1
|
A glutathione-responsive polyphenol - Constructed nanodevice for double roles in apoptosis and ferroptosis. Colloids Surf B Biointerfaces 2021; 205:111902. [PMID: 34107442 DOI: 10.1016/j.colsurfb.2021.111902] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/24/2021] [Accepted: 06/01/2021] [Indexed: 12/12/2022]
Abstract
Combination chemotherapy regimens have been put forward to achieve a synergistic effect and reduce drug doses for the clinical applications of cancer treatment. One of the principal approaches for killing cancer cells involves triggering apoptotic cell death with anti-cancer drugs. Nevertheless, the efficacy of apoptosis induction in tumors is often restricted on account of intrinsic or acquired resistance of cancer cells to apoptosis. Ferroptosis, which involves reactive oxygen species (ROS), is another way to regulate cell death. Doxorubicin (DOX), a commonly used chemotherapeutic agent, can enter the nucleus and destroy tumor cells while also affecting mitochondria by producing semiquinone radicals. Therefore, a drug system combining ferroptosis and apoptosis, bridged by DOX-induced ROS, was proposed to be designed. Herein, we employed a facile and effective self-assembly method to prepare DOX-loaded nanocomplexes by DOX, Pluronic F-68, tannic acid (TA), and iron ions. TA and iron ions could not only improve the stability of nanocarrier but also facilitate achieving a ferroptotic effect. As a result, DOX@F-68/TA/Fe3+ nanocomplexes showed a strong pro-apoptotic effect as well as an increase in intracellular oxidative stress. The improved oxidative stress further resulted in the ferroptosis of tumor cells. In vivo experiments demonstrated that DOX@F-68/TA/Fe3+ efficiently targeted the tumor following intravenous injection and successfully inhibited tumor development.
Collapse
|
2
|
Martinovich GG, Martinovich IV, Vcherashniaya AV, Zenkov NK, Menshchikova EB, Cherenkevich SN. Chemosensitization of Tumor Cells by Phenolic Antioxidants: The Role of the Nrf2 Transcription Factor. Biophysics (Nagoya-shi) 2020. [DOI: 10.1134/s000635092006010x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
3
|
Modulatory Effects of Dexrazoxane Against Genotoxicity and Lipid Peroxidation Induced by Idarubicin in HepG2 Cells. INTERNATIONAL JOURNAL OF CANCER MANAGEMENT 2018. [DOI: 10.5812/ijcm.9675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
4
|
Hasinoff BB, Wu X, Patel D, Kanagasabai R, Karmahapatra S, Yalowich JC. Mechanisms of Action and Reduced Cardiotoxicity of Pixantrone; a Topoisomerase II Targeting Agent with Cellular Selectivity for the Topoisomerase IIα Isoform. J Pharmacol Exp Ther 2016; 356:397-409. [PMID: 26660439 PMCID: PMC4746493 DOI: 10.1124/jpet.115.228650] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 12/09/2015] [Indexed: 01/09/2023] Open
Abstract
Pixantrone is a new noncardiotoxic aza-anthracenedione anticancer drug structurally related to anthracyclines and anthracenediones, such as doxorubicin and mitoxantrone. Pixantrone is approved in the European Union for the treatment of relapsed or refractory aggressive B cell non-Hodgkin lymphoma. This study was undertaken to investigate both the mechanism(s) of its anticancer activity and its relative lack of cardiotoxicity. Pixantrone targeted DNA topoisomerase IIα as evidenced by its ability to inhibit kinetoplast DNA decatenation; to produce linear double-strand DNA in a pBR322 DNA cleavage assay; to produce DNA double-strand breaks in a cellular phospho-histone γH2AX assay; to form covalent topoisomerase II-DNA complexes in a cellular immunodetection of complex of enzyme-to-DNA assay; and to display cross-resistance in etoposide-resistant K562 cells. Pixantrone produced semiquinone free radicals in an enzymatic reducing system, although not in a cellular system, most likely due to low cellular uptake. Pixantrone was 10- to 12-fold less damaging to neonatal rat myocytes than doxorubicin or mitoxantrone, as measured by lactate dehydrogenase release. Three factors potentially contribute to the reduced cardiotoxicity of pixantrone. First, its lack of binding to iron(III) makes it unable to induce iron-based oxidative stress. Second, its low cellular uptake may limit its ability to produce semiquinone free radicals and redox cycle. Finally, because the β isoform of topoisomerase II predominates in postmitotic cardiomyocytes, and pixantrone is demonstrated in this study to be selective for topoisomerase IIα in stabilizing enzyme-DNA covalent complexes, the attenuated cardiotoxicity of this agent may also be due to its selectivity for targeting topoisomerase IIα over topoisomerase IIβ.
