1
|
Minko IG, Moellmer SA, Luzadder MM, Tomar R, Stone MP, McCullough AK, Lloyd RS. Interaction of mitoxantrone with abasic sites - DNA strand cleavage and inhibition of apurinic/apyrimidinic endonuclease 1, APE1. DNA Repair (Amst) 2024; 133:103606. [PMID: 38039951 PMCID: PMC11257150 DOI: 10.1016/j.dnarep.2023.103606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 10/04/2023] [Accepted: 11/15/2023] [Indexed: 12/03/2023]
Abstract
Mitoxantrone (1,4-dihydroxy-5,8-bis[2-(2-hydroxyethylamino)ethylamino]-anthracene-9,10-dione) is a clinically-relevant synthetic anthracenedione that functions as a topoisomerase II poison by trapping DNA double-strand break intermediates. Mitoxantrone binds to DNA via both stacking interactions with DNA bases and hydrogen bonding with the sugar-phosphate backbone. It has been shown that mitoxantrone inhibits apurinic/apyrimidinic (AP) endonuclease 1 (APE1)-catalyzed incision of DNA containing a tetrahydrofuran (THF) moiety and more recently, that mitoxantrone forms Schiff base conjugates at AP sites in DNA. In this study, mitoxantrone-mediated inhibition of APE1 at THF sites was shown to be consistent with preferential binding to, and thermal stabilization of DNA containing a THF site as compared to non-damaged DNA. Investigations into the properties of mitoxantrone at AP and 3' α,β-unsaturated aldehyde sites demonstrated that in addition to being a potent inhibitor of APE1 at these biologically-relevant substrates (∼ 0.5 μM IC50 on AP site-containing DNA), mitoxantrone also incised AP site-containing DNA by catalyzing β- and β/δ-elimination reactions. The efficiency of these reactions to generate the 3' α,β-unsaturated aldehyde and 3' phosphate products was modulated by DNA structure. Although these cell-free reactions revealed that mitoxantrone can generate 3' phosphates, cells lacking polynucleotide kinase phosphatase did not show increased sensitivity to mitoxantrone treatment. Consistent with its ability to inhibit APE1 activity on DNAs containing either an AP site or a 3' α,β-unsaturated aldehyde, combined exposures to clinically-relevant concentrations of mitoxantrone and a small molecule APE1 inhibitor revealed additive cytotoxicity. These data suggest that in a cellular context, mitoxantrone may interfere with APE1 DNA repair functions.
Collapse
Affiliation(s)
- Irina G Minko
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR 97239, USA
| | - Samantha A Moellmer
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR 97239, USA
| | - Michael M Luzadder
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR 97239, USA
| | - Rachana Tomar
- Department of Chemistry and the Vanderbilt-Ingram Cancer Center, Vanderbilt University, Station B Box 351822, Nashville, TN 37235, USA
| | - Michael P Stone
- Department of Chemistry and the Vanderbilt-Ingram Cancer Center, Vanderbilt University, Station B Box 351822, Nashville, TN 37235, USA
| | - Amanda K McCullough
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR 97239, USA; Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA
| | - R Stephen Lloyd
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR 97239, USA; Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
2
|
Reyes ME, Ma J, Grove ML, Ater JL, Morrison AC, Hildebrandt MAT. RNA sequence analysis of inducible pluripotent stem cell-derived cardiomyocytes reveals altered expression of DNA damage and cell cycle genes in response to doxorubicin. Toxicol Appl Pharmacol 2018; 356:44-53. [PMID: 30031762 DOI: 10.1016/j.taap.2018.07.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 06/29/2018] [Accepted: 07/18/2018] [Indexed: 02/03/2023]
Abstract
Anthracyclines, such as doxorubicin, are highly effective chemotherapeutic agents, yet are associated with increased risk of cardiotoxicity. The genes and pathways involved in the development of heart damage following doxorubicin exposure in humans remain elusive. Our objective was to explore time- and dose-dependent changes in gene expression via RNA sequence (RNAseq) that mediate doxorubicin response in human iPSC-cardiomyocytes following 50, 150, or 450 nM exposure for 2, 7, or 12 days. Clustering and differential expression analyses were conducted to identify genes with altered expression. Samples clustered in dose and time-dependent manners, and MCM5, PRC1, NUSAP1, CENPF, CCNB1, MELK, AURKB, and RACGAP1 were consistently significantly differentially expressed between untreated and treated conditions. These genes were also significantly downregulated in pairwise analyses, which was validated by reverse transcription polymerase chain reaction (RT-PCR). Pathway analysis identified the top canonical pathways involved in response, implicating DNA damage repair response and the cell cycle as having roles in the development of doxorubicin-induced cardiotoxicity in the human cardiomyocyte.
