1
|
Goodenow D, Emmanuel F, Berman C, Sahyouni M, Richardson C. Bioflavonoids cause DNA double-strand breaks and chromosomal translocations through topoisomerase II-dependent and -independent mechanisms. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2020; 849:503144. [PMID: 32087851 DOI: 10.1016/j.mrgentox.2020.503144] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/16/2020] [Accepted: 01/17/2020] [Indexed: 11/19/2022]
Abstract
Bioflavonoids have a similar chemical structure to etoposide, the well-characterized topoisomerase II (Top2) poison, and evidence shows that they also induce DNA double-strand breaks (DSBs) and promote genome rearrangements. The purpose of this study was to determine the kinetics of bioflavonoid-induced DSB appearance and repair, and their dependence on Top2. Cells were exposed to bioflavonoids individually or in combination in the presence or absence of the Top2 catalytic inhibitor dexrazoxane. The kinetics of appearance and repair of γH2AX foci were measured. In addition, the frequency of resultant MLL-AF9 breakpoint cluster region translocations was determined. Bioflavonoids readily induced the appearance of γH2AX foci, but bioflavonoid combinations did not act additively or synergistically to promote DSBs. Myricetin-induced DSBs were mostly reduced by dexrazoxane, while genistein and quercetin-induced DSBs were only partially, but significantly, reduced. By contrast, luteolin and kaempferol-induced DSBs increased with dexrazoxane pre-treatment. Sensitivity to Top2 inhibition correlated with a significant reduction of bioflavonoid-induced MLL-AF9 translocations. These data demonstrate that myricetin, genistein, and quercetin act most similar to etoposide although with varying Top2-dependence. By contrast, luteolin and kaempferol have distinct kinetics that are mostly Top2-independent. These findings have implications for understanding the mechanisms of bioflavonoid activity and the potential of individual bioflavonoids to promote chromosomal translocations. Further, they provide direct evidence that specific Top2 inhibitors or targeted drugs could be developed that possess less leukemic potential or suppress chromosomal translocations associated with therapy-related and infant leukemias.
Collapse
Affiliation(s)
- Donna Goodenow
- University of North Carolina at Charlotte, Department of Biological Sciences, 9201 University City Boulevard, Charlotte NC, 28223, United States
| | - Faith Emmanuel
- University of North Carolina at Charlotte, Department of Biological Sciences, 9201 University City Boulevard, Charlotte NC, 28223, United States
| | - Chase Berman
- University of North Carolina at Charlotte, Department of Biological Sciences, 9201 University City Boulevard, Charlotte NC, 28223, United States
| | - Mark Sahyouni
- University of North Carolina at Charlotte, Department of Biological Sciences, 9201 University City Boulevard, Charlotte NC, 28223, United States
| | - Christine Richardson
- University of North Carolina at Charlotte, Department of Biological Sciences, 9201 University City Boulevard, Charlotte NC, 28223, United States.
| |
Collapse
|
2
|
Janežič M, Pogorelčnik B, Brvar M, Solmajer T, Perdih A. 3-substituted-1H-indazoles as Catalytic Inhibitors of the Human DNA Topoisomerase IIα. ChemistrySelect 2017. [DOI: 10.1002/slct.201601554] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Matej Janežič
- National Institute of Chemistry; Hajdrihova 19, SI- 1001 Ljubljana Slovenia
| | | | - Matjaž Brvar
- National Institute of Chemistry; Hajdrihova 19, SI- 1001 Ljubljana Slovenia
| | - Tom Solmajer
- National Institute of Chemistry; Hajdrihova 19, SI- 1001 Ljubljana Slovenia
| | - Andrej Perdih
- National Institute of Chemistry; Hajdrihova 19, SI- 1001 Ljubljana Slovenia
| |
Collapse
|
3
|
Laev SS, Salakhutdinov NF, Lavrik OI. Tyrosyl-DNA phosphodiesterase inhibitors: Progress and potential. Bioorg Med Chem 2016; 24:5017-5027. [PMID: 27687971 DOI: 10.1016/j.bmc.2016.09.045] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 08/30/2016] [Accepted: 09/18/2016] [Indexed: 10/21/2022]
Abstract
DNA topoisomerases are essential during transcription and replication. The therapeutic mechanism of action of topoisomerase inhibitors is enzyme poisoning rather than catalytic inhibition. Tyrosyl-DNA phosphodiesterases 1 or 2 were found as DNA repair enzymes hydrolyzing the covalent bond between the tyrosyl residue of topoisomerases I or II and the 3'- or 5'-phosphate groups in DNA, respectively. Tyrosyl-DNA phosphodiesterase 1 is a key enzyme in DNA repair machinery and a promising target for antitumor and neurodegenerative therapy. Inhibitors of tyrosyl-DNA phosphodiesterase 1 could act synergistically with topoisomerase I inhibitors and thereby potentiate the effects of topoisomerase I poisons. Tyrosyl-DNA phosphodiesterase 2 is an enzyme that specifically repairs DNA damages induced by topoisomerase II poisons and causes resistance to these drugs. Selective inhibition of tyrosyl-DNA phosphodiesterase 2 may be a novel approach to overcome intrinsic or acquired resistance to topoisomerase II-targeted drug therapy. Thus, agents that inhibit tyrosyl-DNA phosphodiesterases 1 and 2 have many applications in biochemical and physiological research and they have the potential to become anticancer and antiviral drugs. The structures, mechanism of action and therapeutic rationale of tyrosyl-DNA phosphodiesterase inhibitors and their development for combinations with topoisomerase inhibitors and DNA damaging agents are discussed.
