(-/-)) and corresponding wild type (MT(+/+)) mice was established to unequivocally determine the effect of MT deficiency on Dox-induced toxicity. MT concentrations in the MT(-/-) cardiomyocytes were about 2.5-fold lower than those in MT(+/+) cardiomyocytes. MT(-/-) cardiomyocytes were more sensitive to Dox-induced cytotoxicity than MT(+/+) cardiomyocytes as measured by morphological alterations, lactate dehydrogenase leakage, cell viability, and apoptosis. Dox time- and concentration-dependently increased reactive oxygen species (ROS) formation in MT(+/+) cardiomyocytes, and this effect was exaggerated in MT(-/-) cardiomyocytes. Antioxidant N-acetylcysteine (NAC) and glutathione (GSH) significantly rescued MT(+/+) but not MT(-/-)cardiomyocytes from Dox-induced cell death and ROS generation. These findings suggest that basal MT provide protection against Dox-induced toxicity in cardiomyocytes, particularly highlight the important role of MT as a cellular antioxidant on scavenging ROS.
Collapse
Affiliation(s)
- Ze Fu
- Evaluation and Research Centre for Toxicology, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, Fengtai District, Beijing, PR China
| | | | | | | | | | | |
Collapse
|
105
|
Hasinoff BB. The cardiotoxicity and myocyte damage caused by small molecule anticancer tyrosine kinase inhibitors is correlated with lack of target specificity. Toxicol Appl Pharmacol 2010; 244:190-5. [DOI: 10.1016/j.taap.2009.12.032] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2009] [Revised: 12/14/2009] [Accepted: 12/22/2009] [Indexed: 10/20/2022]
|
106
|
Minotti G, Salvatorelli E, Menna P. Pharmacological foundations of cardio-oncology. J Pharmacol Exp Ther 2010; 334:2-8. [PMID: 20335321 DOI: 10.1124/jpet.110.165860] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Anthracyclines and many other antitumor drugs induce cardiotoxicity that occurs "on treatment" or long after completing chemotherapy. Dose reductions limit the incidence of early cardiac events but not that of delayed sequelae, possibly indicating that any dose level of antitumor drugs would prime the heart to damage from sequential stressors. Drugs targeted at tumor-specific moieties raised hope for improving the cardiovascular safety of antitumor therapies; unfortunately, however, many such drugs proved unable to spare the heart, aggravated cardiotoxicity induced by anthracyclines, or were safe in selected patients of clinical trials but not in the general population. Cardio-oncology is the discipline aimed at monitoring the cardiovascular safety of antitumor therapies. Although popularly perceived as a clinical discipline that brings oncologists and cardiologists working together, cardio-oncology is in fact a pharmacology-oriented translational discipline. The cardiovascular performance of survivors of cancer will only improve if clinicians joined pharmacologists in the search for new predictive models of cardiotoxicity or mechanistic approaches to explain how a given drug might switch from causing systolic failure to inducing ischemia. The lifetime risk of cardiotoxicity from antitumor drugs needs to be reconciled with the identification of long-lasting pharmacological signatures that overlap with comorbidities. Research on targeted drugs should be reshaped to appreciate that the terminal ballistics of new "magic bullets" might involve cardiomyocytes as innocent bystanders. Finally, the concepts of prevention and treatment need to be tailored to the notion that late-onset cardiotoxicity builds on early asymptomatic cardiotoxicity. The heart of cardio-oncology rests with such pharmacological foundations.
Collapse
Affiliation(s)
- Giorgio Minotti
- Center for Integrated Research, Drug Sciences, University Campus Bio-Medico, Rome, Italy.