Collapse
Affiliation(s)
- Brian B Hasinoff
- College of Pharmacy, Apotex Centre, University of Manitoba, Winnipeg, Manitoba, Canada (B.B.H., X.W., D.P.); and Division of Pharmacology, College of Pharmacy, Ohio State University, Columbus, Ohio (R.K., S.K., J.C.Y.)
| | - Xing Wu
- College of Pharmacy, Apotex Centre, University of Manitoba, Winnipeg, Manitoba, Canada (B.B.H., X.W., D.P.); and Division of Pharmacology, College of Pharmacy, Ohio State University, Columbus, Ohio (R.K., S.K., J.C.Y.)
| | - Daywin Patel
- College of Pharmacy, Apotex Centre, University of Manitoba, Winnipeg, Manitoba, Canada (B.B.H., X.W., D.P.); and Division of Pharmacology, College of Pharmacy, Ohio State University, Columbus, Ohio (R.K., S.K., J.C.Y.)
| | - Ragu Kanagasabai
- College of Pharmacy, Apotex Centre, University of Manitoba, Winnipeg, Manitoba, Canada (B.B.H., X.W., D.P.); and Division of Pharmacology, College of Pharmacy, Ohio State University, Columbus, Ohio (R.K., S.K., J.C.Y.)
| | - Soumendrakrishna Karmahapatra
- College of Pharmacy, Apotex Centre, University of Manitoba, Winnipeg, Manitoba, Canada (B.B.H., X.W., D.P.); and Division of Pharmacology, College of Pharmacy, Ohio State University, Columbus, Ohio (R.K., S.K., J.C.Y.)
| | - Jack C Yalowich
- College of Pharmacy, Apotex Centre, University of Manitoba, Winnipeg, Manitoba, Canada (B.B.H., X.W., D.P.); and Division of Pharmacology, College of Pharmacy, Ohio State University, Columbus, Ohio (R.K., S.K., J.C.Y.)
| |
Collapse
|
5
|
Li N, Oquendo E, Capaldi RA, Robinson JP, He YD, Hamadeh HK, Afshari CA, Lightfoot-Dunn R, Narayanan PK. A systematic assessment of mitochondrial function identified novel signatures for drug-induced mitochondrial disruption in cells. Toxicol Sci 2014; 142:261-73. [PMID: 25163676 DOI: 10.1093/toxsci/kfu176] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Mitochondrial perturbation has been recognized as a contributing factor to various drug-induced organ toxicities. To address this issue, we developed a high-throughput flow cytometry-based mitochondrial signaling assay to systematically investigate mitochondrial/cellular parameters known to be directly impacted by mitochondrial dysfunction: mitochondrial membrane potential (MMP), mitochondrial reactive oxygen species (ROS), intracellular reduced glutathione (GSH) level, and cell viability. Modulation of these parameters by a training set of compounds, comprised of established mitochondrial poisons and 60 marketed drugs (30 nM to 1mM), was tested in HL-60 cells (a human pro-myelocytic leukemia cell line) cultured in either glucose-supplemented (GSM) or glucose-free (containing galactose/glutamine; GFM) RPMI-1640 media. Post-hoc bio-informatic analyses of IC50 or EC50 values for all parameters tested revealed that MMP depolarization in HL-60 cells cultured in GSM was the most reliable parameter for determining mitochondrial dysfunction in these cells. Disruptors of mitochondrial function depolarized MMP at concentrations lower than those that caused loss of cell viability, especially in cells cultured in GSM; cellular GSH levels correlated more closely to loss of viability in vitro. Some mitochondrial respiratory chain inhibitors increased mitochondrial ROS generation; however, measuring an increase in ROS alone was not sufficient to identify mitochondrial disruptors. Furthermore, hierarchical cluster analysis of all measured parameters provided confirmation that MMP depletion, without loss of cell viability, was the key signature for identifying mitochondrial disruptors. Subsequent classification of compounds based on ratios of IC50s of cell viability:MMP determined that this parameter is the most critical indicator of mitochondrial health in cells and provides a powerful tool to predict whether novel small molecule entities possess this liability.