Collapse
Affiliation(s)
- Sergey S Laev
- Vorozhtsov Institute of Organic Chemistry, Siberian Division, Russian Academy of Sciences, pr. akademika Lavrent'eva 9, Novosibirsk 630090, Russian Federation.
| | - Nariman F Salakhutdinov
- Vorozhtsov Institute of Organic Chemistry, Siberian Division, Russian Academy of Sciences, pr. akademika Lavrent'eva 9, Novosibirsk 630090, Russian Federation; Novosibirsk State University, Pirogova Str. 2, Novosibirsk 630090, Russian Federation
| | - Olga I Lavrik
- Novosibirsk State University, Pirogova Str. 2, Novosibirsk 630090, Russian Federation; Novosibirsk Institute of Chemical Biology and Fundamental Medicine, Siberian Division, Russian Academy of Sciences, pr. akademika Lavrent'eva 8, Novosibirsk 630090, Russian Federation
| |
Collapse
|
4
|
Nakazawa N, Mehrotra R, Arakawa O, Yanagida M. ICRF
‐193, an anticancer topoisomerase
II
inhibitor, induces arched telophase spindles that snap, leading to a ploidy increase in fission yeast. Genes Cells 2016; 21:978-93. [DOI: 10.1111/gtc.12397] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 06/26/2016] [Indexed: 12/27/2022]
Affiliation(s)
- Norihiko Nakazawa
- G0 Cell Unit Okinawa Institute of Science and Technology Graduate University Onna‐son Okinawa 904‐0495 Japan
| | - Rajesh Mehrotra
- G0 Cell Unit Okinawa Institute of Science and Technology Graduate University Onna‐son Okinawa 904‐0495 Japan
- Department of Biological Sciences BITS Pilani Rajasthan 333031 India
| | - Orie Arakawa
- G0 Cell Unit Okinawa Institute of Science and Technology Graduate University Onna‐son Okinawa 904‐0495 Japan
| | - Mitsuhiro Yanagida
- G0 Cell Unit Okinawa Institute of Science and Technology Graduate University Onna‐son Okinawa 904‐0495 Japan
| |
Collapse
|
5
|
Pogorelčnik B, Janežič M, Sosič I, Gobec S, Solmajer T, Perdih A. 4,6-Substituted-1,3,5-triazin-2(1H)-ones as monocyclic catalytic inhibitors of human DNA topoisomerase IIα targeting the ATP binding site. Bioorg Med Chem 2015; 23:4218-4229. [PMID: 26183545 DOI: 10.1016/j.bmc.2015.06.049] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 06/15/2015] [Accepted: 06/18/2015] [Indexed: 10/23/2022]
Abstract
Human DNA topoisomerase IIα (htIIα) is a validated target for the development of novel anticancer agents. Starting from our discovered 4-amino-1,3,5-triazine inhibitors of htIIα, we investigated a library of 2,4,6-trisubstituted-1,3,5-triazines for novel inhibitors that bind to the htIIα ATP binding site using a combination of structure-based and ligand-based pharmacophore models and molecular docking. 4,6-substituted-1,3,5-triazin-2(1H)-ones 8, 9 and 14 were identified as novel inhibitors with activity comparable to the established drug etoposide (1). Compound 8 inhibits the htIIα decatenation in a superior fashion to etoposide. Cleavage assays demonstrated that selected compounds 8 and 14 do not act as poisons and antagonize the poison effect of etoposide. Microscale thermophoresis (MST) confirmed binding of compound 8 to the htIIα ATPase domain and compound 14 effectively inhibits the htIIα mediated ATP hydrolysis. The molecular dynamics simulation study provides further insight into the molecular recognition. The 4,6-disubstituted-1,3,5-triazin-2(1H)-ones represent the first validated monocyclic class of catalytic inhibitors that bind to the to the htIIα ATPase domain.
Collapse
Affiliation(s)
| | - Matej Janežič
- National Institute of Chemistry, Hajdrihova 19, 1001 Ljubljana, Slovenia
| | - Izidor Sosič
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Stanislav Gobec
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Tom Solmajer
- National Institute of Chemistry, Hajdrihova 19, 1001 Ljubljana, Slovenia
| | - Andrej Perdih
- National Institute of Chemistry, Hajdrihova 19, 1001 Ljubljana, Slovenia.
| |
Collapse
|
6
|
Mosesso P, Pepe G, Ottavianelli A, Schinoppi A, Cinelli S. Cytogenetic evidence that DNA topoisomerase II is not involved in radiation induced chromsome-type aberrations. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2015; 793:14-8. [PMID: 26520368 DOI: 10.1016/j.mrgentox.2015.06.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 06/09/2015] [Indexed: 11/17/2022]
Abstract
ICRF-187 (Cardioxane™, Chiron) is a catalytic inhibitor of DNA topoisomerase II (Topo II), proposed to act by blocking Topo II-mediated DNA cleavage without stabilizing DNA-Topo II-"cleavable complexes". In this study ICRF-187 was used to evaluate the potential involvement of DNA topoisomerase II in the formation of the radiation-induced chromosome-type aberrations in the G0 phase of the cell cycle in human lymphocytes from three healthy male donors. This is based on many evidences that DNA topoisomerases are involved in DNA recombination, mainly of illegitimate type (non-homologous) both in vitro and in vivo. The results obtained clearly indicated that ICRF-187 did not induce per se any chromosomal damage. When challenged with the non-catalytic Topo II poison VP-16 (etoposide), which acts by stabilizing the "cleavable complex" generating "protein concealed" DSB's and thus chromosomal aberrations, it completely abolished the significant induction of chromosome-type aberrations and formation of dicentric chromosomes. This indicates that ICRF-187 acts effectively as catalytic inhibitor of Topo II. On the other hand, when X-ray treatments were challenged with ICRF-187 using experimental conditions as for VP-16 treatments, no modification of the incidence of chromosome-type aberrations and dicentric chromosomes was observed. On this basis, we conclude that Topo II is not involved in the formation of X-ray-induced chromosome-type aberrations and dicentric chromosomes in human lymphocytes in the G0 phase of the cell cycle.