Collapse
Affiliation(s)
- Monica E Reyes
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jianzhong Ma
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Megan L Grove
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Joann L Ater
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Michelle A T Hildebrandt
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
3
|
Systematic Identification of MCU Modulators by Orthogonal Interspecies Chemical Screening. Mol Cell 2017; 67:711-723.e7. [PMID: 28820965 DOI: 10.1016/j.molcel.2017.07.019] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/05/2017] [Accepted: 07/19/2017] [Indexed: 12/31/2022]
Abstract
The mitochondrial calcium uniporter complex is essential for calcium (Ca2+) uptake into mitochondria of all mammalian tissues, where it regulates bioenergetics, cell death, and Ca2+ signal transduction. Despite its involvement in several human diseases, we currently lack pharmacological agents for targeting uniporter activity. Here we introduce a high-throughput assay that selects for human MCU-specific small-molecule modulators in primary drug screens. Using isolated yeast mitochondria, reconstituted with human MCU, its essential regulator EMRE, and aequorin, and exploiting a D-lactate- and mannitol/sucrose-based bioenergetic shunt that greatly minimizes false-positive hits, we identify mitoxantrone out of more than 600 clinically approved drugs as a direct selective inhibitor of human MCU. We validate mitoxantrone in orthogonal mammalian cell-based assays, demonstrating that our screening approach is an effective and robust tool for MCU-specific drug discovery and, more generally, for the identification of compounds that target mitochondrial functions.
Collapse
|
4
|
Chaudhari U, Nemade H, Wagh V, Gaspar JA, Ellis JK, Srinivasan SP, Spitkovski D, Nguemo F, Louisse J, Bremer S, Hescheler J, Keun HC, Hengstler JG, Sachinidis A. Identification of genomic biomarkers for anthracycline-induced cardiotoxicity in human iPSC-derived cardiomyocytes: an in vitro repeated exposure toxicity approach for safety assessment. Arch Toxicol 2015; 90:2763-2777. [PMID: 26537877 PMCID: PMC5065579 DOI: 10.1007/s00204-015-1623-5] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Accepted: 10/20/2015] [Indexed: 11/25/2022]
Abstract
The currently available techniques for the safety evaluation of candidate drugs are usually cost-intensive and time-consuming and are often insufficient to predict human relevant cardiotoxicity. The purpose of this study was to develop an in vitro repeated exposure toxicity methodology allowing the identification of predictive genomics biomarkers of functional relevance for drug-induced cardiotoxicity in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). The hiPSC-CMs were incubated with 156 nM doxorubicin, which is a well-characterized cardiotoxicant, for 2 or 6 days followed by washout of the test compound and further incubation in compound-free culture medium until day 14 after the onset of exposure. An xCELLigence Real-Time Cell Analyser was used to monitor doxorubicin-induced cytotoxicity while also monitoring functional alterations of cardiomyocytes by counting of the beating frequency of cardiomyocytes. Unlike single exposure, repeated doxorubicin exposure resulted in long-term arrhythmic beating in hiPSC-CMs accompanied by significant cytotoxicity. Global gene expression changes were studied using microarrays and bioinformatics tools. Analysis of the transcriptomic data revealed early expression signatures of genes involved in formation of sarcomeric structures, regulation of ion homeostasis and induction of apoptosis. Eighty-four significantly deregulated genes related to cardiac functions, stress and apoptosis were validated using real-time PCR. The expression of the 84 genes was further studied by real-time PCR in hiPSC-CMs incubated with daunorubicin and mitoxantrone, further anthracycline family members that are also known to induce cardiotoxicity. A panel of 35 genes was deregulated by all three anthracycline family members and can therefore be expected to predict the cardiotoxicity of compounds acting by similar mechanisms as doxorubicin, daunorubicin or mitoxantrone. The identified gene panel can be applied in the safety assessment of novel drug candidates as well as available therapeutics to identify compounds that may cause cardiotoxicity.
Collapse
Affiliation(s)
- Umesh Chaudhari
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Str. 39, 50931, Cologne, NRW, Germany
| | - Harshal Nemade
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Str. 39, 50931, Cologne, NRW, Germany
| | - Vilas Wagh
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Str. 39, 50931, Cologne, NRW, Germany
| | - John Antonydas Gaspar
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Str. 39, 50931, Cologne, NRW, Germany
| | - James K Ellis
- Biomolecular Medicine, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Sureshkumar Perumal Srinivasan
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Str. 39, 50931, Cologne, NRW, Germany
| | - Dimitry Spitkovski
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Str. 39, 50931, Cologne, NRW, Germany
| | - Filomain Nguemo
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Str. 39, 50931, Cologne, NRW, Germany
| | - Jochem Louisse
- Institute for Health and Consumer Protection, Joint Research Centre, European Commission, Ispra, Italy
| | - Susanne Bremer
- Institute for Health and Consumer Protection, Joint Research Centre, European Commission, Ispra, Italy
| | - Jürgen Hescheler
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Str. 39, 50931, Cologne, NRW, Germany
| | - Hector C Keun
- Biomolecular Medicine, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Jan G Hengstler
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), 44139, Dortmund, Germany
| | - Agapios Sachinidis
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Str. 39, 50931, Cologne, NRW, Germany.