Collapse
Affiliation(s)
- Nianyu Li
- Department of Comparative Biology and Safety Sciences, Amgen, Amgen Court West 1201, Seattle, Washington 98119
| | | | | | - J Paul Robinson
- Purdue University Cytometry Laboratories, Department of Basic Medical Sciences, Purdue University, West Lafayette, Indiana 47907
| | - Yudong D He
- Department of Comparative Biology and Safety Sciences, Amgen, Amgen Court West 1201, Seattle, Washington 98119
| | - Hisham K Hamadeh
- Department of Comparative Biology and Safety Sciences, Amgen, 1 Amgen Center Dr, Thousand Oaks, California 91320-1799
| | - Cynthia A Afshari
- Department of Comparative Biology and Safety Sciences, Amgen, 1 Amgen Center Dr, Thousand Oaks, California 91320-1799
| | - Ruth Lightfoot-Dunn
- Department of Comparative Biology and Safety Sciences, Amgen, 1 Amgen Center Dr, Thousand Oaks, California 91320-1799
| | - Padma Kumar Narayanan
- Department of Comparative Biology and Safety Sciences, Amgen, Amgen Court West 1201, Seattle, Washington 98119
| |
Collapse
|
6
|
Doroshow JH. Dexrazoxane for the prevention of cardiac toxicity and treatment of extravasation injury from the anthracycline antibiotics. Curr Pharm Biotechnol 2013; 13:1949-56. [PMID: 22352729 DOI: 10.2174/138920112802273245] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Revised: 02/05/2011] [Accepted: 05/04/2011] [Indexed: 12/11/2022]
Abstract
The cumulative cardiac toxicity of the anthracycline antibiotics and their propensity to produce severe tissue injury following extravasation from a peripheral vein during intravenous administration remain significant problems in clinical oncologic practice. Understanding of the free radical metabolism of these drugs and their interactions with iron proteins led to the development of dexrazoxane, an analogue of EDTA with intrinsic antineoplastic activity as well as strong iron binding properties, as both a prospective cardioprotective therapy for patients receiving anthracyclines and as an effective treatment for anthracycline extravasations. In this review, the molecular mechanisms by which the anthracyclines generate reactive oxygen species and interact with intracellular iron are examined to understand the cardioprotective mechanism of action of dexrazoxane and its ability to protect the subcutaneous tissues from anthracycline-induced tissue necrosis.
Collapse
Affiliation(s)
- James H Doroshow
- Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.
| |
Collapse
|
7
|
Albini A, Cesana E, Donatelli F, Cammarota R, Bucci EO, Baravelli M, Anzà C, Noonan DM. Cardio-oncology in targeting the HER receptor family: the puzzle of different cardiotoxicities of HER2 inhibitors. Future Cardiol 2012; 7:693-704. [PMID: 21929348 DOI: 10.2217/fca.11.54] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The HER family of tyrosine kinase receptors includes several members that are clinically important targets in cancer therapies, in particular HER1 (the EGF receptor) and HER2, other members include HER3 and HER4. Trastuzumab, a humanized monoclonal antibody and lapatinib, a tyrosine kinase inhibitor, are drugs that target HER2, which is highly expressed in 20-30% of breast cancers. Trastuzumab is recommended as an adjuvant therapy for lymph node positive, HER2-positive breast cancers, or node-negative cancer with high-risk of recurrence, as well as in stage IV cancers. One serious side effect of trastuzumab is cardiomyocyte dysfunction, resulting in reduced heart contractile efficiency. The incidence of collateral effects on the heart with trastuzumab therapy increases in people with cardiovascular risk factors, heart disease and when combined with other chemotherapeutics. When cardiotoxicity was observed with trastuzumab, several studies have addressed potential cardiac damage of trastuzumab itself and lapatinib. The differences in cardiovascular effects of these two compounds are somewhat unexpected and suggest distinct mechanisms of action, which have clear implications in clinical application and prevention of cardiotoxicity in cardio-oncological approaches.