Collapse
Affiliation(s)
- P Mosesso
- Dipartimento di Scienze Ecologiche e Biologiche, Università degli Studi della Tuscia, Largo dell'Università s.n.c., 01100 Viterbo, Italy.
| | - G Pepe
- Dipartimento di Scienze Ecologiche e Biologiche, Università degli Studi della Tuscia, Largo dell'Università s.n.c., 01100 Viterbo, Italy
| | - A Ottavianelli
- Dipartimento di Scienze Ecologiche e Biologiche, Università degli Studi della Tuscia, Largo dell'Università s.n.c., 01100 Viterbo, Italy
| | - A Schinoppi
- Dipartimento di Scienze Ecologiche e Biologiche, Università degli Studi della Tuscia, Largo dell'Università s.n.c., 01100 Viterbo, Italy
| | - S Cinelli
- Research Toxicology Centre, Via Tito Speri 12/14, 00040, Pomezia, Roma, Italy
| |
Collapse
|
7
|
Pogorelčnik B, Brvar M, Žegura B, Filipič M, Solmajer T, Perdih A. Discovery of Mono- and Disubstituted 1H-Pyrazolo[3,4]pyrimidines and 9H-Purines as Catalytic Inhibitors of Human DNA Topoisomerase IIα. ChemMedChem 2014; 10:345-59. [DOI: 10.1002/cmdc.201402459] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Indexed: 11/07/2022]
|
8
|
Pogorelčnik B, Brvar M, Zajc I, Filipič M, Solmajer T, Perdih A. Monocyclic 4-amino-6-(phenylamino)-1,3,5-triazines as inhibitors of human DNA topoisomerase IIα. Bioorg Med Chem Lett 2014; 24:5762-5768. [PMID: 25453816 DOI: 10.1016/j.bmcl.2014.10.042] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 10/09/2014] [Accepted: 10/14/2014] [Indexed: 10/24/2022]
Abstract
Human DNA topoisomerase IIα (htIIα) is a validated target for the development of anticancer agents. Starting from the available information about the binding of the purine-based htIIα inhibitors in the ATP binding site we designed a virtual screening campaign combining structure-based and ligand-based pharmacophores with a molecular docking calculation searching for compounds that would contain a monocycle mimetic of the purine moiety. We discovered novel 4-amino-6-(phenylamino)-1,3,5-triazines 6, 7 and 11 as monocyclic htIIα inhibitors targeting the ATP binding site. Compound 6 from the 1,3,5-triazine series also displayed cytotoxicity properties in hepatocellular carcinoma (HepG2) cell lines and selectivity against human umbilical vein endothelial (HUVEC) cell lines.
Collapse
Affiliation(s)
| | - Matjaž Brvar
- National Institute of Chemistry, Hajdrihova 19, 1001 Ljubljana, Slovenia
| | - Irena Zajc
- National Institute of Biology, Department of Genetic Toxicology and Cancer Biology, Večna pot 111, 1000 Ljubljana, Slovenia
| | - Metka Filipič
- National Institute of Biology, Department of Genetic Toxicology and Cancer Biology, Večna pot 111, 1000 Ljubljana, Slovenia
| | - Tom Solmajer
- National Institute of Chemistry, Hajdrihova 19, 1001 Ljubljana, Slovenia
| | - Andrej Perdih
- National Institute of Chemistry, Hajdrihova 19, 1001 Ljubljana, Slovenia.
| |
Collapse
|
9
|
Yang YC, Chou HYE, Shen TL, Chang WJ, Tai PH, Li TK. Topoisomerase II-mediated DNA cleavage and mutagenesis activated by nitric oxide underlie the inflammation-associated tumorigenesis. Antioxid Redox Signal 2013; 18:1129-40. [PMID: 22998676 DOI: 10.1089/ars.2012.4620] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
AIMS Both cancer-suppressing and cancer-promoting properties of reactive nitrogen and oxygen species (RNOS) have been suggested to play a role in tumor pathology, particularly those activities associated with chronic inflammation. Here, we address the impact of nitric oxide (NO) on the induction of DNA damage and genome instability with a specific focus on the involvement of topoisomerase II (TOP2). We also investigate the contribution of NO to the formation of skin melanoma in mice. RESULTS Similar to the TOP2-targeting drug, etoposide (VP-16), the NO-donor, S-nitrosoglutathione (GSNO), induces skin melanomas formation in 7,12-dimethyl- benz[a]anthracene (DMBA)-initiated mice. To explore the mechanism(s) underlying this NO-induced tumorigenesis, we use a co-culture model system to demonstrate that inflamed macrophages with inducible NO synthase (iNOS) expression cause γ-H2AX activation, p53 phosphorylation, and chromosome DNA breaks in the target cells. Inhibitor experiments revealed that NO and TOP2 isozymes are responsible for the above described cellular phenotypes. Notably, NO, unlike VP-16, preferentially induces the formation of TOP2β cleavable complexes (TOP2βcc) in cells. Moreover, GSNO induced TOP2-dependent DNA sequence rearrangements and cytotoxicity. Furthermore, the incidences of GSNO- and VP-16-induced skin melanomas were also observed to be lower in the skin-specific top2β-knockout mice. Our results suggest that TOP2 isozymes contribute to NO-induced mutagenesis and subsequent cancer development during chronic inflammation. INNOVATION AND CONCLUSIONS We provide the first experimental evidence for the functional role of TOP2 in NO-caused DNA damage, mutagenesis, and carcinogenesis. Notably, these studies contribute to our molecular understanding of the cancer-promoting actions of RNOS during chronic inflammation.