| | | | | |
Collapse
|
107
|
Influence of the phosphodiesterase-5 inhibitor, sildenafil, on sensitivity to chemotherapy in breast tumor cells. Breast Cancer Res Treat 2010; 124:349-60. [DOI: 10.1007/s10549-010-0765-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2009] [Accepted: 01/20/2010] [Indexed: 12/22/2022]
|
108
|
Raschi E, Vasina V, Ursino MG, Boriani G, Martoni A, De Ponti F. Anticancer drugs and cardiotoxicity: Insights and perspectives in the era of targeted therapy. Pharmacol Ther 2010; 125:196-218. [DOI: 10.1016/j.pharmthera.2009.10.002] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2009] [Accepted: 10/05/2009] [Indexed: 01/01/2023]
|
109
|
Qian JY, Guo L. Altered cytosolic Ca2+ dynamics in cultured Guinea pig cardiomyocytes as an in vitro model to identify potential cardiotoxicants. Toxicol In Vitro 2010; 24:960-72. [PMID: 20064605 DOI: 10.1016/j.tiv.2009.12.027] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2009] [Revised: 12/07/2009] [Accepted: 12/29/2009] [Indexed: 01/07/2023]
Abstract
Altered intracellular calcium (Ca(i)(2+)) handling by cardiomyocytes has been implicated in drug-induced cardiomyopathy and arrhythmogenesis. To explore whether such alterations predict cardiotoxicity, Ca(i)(2+) imaging was conducted in cultured, spontaneously contracting Guinea pig cardiomyocytes to characterize the effects of 13 cardiotoxicants and 2 safe drugs. All cardiotoxicants perturbed Ca(i)(2+) at therapeutically relevant concentrations. The cytotoxic chemotherapeutics doxorubicin and epirubicin, known to cause cardiomyopathy, preferentially reduced Ca(i)(2+) transient amplitude and sarcoplasmic reticulum (SR) Ca(2+) content, whereas Torsade de Pointes (TdP) inducers and potent hERG channel blockers (amiodarone, cisapride, dofetilide, E-4031 and terfenadine) predominately suppressed diastolic Ca(i)(2+) and contraction rate, and prolonged Ca(i)(2+) transient duration. The molecularly targeted cancer therapeutics, sunitinib and imatinib, exhibited profound effects on Ca(i)(2+), combining effects of cytotoxic chemotherapeutics, TdP inducers and potent hERG channel blockers. TdP inducers lacking direct hERG inhibition, ouabain and pentamidine, significantly elevated Ca(i)(2+) transient amplitude and SR Ca(2+) content while aconitine primarily accelerated automaticity and elevated diastolic Ca(i)(2+) similar to ouabain. Finally, amoxicillin and aspirin did not exert any significant effects on Ca(i)(2+) at concentrations up to 100 microM. These results suggest that detecting altered Ca(i)(2+) handling in cultured cardiomyocytes may be used as an in vitro model for early cardiac drug safety assessment.
Collapse
Affiliation(s)
- Jian-Yong Qian
- Safety Pharmacology, Department of Toxicology and Pathology, Non-Clinical Safety, Hoffmann-La Roche, Inc, Nutley, NJ 07110, USA
| | | |
Collapse
|
110
|
Maita R, Strauss M, Anselmi G. Skeletal Muscle for Endomyocardial Biopsy: Comparable Stress Response in Doxorubicin Cardio-myopathy. J Toxicol Pathol 2009; 22:273-9. [PMID: 22272002 PMCID: PMC3234599 DOI: 10.1293/tox.22.273] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2009] [Accepted: 07/17/2009] [Indexed: 11/28/2022] Open
Abstract
In the present study, we compared the cell damage response in skeletal and
cardiac muscle tissue when exposed to doxorubicin. This was carried out by means
of a less invasive informative substitute to endomyocardiac biopsy based on
Hsp70 immunodetection and a subcellular analysis of the nucleolus. Male Sprague
Dawley rats (62 g body weight) were randomly distributed into 3 group, the
control and doxorubicin I and doxorubicin II groups, in which 15 and 25 mg/kg
body weight of doxorubicin (0.1 ml, i.v.) was administered, respectively. After
15, 30, 45 and 60 minutes, portions of the left and right ventricle wall and
interventricle wall, together with skeletal muscle from the posterior and
anterior member, were prepared for Hsp70 immunodetection by Western blot
analysis and ultrastructural study using the thin cut technique. Differential
cell response between the control and treated groups was observed in Hsp70
immunodetection and at the subcellular level. In the control group, the Hsp70
recognition levels and typical normal nucleolar morphology were similar, while
the treated groups showed variable-dependent Hsp70 recognition and segregation
of nucleolar components, forming ring-like figures of a variable-independent
nature. Comparison of cardiac and skeletal muscle tissue cell response to
doxorubicin toxic aggression revealed parallelism in terms of Hsp70 accumulation
in certain regions of both tissues (15 mg/kg body weight of doxorubicin), which
suggests that replacing endomyocardiac biopsy analysis with skeletal muscle
analysis may be a safe option.