Collapse
Affiliation(s)
- Adriana Albini
- Oncology Research Division, Polo Scientifico e Tecnologico-IRCCS MultiMedica, Via Fantoli 16/15-20138, Milan, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Wu EC, Andrew JS, Buyanin A, Kinsella JM, Sailor MJ. Suitability of porous silicon microparticles for the long-term delivery of redox-active therapeutics. Chem Commun (Camb) 2011; 47:5699-701. [PMID: 21503283 DOI: 10.1039/c1cc10993f] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Thermal oxidation of porous Si microparticles provides an inert carrier for the long-term release of the anthracycline drug daunorubicin. Without prior oxidation, porous Si undergoes an undesirable side reaction with this redox active drug.
Collapse
Affiliation(s)
- Elizabeth C Wu
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | | | | | | | | |
Collapse
|
9
|
Wu EC, Andrew JS, Cheng L, Freeman WR, Pearson L, Sailor MJ. Real-time monitoring of sustained drug release using the optical properties of porous silicon photonic crystal particles. Biomaterials 2010; 32:1957-66. [PMID: 21122914 DOI: 10.1016/j.biomaterials.2010.11.013] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Accepted: 11/06/2010] [Indexed: 10/18/2022]
Abstract
A controlled and observable drug delivery system that enables long-term local drug administration is reported. Biodegradable and biocompatible drug-loaded porous Si microparticles were prepared from silicon wafers, resulting in a porous 1-dimensional photonic crystal (rugate filter) approx. 12 μm thick and 35 μm across. An organic linker, 1-undecylenic acid, was attached to the Si-H terminated inner surface of the particles by hydrosilylation and the anthracycline drug daunorubicin was bound to the carboxy terminus of the linker. Degradation of the porous Si matrix in vitro was found to release the drug in a linear and sustained fashion for 30 d. The bioactivity of the released daunorubicin was verified on retinal pigment epithelial (RPE) cells. The degradation/drug delivery process was monitored in situ by digital imaging or spectroscopic measurement of the photonic resonance reflected from the nanostructured particles, and a simple linear correlation between observed wavelength and drug release was observed. Changes in the optical reflectance spectrum were sufficiently large to be visible as a distinctive red to green color change.
Collapse
Affiliation(s)
- Elizabeth C Wu
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, United States
| | | | | | | | | | | |
Collapse
|
10
|
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 2010; 332:87-96. [PMID: 19841470 DOI: 10.1124/jpet.109.159525] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [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
MESH Headings
- Actins/metabolism
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/adverse effects
- Antineoplastic Agents/administration & dosage
- Antineoplastic Agents/adverse effects
- Apoptosis/drug effects
- Cardiotonic Agents/pharmacology
- Cell Culture Techniques
- Cell Death/drug effects
- Cell Line
- Dose-Response Relationship, Drug
- Drug Synergism
- Epirubicin/administration & dosage
- Epirubicin/adverse effects
- Glycoproteins/antagonists & inhibitors
- Immunoblotting
- Membrane Potential, Mitochondrial/drug effects
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Rats
- Rats, Sprague-Dawley
- Razoxane/pharmacology
- Receptor, ErbB-2
Collapse
Affiliation(s)
- Paolo Spallarossa
- Research Center of Cardiovascular Biology, Division of Cardiology, Department of Internal Medicine, University of Genova, Genova, Italy
| | | | | | | | | | | | | |
Collapse
|
11
|
Martin E, Thougaard AV, Grauslund M, Jensen PB, Bjorkling F, Hasinoff BB, Tjørnelund J, Sehested M, Jensen LH. Evaluation of the topoisomerase II-inactive bisdioxopiperazine ICRF-161 as a protectant against doxorubicin-induced cardiomyopathy. Toxicology 2008; 255:72-9. [PMID: 19010377 DOI: 10.1016/j.tox.2008.10.011] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2008] [Revised: 10/08/2008] [Accepted: 10/09/2008] [Indexed: 01/08/2023]
Abstract