Collapse
Affiliation(s)
- Yu-Chen Yang
- Department and Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
10
|
Abstract
Topoisomerases are ubiquitous enzymes that control DNA supercoiling and entanglements. They are essential during transcription and replication, and topoisomerase inhibitors are among the most effective and most commonly used anticancer and antibacterial drugs. This review consists of two parts. In the first part ("Lessons"), it gives background information on the catalytic mechanisms of the different enzyme families (6 different genes in humans and 4 in most bacteria), describes the "interfacial inhibition" by which topoisomerase-targeted drugs act as topoisomerase poisons, and describes clinically relevant topoisomerase inhibitors. It generalizes the interfacial inhibition principle, which was discovered from the mechanism of action of topoisomerase inhibitors, and discusses how topoisomerase inhibitors kill cells by trapping topoisomerases on DNA rather than by classical enzymatic inhibition. Trapping protein-DNA complexes extends to a novel mechanism of action of PARP inhibitors and could be applied to the targeting of transcription factors. The second part of the review focuses on the challenges for discovery and precise use of topoisomerase inhibitors, including targeting topoisomerase inhibitors using chemical coupling and encapsulation for selective tumor delivery, use of pharmacodynamic biomarkers to follow drug activity, complexity of the response determinants for anticancer activity and patient selection, prospects of rational combinations with DNA repair inhibitors targeting tyrosyl-DNA-phosphodiesterases 1 and 2 (TDP1 and TDP2) and PARP, and the unmet need to develop inhibitors for type IA enzymes.
Collapse
Affiliation(s)
- Yves Pommier
- Laboratory of Molecular
Pharmacology, Center for Cancer
Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
11
|
Marchand C, Pommier Y. Topoisomerases Inhibitors: A Paradigm for Interfacial Inhibition. ACTA ACUST UNITED AC 2011. [DOI: 10.1007/978-1-4614-0323-4_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
|
12
|
Medikayala S, Piteo B, Zhao X, Edwards JG. Chronically elevated glucose compromises myocardial mitochondrial DNA integrity by alteration of mitochondrial topoisomerase function. Am J Physiol Cell Physiol 2010; 300:C338-48. [PMID: 21123731 DOI: 10.1152/ajpcell.00248.2010] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Mitochondrial dysfunction has a significant role in the development and complications of diabetic cardiomyopathy. Mitochondrial dysfunction and mitochondrial DNA (mtDNA) mutations are also associated with different types of cancer and neurodegenerative diseases. The goal of this study was to determine if chronically elevated glucose increase in mtDNA damage contributed to mitochondrial dysfunction and identify the underlying basis for mtDNA damage. H9c2 myotubes (a cardiac-derived cell line) were studied in the presence of 5.5, 16.5, or 33.0 mM glucose for up to 13 days. Tests of mitochondria function (Complex I and IV activity and ATP generation) were all significantly depressed by elevated media glucose. Intramitochondrial superoxide and intracellular superoxide levels were transiently increased during the experimental period. AnnexinV binding (a marker of apoptosis) was significantly increased after 7 and 13 days of high glucose. Thirteen days of elevated glucose significantly increased mtDNA damage globally and across the region encoding for the three subunits of cytochrome oxidase. Using mitochondria isolated from cells chronically exposed to elevated glucose, we observed significant increases in topoisomerase-linked DNA cleavage. Mitochondria-dependent DNA cleavage was significantly exacerbated by H(2)O(2) and that immunoprecipitation of mitochondrial extracts with a mtTOP1 antibody significantly decreased DNA cleavage, indicating that at least part of this activity could be attributed to mtTOP1. We conclude that even mild increases in glucose presentation compromised mitochondrial function as a result of a decline in mtDNA integrity. Separate from a direct impact of oxidative stress on mtDNA, ROS-induced alteration of mitochondrial topoisomerase activity exacerbated and propagated increases in mtDNA damage. These findings are significant in that the activation/inhibition state of the mitochondrial topoisomerases will have important consequences for mitochondrial DNA integrity and the well being of the myocardium.