Collapse
Affiliation(s)
- Rosa Maita
- Sección de Biología Celular, Instituto de Medicina Tropical, Facultad de Medicina, Universidad Central de Venezuela, Caracas, Venezuela
| | | | | |
Collapse
|
111
|
Berdichevski A, Meiry G, Milman F, Reiter I, Sedan O, Eliyahu S, Duffy HS, Youdim MB, Binah O. TVP1022 protects neonatal rat ventricular myocytes against doxorubicin-induced functional derangements. J Pharmacol Exp Ther 2009; 332:413-20. [PMID: 19915070 DOI: 10.1124/jpet.109.161158] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Our recent studies demonstrated that propargylamine derivatives such as rasagiline (Azilect, Food and Drug Administration-approved anti-Parkinson drug) and its S-isomer TVP1022 protect cardiac and neuronal cell cultures against apoptotic-inducing stimuli. Studies on structure-activity relationship revealed that their neuroprotective effect is associated with the propargylamine moiety, which protects mitochondrial viability and prevents apoptosis by activating Bcl-2 and protein kinase C-epsilon and by down-regulating the proapoptotic protein Bax. Based on the established cytoprotective and neuroprotective efficacies of propargylamine derivatives, as well as on our recent study showing that TVP1022 attenuates serum starvation-induced and doxorubicin-induced apoptosis in neonatal rat ventricular myocytes (NRVMs), we tested the hypothesis that TVP1022 will also provide protection against doxorubicin-induced NRVM functional derangements. The present study demonstrates that pretreatment of NRVMs with TVP1022 (1 microM, 24 h) prevented doxorubicin (0.5 microM, 24 h)-induced elevation of diastolic [Ca(2+)](i), the slowing of [Ca(2+)](i) relaxation kinetics, and the decrease in the rates of myocyte contraction and relaxation. Furthermore, pretreatment with TVP1022 attenuated the doxorubicin-induced reduction in the protein expression of sarco/endoplasmic reticulum calcium (Ca(2+)) ATPase, Na(+)/Ca(2+) exchanger 1, and total connexin 43. Finally, TVP1022 diminished the inhibitory effect of doxorubicin on gap junctional intercellular coupling (measured by means of Lucifer yellow transfer) and on conduction velocity, the amplitude of the activation phase, and the maximal rate of activation (dv/dt(max)) measured by the Micro-Electrode-Array system. In summary, our results indicate that TVP1022 acts as a novel cardioprotective agent against anthracycline cardiotoxicity, and therefore potentially can be coadmhence, theinistered with doxorubicin in the treatment of malignancies in humans.
Collapse
Affiliation(s)
- Alexandra Berdichevski
- Department of Physiology, Ruth and Bruce Rappaport Faculty of Medicine, Haifa 31096, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
112
|
Moss LS, Starbuck MF, Mayer DK, Harwood EB, Glotzer J. Trastuzumab-Induced Cardiotoxicity. Oncol Nurs Forum 2009; 36:676-85. [DOI: 10.1188/09.onf.676-685] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
113
|
Spallarossa P, Altieri P, Pronzato P, Aloi C, Ghigliotti G, Barsotti A, Brunelli C. Sublethal doses of an anti-erbB2 antibody leads to death by apoptosis in cardiomyocytes sensitized by low prosenescent doses of epirubicin: the protective role of dexrazoxane. J Pharmacol Exp Ther 2009; 332:87-96. [PMID: 19841470 DOI: 10.1124/jpet.109.159525] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The cardiotoxic synergism resulting from the sequential treatment with anthracyclines and trastuzumab has been attributed to the trastuzumab-induced loss of the erbB2-related functions that serve as a salvage pathway against the damaging effects of anthracyclines. Cellular senescence is a novel mechanism of cardiotoxicity induced by subapoptotic doses of anthracyclines. After having identified prosenescent and proapoptotic doses of epirubicin and rat MAb c-erbB2/Her-2/neu Ab-9 clone B10 (B10), an anti-erbB2 monoclonal antibody, we investigated the effects of the sequential treatment with prosenescent doses of both drugs on H9c2 cells and neonatal rat cardiomyocytes pretreated with or without the cardioprotective agent dexrazoxane. Cells were analyzed by senescence-associated beta-galactosidase, single-stranded DNA, annexin/propidium double staining, F-actin, and mitochondrial transmembrane potential. ErbB2 expression levels, AKT activation, and the effects of the inhibition of nicotinamide adenine dinucleotide phosphate oxidase [NAD(P)H oxidase] and phosphoinositide-3-OH kinase (PI3K) were also assessed. Data demonstrate that 1) the toxic effects of epirubicin mainly occur through NAD(P)H oxidase activation; 2) the erbB2 overexpression induced by epirubicin is a redox-sensitive mechanism largely dependent on NAD(P)H oxidase; 3) the loss of erbB2-related functions caused by B10 determines marginal cellular changes in untreated cells, but causes massive death by apoptosis in cells previously exposed to a prosenescent dose of epirubicin, 4) dexrazoxane promotes survival pathways, as demonstrated by the activation of Akt and the PI3K-dependent erbB2 overexpression; and 5) it also prevents epirubicin-induced senescence and renders epirubicin-treated cells more resistant to treatment with B10. Data underline the importance of NAD(P)H oxidase in epirubicin-induced cardiotoxicity and shed new light on the protective mechanisms of dexrazoxane.