Anthracycline-induced cardiomyopathy is a major problem in anti-cancer therapy. The only approved agent for alleviating this serious dose limiting side effect is ICRF-187 (dexrazoxane). The current thinking is that the ring-opened hydrolysis product of this agent, ADR-925, which is formed inside cardiomyocytes, removes iron from its complexes with anthracyclines, hereby reducing the concentration of highly toxic iron-anthracycline complexes that damage cardiomyocytes by semiquinone redox recycling and the production of free radicals. However, the 2 carbon linker ICRF-187 is also is a catalytic inhibitor of topoisomerase II, resulting in the risk of additional myelosuppression in patients receiving ICRF-187 as a cardioprotectant in combination with doxorubicin. The development of a topoisomerase II-inactive iron chelating compound thus appeared attractive. In the present paper we evaluate the topoisomerase II-inactive 3 carbon linker bisdioxopiperazine analog ICRF-161 as a cardioprotectant. We demonstrate that this compound does chelate iron and protects against doxorubicin-induced LDH release from primary rat cardiomyocytes in vitro, similarly to ICRF-187. The compound does not target topoisomerase II in vitro or in cells, it is well tolerated and shows similar exposure to ICRF-187 in rodents, and it does not induce myelosuppression when given at high doses to mice as opposed to ICRF-187. However, when tested in a model of chronic anthracycline-induced cardiomyopathy in spontaneously hypertensive rats, ICRF-161 was not capable of protecting against the cardiotoxic effects of doxorubicin. Modulation of the activity of the beta isoform of the topoisomerase II enzyme by ICRF-187 has recently been proposed as the mechanism behind its cardioprotection. This concept is thus supported by the present study in that iron chelation alone does not appear to be sufficient for protection against anthracycline-induced cardiomyopathy.
Collapse
Affiliation(s)
- Elke Martin
- TopoTarget A/S, Symbion Science Park, Fruebjergvej 3, Copenhagen 2100, Denmark
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Panjrath GS, Jain D. Monitoring chemotherapy-induced cardiotoxicity: role of cardiac nuclear imaging. J Nucl Cardiol 2007; 13:415-26. [PMID: 16750786 DOI: 10.1016/j.nuclcard.2006.03.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Cardiotoxicity may result from a range of chemotherapeutic agents. The prevalence of cardiotoxicity from certain cytotoxic agents is reported to be significantly high. In addition to serious side effects and increased long-lasting morbidity and mortality, dose limitation and suboptimal usage is an important adverse effect. Nuclear cardiac imaging has played a quintessential and important role in identifying patients at risk and in the prevention and reduction of cardiac injury resulting from cytotoxic agents. Despite exploring a number of other diagnostic imaging or biochemical tools for identification of cardiac injury, nuclear cardiac imaging in the form of radionuclide angiocardiography continues to be the most suitable and cost-effective tool for reducing the prevalence of cases of cardiac dysfunction resulting from chemotherapy. This article reviews the prevalence, mechanisms, and prevention strategies for cardiotoxicity associated with some of the commonly known cytotoxic agents and the role of nuclear cardiac imaging in its monitoring and prevention, along with recent advances in this area.
Collapse
Affiliation(s)
- Gurusher Singh Panjrath
- Department of Internal Medicine, St. Luke's-Roosevelt Hospital Center, New York, NY 10025, USA.
| | | |
Collapse
|
13
|
Hasinoff BB, Begleiter A. The reductive activation of the antitumor drug RH1 to its semiquinone free radical by NADPH cytochrome P450 reductase and by HCT116 human colon cancer cells. Free Radic Res 2006; 40:974-8. [PMID: 17015278 DOI: 10.1080/10715760600833077] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
RH1 (2,5-diaziridinyl-3-(hydroxymethyl)-6-methyl-1,4-benzoquinone), which is currently in clinical trials, is a diaziridinyl benzoquinone bioreductive anticancer drug that was designed to be activated by the obligate two-electron reductive enzyme NAD(P)H quinone oxidoreductase 1 (NQO1). In this electron paramagnetic resonance (EPR) study we showed that RH1 was reductively activated by the one-electron reductive enzyme NADPH cytochrome P450 reductase and by a suspension of HCT116 human colon cancer cells to yield a semiquinone free radical. As shown by EPR spin trapping experiments RH1 was reductively activated by cytochrome P450 reductase and underwent redox cycling to produce damaging hydroxyl radicals in reactions that were both H2O2- and iron-dependent. Thus, reductive activation by cytochrome P450 reductase or other reductases to produce a semiquinone that can redox cycle to produce damaging hydroxyl radicals and/or DNA-reactive alkylating species may contribute to the potent cell growth inhibitory effects of RH1. These results also suggest that selection of patients for treatment with RH1 based on their expression levels of NQO1 may be problematic.