Collapse
Affiliation(s)
- S Medikayala
- Dept. of Physiology, New York Medical College, Valhalla, NY 10595, USA
| | | | | | | |
Collapse
|
13
|
Toyoda E, Kagaya S, Cowell IG, Kurosawa A, Kamoshita K, Nishikawa K, Iiizumi S, Koyama H, Austin CA, Adachi N. NK314, a topoisomerase II inhibitor that specifically targets the alpha isoform. J Biol Chem 2008; 283:23711-20. [PMID: 18596031 PMCID: PMC3259784 DOI: 10.1074/jbc.m803936200] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2008] [Revised: 06/27/2008] [Indexed: 11/06/2022] Open
Abstract
Topoisomerase II (Top2) is a ubiquitous nuclear enzyme that relieves torsional stress in chromosomal DNA during various cellular processes. Agents that target Top2, involving etoposide, doxorubicin, and mitoxantrone, are among the most effective anticancer drugs used in the clinic. Mammalian cells possess two genetically distinct Top2 isoforms, both of which are the target of these agents. Top2alpha is essential for cell proliferation and is highly expressed in vigorously growing cells, whereas Top2beta is nonessential for growth and has recently been implicated in treatment-associated secondary malignancies, highlighting the validity of a Top2alpha-specific drug for future cancer treatment; however, no such agent has been hitherto reported. Here we show that NK314, a novel synthetic benzo[c]phenanthridine alkaloid, targets Top2alpha and not Top2beta in vivo. Unlike other Top2 inhibitors, NK314 induces Top2-DNA complexes and double-strand breaks (DSBs) in an alpha isoform-specific manner. Heterozygous disruption of the human TOP2alpha gene confers increased NK314 resistance, whereas TOP2beta homozygous knock-out cells display increased NK314 sensitivity, indicating that the alpha isoform is the cellular target. We further show that the absence of Top2beta does not alleviate NK314 hypersensitivity of cells deficient in non-homologous end-joining, a critical pathway for repairing Top2-mediated DSBs. Our results indicate that NK314 acts as a Top2alpha-specific poison in mammalian cells, with excellent potential as an efficacious and safe chemotherapeutic agent. We also suggest that a series of human knock-out cell lines are useful in assessing DNA damage and repair induced by potential topoisomerase-targeting agents.
Collapse
Affiliation(s)
- Eriko Toyoda
- International Graduate School of Arts and
Sciences, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027,
Japan, the Pharmaceutical Research Laboratories,
Nippon Kayaku Co., Ltd., 31-12, Shimo 3-chome, Kita-ku, Tokyo 115-8588, Japan,
and the Institute for Cell and Molecular
Biosciences, The Medical School, Newcastle University, Newcastle-upon-Tyne,
NE2 4HH United Kingdom
| | - Shigehide Kagaya
- International Graduate School of Arts and
Sciences, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027,
Japan, the Pharmaceutical Research Laboratories,
Nippon Kayaku Co., Ltd., 31-12, Shimo 3-chome, Kita-ku, Tokyo 115-8588, Japan,
and the Institute for Cell and Molecular
Biosciences, The Medical School, Newcastle University, Newcastle-upon-Tyne,
NE2 4HH United Kingdom
| | - Ian G. Cowell
- International Graduate School of Arts and
Sciences, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027,
Japan, the Pharmaceutical Research Laboratories,
Nippon Kayaku Co., Ltd., 31-12, Shimo 3-chome, Kita-ku, Tokyo 115-8588, Japan,
and the Institute for Cell and Molecular
Biosciences, The Medical School, Newcastle University, Newcastle-upon-Tyne,
NE2 4HH United Kingdom
| | - Aya Kurosawa
- International Graduate School of Arts and
Sciences, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027,
Japan, the Pharmaceutical Research Laboratories,
Nippon Kayaku Co., Ltd., 31-12, Shimo 3-chome, Kita-ku, Tokyo 115-8588, Japan,
and the Institute for Cell and Molecular
Biosciences, The Medical School, Newcastle University, Newcastle-upon-Tyne,
NE2 4HH United Kingdom
| | - Keiichi Kamoshita
- International Graduate School of Arts and
Sciences, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027,
Japan, the Pharmaceutical Research Laboratories,
Nippon Kayaku Co., Ltd., 31-12, Shimo 3-chome, Kita-ku, Tokyo 115-8588, Japan,
and the Institute for Cell and Molecular
Biosciences, The Medical School, Newcastle University, Newcastle-upon-Tyne,
NE2 4HH United Kingdom
| | - Kiyohiro Nishikawa
- International Graduate School of Arts and
Sciences, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027,
Japan, the Pharmaceutical Research Laboratories,
Nippon Kayaku Co., Ltd., 31-12, Shimo 3-chome, Kita-ku, Tokyo 115-8588, Japan,
and the Institute for Cell and Molecular
Biosciences, The Medical School, Newcastle University, Newcastle-upon-Tyne,
NE2 4HH United Kingdom
| | - Susumu Iiizumi
- International Graduate School of Arts and
Sciences, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027,
Japan, the Pharmaceutical Research Laboratories,
Nippon Kayaku Co., Ltd., 31-12, Shimo 3-chome, Kita-ku, Tokyo 115-8588, Japan,
and the Institute for Cell and Molecular
Biosciences, The Medical School, Newcastle University, Newcastle-upon-Tyne,
NE2 4HH United Kingdom
| | - Hideki Koyama
- International Graduate School of Arts and
Sciences, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027,
Japan, the Pharmaceutical Research Laboratories,
Nippon Kayaku Co., Ltd., 31-12, Shimo 3-chome, Kita-ku, Tokyo 115-8588, Japan,
and the Institute for Cell and Molecular
Biosciences, The Medical School, Newcastle University, Newcastle-upon-Tyne,
NE2 4HH United Kingdom
| | - Caroline A. Austin
- International Graduate School of Arts and