Collapse
Affiliation(s)
- Paolo Spallarossa
- Research Center of Cardiovascular Biology, Division of Cardiology, Department of Internal Medicine, University of Genova, Genova, Italy
| | | | | | | | | | | | | |
Collapse
|
114
|
Tong TR. Therapies for coronaviruses. Part I of II -- viral entry inhibitors. Expert Opin Ther Pat 2009; 19:357-67. [PMID: 19449500 DOI: 10.1517/13543770802609384] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Severe acute respiratory syndrome (SARS) coronavirus emerged fleetingly in the winter of 2002 and again in the winter of 2003, resulting in the infection of ~8,000 people and the death of ~800. The identification of the putative natural reservoir suggests that a re-emergence is possible. The functions of many coronaviral proteins have now been elucidated, resulting in many novel approaches to therapy. OBJECTIVE To review anticoronaviral therapies based on inhibition of viral entry into the host cell and to cast light on promising approaches and future developments. METHOD The published literature, in particular patent publications, is searched for relevant documents. The information is organized and critiqued. RESULTS/CONCLUSION The approaches to combating coronaviral infections are built on the foundation of antivirals against other viruses and the fundamental insights gained by dissection of the coronaviral lifecycle. These approaches include the prevention of viral entry, reviewed here, and interference with the intracellular lifecycle of the virus in the infected cell, reviewed next. Of the viral-entry inhibitors, monoclonal antibodies have demonstrated efficacy, clinical application in other viral infections, and the potential to impact a future epidemic. Moreover, combinations of monoclonal antibodies have been shown to have a broader spectrum of antiviral activity.
Collapse
Affiliation(s)
- Tommy R Tong
- Jack D Weiler Hospital, Montefiore Medical Center, Department of Pathology, Bronx, NY 10461, USA.
| |
Collapse
|
115
|
Simůnek T, Stérba M, Popelová O, Adamcová M, Hrdina R, Gersl V. Anthracycline-induced cardiotoxicity: overview of studies examining the roles of oxidative stress and free cellular iron. Pharmacol Rep 2009; 61:154-71. [PMID: 19307704 DOI: 10.1016/s1734-1140(09)70018-0] [Citation(s) in RCA: 546] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2008] [Revised: 02/02/2009] [Indexed: 12/23/2022]
Abstract
The risk of cardiotoxicity is the most serious drawback to the clinical usefulness of anthracycline antineoplastic antibiotics, which include doxorubicin (adriamycin), daunorubicin or epirubicin. Nevertheless, these compounds remain among the most widely used anticancer drugs. The molecular pathogenesis of anthracycline cardiotoxicity remains highly controversial, although the oxidative stress-based hypothesis involving intramyocardial production of reactive oxygen species (ROS) has gained the widest acceptance. Anthracyclines may promote the formation of ROS through redox cycling of their aglycones as well as their anthracycline-iron complexes. This proposed mechanism has become particularly popular in light of the high cardioprotective efficacy of dexrazoxane (ICRF-187). The mechanism of action of this drug has been attributed to its hydrolytic transformation into the iron-chelating metabolite ADR-925, which may act by displacing iron from anthracycline-iron complexes or by chelating free or loosely bound cellular iron, thus preventing site-specific iron-catalyzed ROS damage. However, during the last decade, calls for the critical reassessment of this "ROS and iron" hypothesis have emerged. Numerous antioxidants, although efficient in cellular or acute animal experiments, have failed to alleviate anthracycline cardiotoxicity in clinically relevant chronic animal models or clinical trials. In addition, studies with chelators that are stronger and more selective for iron than ADR-925 have also yielded negative or, at best, mixed outcomes. Hence, several lines of evidence suggest that mechanisms other than the traditionally emphasized "ROS and iron" hypothesis are involved in anthracycline-induced cardiotoxicity and that these alternative mechanisms may be better bases for designing approaches to achieve efficient and safe cardioprotection.