Collapse
Affiliation(s)
- Brian B Hasinoff
- Faculty of Pharmacy, University of Manitoba, 50 Sifton Road, Winnipeg, Manitoba, Canada.
| | | |
Collapse
|
14
|
Korniłłowicz-Kowalska T, Wrzosek M, Ginalska G, Iglik H, Bancerz R. Identification and application of a new fungal strain Bjerkandera adusta R59 in decolorization of daunomycin wastes. Enzyme Microb Technol 2006. [DOI: 10.1016/j.enzmictec.2005.10.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
15
|
Barnabé N, Zastre JA, Venkataram S, Hasinoff BB. Deferiprone protects against doxorubicin-induced myocyte cytotoxicity. Free Radic Biol Med 2002; 33:266-75. [PMID: 12106822 DOI: 10.1016/s0891-5849(02)00873-0] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The iron chelating hydroxypyridinone deferiprone (CP20, L1) and the clinically approved cardioprotective agent dexrazoxane (ICRF-187) were examined for their ability to protect neonatal rat cardiac myocytes from doxorubicin-induced damage. Doxorubicin is thought to induce oxidative stress on the heart muscle, both through reductive activation to its semiquinone form, and by the production of hydroxyl radicals mediated by its complex with iron. The results of this study showed that both deferiprone and dexrazoxane were able to protect myocytes from doxorubicin-induced lactate dehydrogenase release. Deferiprone quickly and efficiently removed iron(III) from its complex with doxorubicin. In addition, this study also showed that deferiprone rapidly entered myocytes and displaced iron from a fluorescence-quenched trapped intracellular iron-calcein complex, suggesting that in the myocyte, deferiprone should also be able to displace iron from its complex with doxorubicin. It was shown by electron paramagnetic resonance spectroscopy that under hypoxic conditions myocytes were able to reduce doxorubicin to its semiquinone free radical. Deferiprone also greatly reduced hydroxyl radical production by the iron(III)-doxorubicin complex in the xanthine oxidase/xanthine superoxide generating system. Together these results suggest that deferiprone may protect against doxorubicin-induced damage to myocytes by displacing iron bound to doxorubicin, or chelating free or loosely bound iron, thus preventing site-specific iron-based oxygen radical damage.
Collapse
Affiliation(s)
- Norman Barnabé
- Faculty of Pharmacy, University of Manitoba, Winnipeg, MB, Canada
| | | | | | | |
Collapse
|
16
|
Liu FT, Kelsey SM, Newland AC, Jia L. Generation of reactive oxygen species is not involved in idarubicin-induced apoptosis in human leukaemic cells. Br J Haematol 2001; 115:817-25. [PMID: 11843815 DOI: 10.1046/j.1365-2141.2001.03216.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The anthracycline antibiotic idarubicin (IDA) induces double-stranded DNA breaks, the generation of reactive oxygen species (ROS) and apoptosis in human leukaemic cells. It is unclear whether the generation of ROS is associated with the apoptotic process. Using the T-lymphoblastic leukaemic CEM cell line, we found that IDA-induced DNA breaks were correlated with final cell death. The reduction in mitochondrial membrane potential (Deltapsim) and the generation of ROS occurred simultaneously with IDA-induced activation of caspase-9 and caspase-3. Inhibition of caspases by a pan-caspase inhibitor, benzyloxycarbonyl-Val-Ala-Asp-fluoromethyl ketone (Z-VAD-fmk) completely blocked IDA-induced reduction of Deltapsim, apoptosis and final cell death. Interestingly, ROS generation was significantly enhanced by Z-VAD-fmk. ROS generation was neither caspase dependent nor part of the apoptotic process. IDA-mediated reduction in Deltapsim is caspase dependent and is not a consequence of the generation of ROS. These results indicate that IDA-induced generation of ROS and apoptosis are separate events. Inhibition of caspases facilitates IDA-mediated generation of ROS.