Sciences, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027,
Japan, the Pharmaceutical Research Laboratories,
Nippon Kayaku Co., Ltd., 31-12, Shimo 3-chome, Kita-ku, Tokyo 115-8588, Japan,
and the Institute for Cell and Molecular
Biosciences, The Medical School, Newcastle University, Newcastle-upon-Tyne,
NE2 4HH United Kingdom
| | - Noritaka Adachi
- International Graduate School of Arts and
Sciences, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027,
Japan, the Pharmaceutical Research Laboratories,
Nippon Kayaku Co., Ltd., 31-12, Shimo 3-chome, Kita-ku, Tokyo 115-8588, Japan,
and the Institute for Cell and Molecular
Biosciences, The Medical School, Newcastle University, Newcastle-upon-Tyne,
NE2 4HH United Kingdom
| |
Collapse
|
14
|
Lyu YL, Kerrigan JE, Lin CP, Azarova AM, Tsai YC, Ban Y, Liu LF. Topoisomerase IIbeta mediated DNA double-strand breaks: implications in doxorubicin cardiotoxicity and prevention by dexrazoxane. Cancer Res 2007; 67:8839-46. [PMID: 17875725 DOI: 10.1158/0008-5472.can-07-1649] [Citation(s) in RCA: 442] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Doxorubicin is among the most effective and widely used anticancer drugs in the clinic. However, cardiotoxicity is one of the life-threatening side effects of doxorubicin-based therapy. Dexrazoxane (Zinecard, also known as ICRF-187) has been used in the clinic as a cardioprotectant against doxorubicin cardiotoxicity. The molecular basis for doxorubicin cardiotoxicity and the cardioprotective effect of dexrazoxane, however, is not fully understood. In the present study, we showed that dexrazoxane specifically abolished the DNA damage signal gamma-H2AX induced by doxorubicin, but not camptothecin or hydrogen peroxide, in H9C2 cardiomyocytes. Doxorubicin-induced DNA damage was also specifically abolished by the proteasome inhibitors bortezomib and MG132 and much reduced in top2beta(-/-) mouse embryonic fibroblasts (MEF) compared with TOP2beta(+/+) MEFs, suggesting the involvement of proteasome and DNA topoisomerase IIbeta (Top2beta). Furthermore, in addition to antagonizing Top2 cleavage complex formation, dexrazoxane also induced rapid degradation of Top2beta, which paralleled the reduction of doxorubicin-induced DNA damage. Together, our results suggest that dexrazoxane antagonizes doxorubicin-induced DNA damage through its interference with Top2beta, which could implicate Top2beta in doxorubicin cardiotoxicity. The specific involvement of proteasome and Top2beta in doxorubicin-induced DNA damage is consistent with a model in which proteasomal processing of doxorubicin-induced Top2beta-DNA covalent complexes exposes the Top2beta-concealed DNA double-strand breaks.
Collapse
Affiliation(s)
- Yi Lisa Lyu
- Department of Pharmacology, University of Medicine and Dentistry of New Jersey-Robert Wood Johnson Medical School, Piscataway, New Jersey 08854-5635, USA.
| | | | | | | | | | | | | |
Collapse
|
15
|
Swift LP, Cutts SM, Nudelman A, Levovich I, Rephaeli A, Phillips DR. The cardio-protecting agent and topoisomerase II catalytic inhibitor sobuzoxane enhances doxorubicin-DNA adduct mediated cytotoxicity. Cancer Chemother Pharmacol 2007; 61:739-49. [PMID: 17594094 DOI: 10.1007/s00280-007-0528-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2007] [Accepted: 05/11/2007] [Indexed: 11/24/2022]
Abstract
PURPOSE The importance of understanding the mechanism of action of anticancer agents is sometimes overlooked in the pursuit of new and therapeutically advantageous compounds. Doxorubicin has long been identified as an inhibitor of the DNA-decatenating enzyme topoisomerase II, this being believed to be the major mechanism of action of this drug. However, the complex nature of cytotoxicity induced by doxorubicin suggests that more than one mechanism of action is responsible for cell kill. Investigation into various other cellular effects has shown that doxorubicin can, in the presence of formaldehyde, form doxorubicin-DNA adducts, resulting in enhanced cell death. METHODS We have used six catalytic inhibitors of topoisomerase II (aclarubicin, merbarone, suramin, staurosporine, maleimide and sobuzoxane) to investigate the role of topoisomerase II mediated cell effects in doxorubicin-DNA adduct inducing treatments. Adduct levels were determined by scintillation counting of [14C]doxorubicin-DNA lesions and DNA damage responses by Comet analysis and flow cytometry (apoptosis). RESULTS Here we show that sobuzoxane inhibits topoisomerase II but in the presence of doxorubicin also enhances the production of doxorubicin-DNA adducts resulting in an enhanced cytotoxic response. We show that the formation of doxorubicin-DNA adducts is mediated by formaldehyde released from sobuzoxane when it is metabolised. CONCLUSIONS Sobuzoxane has also been shown to decrease the normally dose limiting cardiotoxicity commonly exhibited with clinical use of doxorubicin. The potential combination of doxorubicin and sobuzoxane in cancer chemotherapy has two advantages. First, the mechanism of doxorubicin toxicity is shifted away from topoisomerase II inhibition and towards drug-DNA adduct formation which may allow for a lower drug dose to be used and circumvent some drug resistance problems. Second, the addition of a cardioprotecting agent will counteract the commonly dose limiting side effect of cardiac damage resulting from doxorubicin treatment. The importance of the potentiation of cell kill of doxorubicin and sobuzoxane provides a rationalisation of a mechanistic-based combination of anticancer drugs for an improved clinical outcome.