Collapse
Affiliation(s)
- Tomás Simůnek
- Charles University in Prague, Faculty of Pharmacy in Hradec Králové, Hradec Králové, Czech Republic.
| | | | | | | | | | | |
Collapse
|
116
|
Abstract
Recent molecular studies have expanded the biological contexts in which topoisomerase II (TOP2) has crucial functions, including DNA replication, transcription and chromosome segregation. Although the biological functions of TOP2 are important for ensuring genomic integrity, the ability to interfere with TOP2 and generate enzyme-mediated DNA damage is an effective strategy for cancer chemotherapy. The molecular tools that have allowed an understanding of the biological functions of TOP2 are also being applied to understanding the details of drug action. These studies promise refined targeting of TOP2 as an effective anticancer strategy.
Collapse
Affiliation(s)
- John L Nitiss
- Molecular Pharmacology Department, St Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
117
|
Salvatorelli E, Menna P, Lusini M, Covino E, Minotti G. Doxorubicinolone Formation and Efflux: A Salvage Pathway against Epirubicin Accumulation in Human Heart. J Pharmacol Exp Ther 2009; 329:175-84. [DOI: 10.1124/jpet.108.149260] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
118
|
Hasinoff BB, Patel D, O'Hara KA. Mechanisms of myocyte cytotoxicity induced by the multiple receptor tyrosine kinase inhibitor sunitinib. Mol Pharmacol 2008; 74:1722-8. [PMID: 18815214 DOI: 10.1124/mol.108.050104] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The anticancer tyrosine kinase inhibitor sunitinib has been shown recently to be cardiotoxic. Using a neonatal rat myocyte model, we investigated various mechanisms that might be responsible for its cardiotoxicity. Sunitinib potently inhibited the enzyme activity of both AMP-activated protein kinase (AMPK) and the ribosomal S6 kinase RSK1 at therapeutically relevant concentrations. Heart tissue with its high energy needs might be particularly sensitive to inhibition of AMPK because of its role as an energy sensor regulating ATP levels. As measured by lactate dehydrogenase release, sunitinib treatment of myocytes caused dose-dependent damage at therapeutic levels. Sunitinib treatment also caused a dose-dependent reduction in myocyte protein levels of the phosphorylated alpha and beta isoforms of the AMPK phosphorylation target acetyl-Coenzyme A carboxylase. However, myocytes were not protected from sunitinib treatment by pretreating them with the AMPK-activating antidiabetic drug metformin. Sunitinib treatment of myocytes also did not affect cellular ATP levels. Together, these last two results do not suggest a major role for inhibition of AMPK in sunitinib-induced myocyte damage. Dexrazoxane, which is a clinically approved doxorubicin cardioprotective agent, also did not protect myocytes from damage, which suggests that sunitinib did not induce oxidative damage. In conclusion, even though sunitinib potently inhibits AMPK and RSK1, given the extreme lack of kinase selectivity that sunitinib exhibits, it is likely that inhibition of other kinases or combinations of kinases are responsible for the cardiotoxic effects of sunitinib.