Collapse
Affiliation(s)
- F T Liu
- Department of Haematology/Oncology, St Bartholomew's and The Royal London School of Medicine and Dentistry, London E1 2AD, UK
| | | | | | | |
Collapse
|
17
|
Saadane N, Yue P, Alpert L, Mitmaker B, Kirby GM, Chalifour LE. Diminished molecular response to doxorubicin and loss of cardioprotective effect of dexrazoxane inEgr-1deficient female mice. Can J Physiol Pharmacol 2001. [DOI: 10.1139/y01-021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Doxorubicin (DOX) and VP16 are DNA topoisomerase II inhibitors yet only DOX induces an irreversible cardiotoxicity, likely through DOX-induced oxidative stress. Egr-1 is overexpressed after many stimuli that increase oxidative stress in vitro and after DOX-injection into adult mice in vivo. To investigate Egr-1 function in the heart, we compared the molecular and histological responses of wild type (+/+) and Egr-1 deficient (/) female mice to saline, DOX, VP16, the cardioprotectant dexrazoxane (DZR), or DOX+DZR injection. DOX, and to a lesser extent VP16, induced characteristic increases in cardiac muscle and non-muscle genes typical of cardiac damage in +/+ mice, whereas only β-MHC and Sp1 were increased in / mice. DZR-alone treated +/+ mice showed increased cardiomyocyte transnuclear width without a change to the heart to body weight (HW/BW) ratio. However, DZR-alone treated / mice had an increased HW/BW, increased cardiomyocyte transnuclear width, and gene expression changes similar to DOX-injected +/+ mice. DZR pre-injection alleviated DOX-induced gene changes in +/+ mice; in DZR+DOX injected / mice the increases in cardiac and non-muscle gene expression were equal to, or exceeded that, detected after DOX-alone or DZR-alone injections. We conclude that Egr-1 is required for DOX-induced molecular changes and for DZR-mediated cardioprotection.Key words: mice, gene expression, doxorubicin, DNA topoisomerase II inhibitors, cardioprotection.
Collapse
|
18
|
Kwok JC, Richardson DR. The cardioprotective effect of the iron chelator dexrazoxane (ICRF-187) on anthracycline-mediated cardiotoxicity. Redox Rep 2001; 5:317-24. [PMID: 11140743 DOI: 10.1179/135100000101535898] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
The cardiotoxic effect of anthracyclines limits their use in the treatment of a variety of cancers. The reason for the high susceptibility of cardiac muscle to anthracyclines remains unclear, but it appears to be due, at least in part, to the interaction of these drugs with intracellular iron (Fe). The suggestion that Fe plays an important role in anthracycline cardiotoxicity has been strengthened by observation that the chelator, dexrazoxane (ICRF-187), has a potent cardioprotective effect. In the present review, the role of Fe in the cardiotoxicity of anthracyclines is discussed together with the possible role of Fe chelation therapy as a cardioprotective strategy that may also result in enhanced antitumour activity.
Collapse
Affiliation(s)
- J C Kwok
- The Iron Metabolism and Chelation Group, The Heart Research Institute, Sydney, New South Wales, Australia
| | | |
Collapse
|
19
|
Abstract
1. Dexrazoxane (ICRF-187) is the only clinically approved drug for use in cancer patients to prevent anthracycline mediated cardiotoxicity. 2. The mode of action appears to be mainly due to the potential of the drug to remove iron from iron/anthracycline complexes and thus reduce free radical formation by these complexes. 3. Dexrazoxane also influences cell biology by its ability to inhibit topoisomerase II and its effects on the regulation of cellular iron homeostasis. 4. Although the cardioprotective effect of dexrazoxane in cancer patients undergoing chemotherapy with anthracyclines is well documented, the potential of this drug to modulate topoisomerase II activity and cellular iron metabolism may hold the key for future applications of dexrazoxane in cancer therapy, immunology, or infectious diseases.
Collapse
Affiliation(s)
- G Weiss
- Department of Internal Medicine, University Hospital, Innsbruck, Austria.
| | | | | |
Collapse
|