Collapse
Affiliation(s)
- Lonnie P Swift
- Department of Biochemistry, La Trobe University, Bundoora, VIC 3086, Australia
| | | | | | | | | | | |
Collapse
|
16
|
Studies on the reduction and reductive alkylation of amino acid-derived spirocyclic 2,6-dioxopiperazines. Tetrahedron 2007. [DOI: 10.1016/j.tet.2007.01.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
17
|
Gabibov A, Yakubovskaya E, Lukin M, Favorov P, Reshetnyak A, Monastyrsky M. Catalytic transformations of supercoiled DNA as studied by flow linear dichroism technique. FEBS J 2006; 272:6336-43. [PMID: 16336270 DOI: 10.1111/j.1742-4658.2005.05027.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
A catalytic turnover of supercoiled DNA (scDNA) transformation mediated by topoisomerases leads to changes in the linking number (Lk) of the polymeric substrate by 1 or 2 per cycle. As a substrate of the topoisomerization reaction it is chemically identical to its product; even a single catalytic event results in the quantum leap in the scDNA topology. Non-intrusive continuous assay to measure the kinetics of the scDNA topoisomerization was performed. The development of such a technique was hindered because of multiple DNA species of intermediate topology present in the reaction mixture. The interrelation of DNA topology, its hydrodynamics, and optical anisotropy enable us to use the flow linear dichroism technique (FLD) for continuous monitoring of the scDNA topoisomerization reaction. This approach permits us to study the kinetics of DNA transformation catalyzed by eukaryotic topoisomerases I and II, as well as mechanistic characteristics of these enzymes and their interactions with anticancer drugs. Moreover, FLD assay can be applied to any enzymatic reaction that involves scDNA as a substrate. It also provides a new way of screening drugs dynamically and is likely to be potent in various biomedical applications.
Collapse
Affiliation(s)
- Alexander Gabibov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.
| | | | | | | | | | | |
Collapse
|
18
|
|
19
|
Wang L, Eastmond DA. Catalytic inhibitors of topoisomerase II are DNA-damaging agents: induction of chromosomal damage by merbarone and ICRF-187. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2002; 39:348-356. [PMID: 12112387 DOI: 10.1002/em.10072] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Merbarone is a catalytic inhibitor of topoisomerase II (topo II) that has been proposed to act primarily by blocking topo II-mediated DNA cleavage without stabilizing DNA-topo II-cleavable complexes. In this study merbarone was used as a model compound to investigate the genotoxic effects of catalytic inhibitors of topo II. The clastogenic properties of merbarone were evaluated using in vitro and in vivo micronucleus (MN) assays combined with CREST staining. For the in vitro MN assay, ICRF-187, a different type of catalytic inhibitor, and etoposide, a topo II poison, were used for comparison. Treatment of TK6 cells with all three of these drugs resulted in highly significant dose-related increases in kinetochore-lacking MN and, to a lesser extent, kinetochore-containing MN. In addition, a good correlation between p53 accumulation and MN formation was seen in the drug-treated cells. A mouse MN assay was performed to confirm that similar DNA-damaging effects would occur in vivo. Bone marrow smears from merbarone-treated B6C3F1 mice showed a dose-related increase in micronucleated polychromatic erythrocytes with a mean of 26 MN per 1000 cells being seen at the 60 mg/kg dose. Almost all MN lacked a kinetochore signal, indicating that merbarone was predominantly clastogenic under these conditions in vivo. The present study clearly shows that merbarone is genotoxic both in vitro and in vivo, and demonstrates the inaccuracy of earlier statements that merbarone and other catalytic inhibitors block the enzymatic activity of topo II without damaging DNA.
Collapse
Affiliation(s)
- Ling Wang
- Environmental Toxicology Graduate Program, Department of Cell Biology and Neuroscience, University of California, Riverside, California 92521, USA
| | | |
Collapse
|
20
|
Morgan SE, Beck WT. Role of an inverted CCAAT element in human topoisomerase IIalpha gene expression in ICRF-187-sensitive and -resistant CEM leukemic cells. Mol Pharmacol 2001; 59:203-11. [PMID: 11160854 DOI: 10.1124/mol.59.2.203] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
DNA topoisomerase (topo) IIalpha gene expression or activity is altered in tumor cells selected for resistance to inhibitors of topoII. To better understand the mechanisms by which topoIIalpha expression levels are modulated, we examined topoIIalpha transcriptional regulation in ICRF-187-sensitive and ICRF-187-resistant human leukemic cell lines that express an increased amount of topoIIalpha protein and mRNA. Transient transfections of luciferase reporter plasmids containing either the full-length human topoIIalpha promoter or fragments of it revealed that topoIIalpha transcriptional activity was significantly increased in the drug-resistant CEM/ICRF-8 cells, compared with CEM cells. Specifically, the transcriptional activity of the full-length topoIIalpha promoter (nucleotides -557 to +90) was doubled in CEM/ICRF-8 compared with CEM cells. Serial deletion of the topoIIalpha promoter permitted localization of the region responsible for its up-regulation in the drug-resistant cells between nucleotides -557 and -162, which includes the last three inverted CCAAT elements (ICE) 3 to 5. Note that construction of a point mutation in ICE3 resulted in a significant increase in transcriptional activity of the topoIIalpha promoter in the drug-sensitive CEM cells. In addition, by electrophoretic mobility shift assay, ICE3 was recognized by a protein complex containing NF-YB that was present at reduced levels in the topoIIalpha-overexpressing CEM/ICRF-8 extracts, suggesting that ICE3 plays a negative regulatory role in human topoIIalpha gene expression. This is the first study to show that topoIIalpha transcriptional up-regulation in ICRF-187-resistant cells is mediated in part by altered regulation of the third inverted CCAAT box in the topoIIalpha promoter.