Collapse
Affiliation(s)
- Brian B Hasinoff
- Faculty of Pharmacy, Apotex Centre, University of Manitoba, Winnipeg, Manitoba, Canada.
| | | | | |
Collapse
|
119
|
Fang C, Gu J, Xie F, Behr M, Yang W, Abel ED, Ding X. Deletion of the NADPH-cytochrome P450 reductase gene in cardiomyocytes does not protect mice against doxorubicin-mediated acute cardiac toxicity. Drug Metab Dispos 2008; 36:1722-8. [PMID: 18463197 DOI: 10.1124/dmd.108.021881] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
A genetic mouse model (designated cardiomyocyte-Cpr-null) with cardiomyocyte-specific deletion of the cytochrome P450 (P450) reductase (Cpr) gene was generated in this study. CPR protein levels, as well the enzyme activity of P450s, were greatly reduced in heart microsomes from the null mice compared with wild-type mice, whereas CPR expression in other organs remained unchanged. Nonetheless, homozygous null mice were normal in appearance, gross anatomy, tissue morphology, and general cardiac functional parameters, and there was no indication of embryonic lethality or premature mortality in contrast to the recognized role of CPR in embryonic development. Thus, this new mouse model should be useful for determination of the in vivo roles of cardiomyocyte CPR and CPR-dependent enzymes, including microsomal P450s, not only in the metabolism and toxicity of numerous xenobiotic compounds but also in cardiac pathophysiology. As a first application, we studied the role of cardiomyocyte CPR and CPR-dependent enzymes in doxorubicin (Dox)-mediated acute cardiotoxicity. Wild-type and null mice were treated with a single i.p. dose of Dox at 5, 10, or 20 mg/kg. The Dox treatment caused apoptosis and vacuolization in cardiomyocytes at the dose of 20 mg/kg and a significant increase in the levels of serum creatine kinase at 10 and 20 mg/kg in both wild-type and null mice. However, there was no significant difference in the extent of Dox-induced cardiac injury between the two strains; this lack of difference suggests that cardiomyocyte CPR and CPR-dependent enzymes do not play critical roles in the acute cardiotoxicity induced by Dox.
Collapse
Affiliation(s)
- Cheng Fang
- Wadsworth Center, New York State Department of Health, Empire State Plaza, Albany, NY, USA
| | | | | | | | | | | | | |
Collapse
|
120
|
Jung R, Wendeler MW, Danevad M, Himmelbauer H, Geßner R. Phylogenetic origin of LI-cadherin revealed by protein and gene structure analysis. Cell Mol Life Sci 2004; 61:1157-66. [PMID: 15141301 PMCID: PMC11138757 DOI: 10.1007/s00018-004-3470-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The intestine specific LI-cadherin differs in its overall structure from classical and desmosomal cadherins by the presence of seven instead of five cadherin repeats and a short cytoplasmic domain. Despite the low sequence similarity, a comparative protein structure analysis revealed that LI-cadherin may have originated from a five-repeat predecessor cadherin by a duplication of the first two aminoterminal repeats. To test this hypothesis, we cloned the murine LI-cadherin gene and compared its structure to that of other cadherins. The intron-exon organization, including the intron positions and phases, is perfectly conserved between repeats 3-7 of LI-cadherin and 1-5 of classical cadherins. Moreover, the genomic structure of the repeats 1-2 and 3-4 is identical for LI-cadherin and highly similar to that of the repeats 1-2 of classical cadherins. These findings strengthen our assumption that LI-cadherin originated from an ancestral cadherin with five domains by a partial gene duplication event.
Collapse
Affiliation(s)
- R. Jung
- Institute of Laboratory Medicine and Biochemistry, Virchow-Hospital of Charité Medical School, Humboldt University of Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Schering AG, Müllerstr. 178, 13342 Berlin, Germany
| | - M. W. Wendeler
- Institute of Laboratory Medicine and Biochemistry, Virchow-Hospital of Charité Medical School, Humboldt University of Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - M. Danevad
- Institute of Laboratory Medicine and Biochemistry, Virchow-Hospital of Charité Medical School, Humboldt University of Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - H. Himmelbauer
- Max-Planck-Institute of Molecular Genetics, Ihnestr. 73, 14195 Berlin, Germany
| | - R. Geßner
- Institute of Laboratory Medicine and Biochemistry, Virchow-Hospital of Charité Medical School, Humboldt University of Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| |
Collapse
|