Collapse
Affiliation(s)
- S E Morgan
- Division of Molecular Pharmacology, Departments of Molecular Genetics, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| | | |
Collapse
|
21
|
Patel S, Jazrawi E, Creighton AM, Austin CA, Fisher LM. Probing the interaction of the cytotoxic bisdioxopiperazine ICRF-193 with the closed enzyme clamp of human topoisomerase IIalpha. Mol Pharmacol 2000; 58:560-8. [PMID: 10953049 DOI: 10.1124/mol.58.3.560] [Citation(s) in RCA: 25] [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
Topoisomerase II is an ATP-operated protein clamp that captures a DNA helix and transports it through another DNA duplex, allowing chromosome segregation at mitosis. A number of cytotoxic bisdioxopiperazines such as ICRF-193 target topoisomerase II by binding and trapping the closed enzyme clamp. To investigate this unusual mode of action, we have used yeast to select plasmid-borne human topoisomerase IIalpha alleles resistant to ICRF-193. Mutations in topoisomerase IIalpha of Leu-169 to Phe (L169F) (in the N-terminal ATPase domain) and Ala-648 to Pro (A648P) (in the core domain) were identified as conferring >50-fold and 5-fold resistance to ICRF-193 in vivo, respectively. The L169F mutation, located next to the Walker A box ATP-binding sequence, resulted in a mutant enzyme displaying ICRF-193-resistant topoisomerase and ATPase activities and whose closed clamp was refractory to ICRF-193-mediated trapping as an annulus on closed circular DNA. These data imply that the mutation interferes directly with ICRF-193 binding to the N-terminal ATPase gate. In contrast, the A648P enzyme displayed topoisomerase activities exhibiting wild-type sensitivity to ICRF-193. We suggest that the inefficient trapping of the A648P closed clamp results either from the observed increased ATP requirement, or more likely, from lowered salt stability, perhaps involving destabilization of ICRF-193 interactions with the B'-B' interface in the core domain. These results provide evidence for at least two different phenotypic classes of ICRF-193 resistance mutations and suggest that bisdioxopiperazine action involves the interplay of both the ATPase and core domains of topoisomerase IIalpha.
Collapse
Affiliation(s)
- S Patel
- Molecular Genetics Group, Department of Biochemistry and Immunology, St. George's Hospital Medical School, University of London, London, United Kingdom
| | | | | | | | | |
Collapse
|
22
|
Hammonds TR, Foster SR, Maxwell A. Increased sensitivity to quinolone antibacterials can be engineered in human topoisomerase IIalpha by selective mutagenesis. J Mol Biol 2000; 300:481-91. [PMID: 10884345 DOI: 10.1006/jmbi.2000.3892] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A potential region of drug-DNA interaction in the A subunit of DNA gyrase has previously been identified from crystallographic studies. The local amino acid sequence has been compared with similar regions in yeast topoisomerase II and human topoisomerase IIalpha. Three non- conserved, potentially solvent-accessible residues at positions 762, 763 and 766 in human topoisomerase IIalpha lie between well-conserved regions. The corresponding residues in GyrA (83, 84 and 87) have a high frequency of mutation in quinolone-resistant bacteria. Mutations in human topoisomerase IIalpha have been generated in an attempt to engineer ciprofloxacin sensitivity into this enzyme: M762S, S763A and M766D (each mutated to the identical amino acid present in gyrase), along with an M762S/S763A double mutant and a triple mutant. These enzymes were introduced into a temperature-sensitive yeast strain, deficient in topoisomerase II, for in vivo studies, and were overproduced for in vitro studies. The M766D mutation renders the enzyme incapable of supporting the temperature-sensitive strain at a non-permissive temperature. However, both M766D and the triple mutant enzymes can be overproduced and are fully active in vitro. The double mutant was impaired in its ability to cleave DNA and had reduced catalytic activity. The triple mutation confers a three-fold increase in sensitivity to ciprofloxacin in vitro and similar sensitivities to a range of other quinolones. The activity of the quinolone CP-115,953, a bacterial and eukaryotic topoisomerase II poison, was unaffected by any of these mutations. Mutations in this region were found to increase the sensitivity of the enzyme to the DNA intercalating anti-tumour agents m-AMSA and ellipticine, but confer resistance to the non-intercalating agents etoposide, teniposide and merbarone, an effect that was maximal in the triple mutant. We have therefore shown the importance of this region in determining the sensitivity of topoisomerase II to drugs and have engineered increased sensitivity to quinolones.
Collapse
Affiliation(s)
- T R Hammonds
- Department of Biochemistry, University of Leicester, Leicester, LE1 7RH, UK.
| | | | | |
Collapse
|
23
|
Ravandi-Kashani F, Sriswasdi C, Lynott A, Giles FJ. HTLV-1 and Adult T-Cell Leukemia/Lymphoma: A Review. Hematology 1998; 3:429-41. [PMID: 27420330 DOI: 10.1080/10245332.1998.11746417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Infection with the human T-lymphotropic virus type 1 (HTLV-1) has been shown to be fundamental to the etiology of Adult T-cell Leukemia/Lymphoma (ATL). The disease is endemic in specific geographic areas but is increasingly reported from non-endemic regions. With increasing number of patients with this entity, the diversity in the clinical features has become apparent. In the past treatment strategies using combination chemotherapy have been unsatisfactory, but more recent trials using adenosine analouges, interferons, and combination of interferons and AZT have shown promise. With increased understanding of the etiology and molecular basis of the disease more effective therapies can be anticipated.
Collapse
Affiliation(s)
- F Ravandi-Kashani
- a Department of Leukemia , University of Texas , M.D. Anderson Cancer Center , Houston , Texas
| | - C Sriswasdi
- a Department of Leukemia , University of Texas , M.D. Anderson Cancer Center , Houston , Texas
| | - A Lynott
- b International Oncology Study Group , Houston , Texas
| | - F J Giles
- a Department of Leukemia , University of Texas , M.D. Anderson Cancer Center , Houston , Texas
| |
Collapse
|