| |
Collapse
|
5
|
Ghanbarzadeh S, Arami S, Pourmoazzen Z, Ghasemian-Yadegari J, Khorrami A. Plasma stable, pH-sensitive fusogenic polymer-modified liposomes: A promising carrier for mitoxantrone. J Biomater Appl 2013; 29:81-92. [PMID: 24336315 DOI: 10.1177/0885328213515288] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
pH-sensitive liposomes are designed to undergo acid-triggered destabilization. In the present study, we prepared polymer-modified, plasma stable, pH-sensitive fusogenic mitoxantrone liposomes to increase efficacy and selectivity on cancer cell lines. Conventional liposomes were prepared using cholesterol and dipalmitoyl-sn-glycero-3-phosphatidylethanolamine. Dioleoylphosphatidylethanolamine and a cholesteryl derivative, poly(monomethylitaconate)-co-poly(N,N-dimethylaminoethyl methacrylate) (PMMI-co-PDMAEMA), were used for the preparation of pH-sensitive fusogenic liposomes. Using polyethylene glycol (PEG)-poly(monomethylitaconate)-CholC6 (PEG-PMMI-CholC6) copolymers instead of cholesterol introduced pH-sensitive and plasma stability properties simultaneously in prepared liposomes. All formulations were prepared by thin film hydration method and subsequently, pH-sensitivity and stability in human serum were evaluated. The ability of pH-sensitive fusogenic liposomes to enhance the mitoxantrone cytotoxicity and selectivity in cancerous cell lines was assessed in vitro compared to normal cell line using human breast cancer cell line (MCF-7), human prostate cancer cell line (PC-3), and human umbilical vein endothelial cells line. Results revealed that both PMMI-co-PDMAEMA and PEG-PMMI-CholC6-based formulations showed pH-sensitive property and were found to rapidly release mitoxantrone under mildly acidic conditions. Nevertheless, only the PEG-PMMI-CholC6-based liposomes preserved pH-sensitivity after incubation in plasma. Mitoxantrone loaded-pH-sensitive fusogenic liposomes exhibited a higher cytotoxicity than the control conventional liposomes on MCF-7 and PC-3 cell lines. On the contrary, both pH-sensitive fusogenic liposomes showed lower cytotoxic effect on human umbilical vein endothelial cell line. Plasma stable, pH-sensitive fusogenic liposomes are promising carriers for enhancing the efficiency and selectivity, besides reduction of the side effects of anticancer agents.
Collapse
Affiliation(s)
- Saeed Ghanbarzadeh
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran Student Research Committee, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sanam Arami
- Student Research Committee, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran Pharmaceutical Biotechnology Department, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zhaleh Pourmoazzen
- Chemistry Department, Science Faculty, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Javad Ghasemian-Yadegari
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran Department of Pharmacognosy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Arash Khorrami
- Student Research Committee, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
6
|
Falk IJ, Fyrberg A, Paul E, Nahi H, Hermanson M, Rosenquist R, Höglund M, Palmqvist L, Stockelberg D, Wei Y, Gréen H, Lotfi K. Decreased survival in normal karyotype AML with single-nucleotide polymorphisms in genes encoding the AraC metabolizing enzymes cytidine deaminase and 5'-nucleotidase. Am J Hematol 2013; 88:1001-6. [PMID: 23873772 DOI: 10.1002/ajh.23549] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 07/10/2013] [Accepted: 07/15/2013] [Indexed: 11/09/2022]
Abstract
De novo acute myeloid leukemia with normal karyotype (NK-AML) comprises a large group of patients with no common cytogenetic alterations and with a large variation in treatment response. Single-nucleotide polymorphisms (SNPs) in genes related to the metabolism of the nucleoside analogue AraC, the backbone in AML treatment, might affect drug sensitivity and treatment outcome. Therefore, SNPs may serve as prognostic biomarkers aiding clinicians in individualized treatment decisions, with the aim of improving patient outcomes. We analyzed polymorphisms in genes encoding cytidine deaminase (CDA 79A>C rs2072671 and -451C>T rs532545), 5'-nucleotidase (cN-II 7A>G rs10883841), and deoxycytidine kinase (DCK 3'UTR 948T>C rs4643786) in 205 de novo NK-AML patients. In FLT3-internal tandem duplication (ITD)-positive patients, the CDA 79C/C and -451T/T genotypes were associated with shorter overall survival compared to other genotypes (5 vs. 24 months, P < 0.001 and 5 vs. 23 months, P = 0.015, respectively), and this was most pronounced in FLT3-ITD-positive/NPM1-positive patients. We observed altered in vitro sensitivity to topoisomerase inhibitory drugs, but not to nucleoside analogues, and a decrease in global DNA methylation in cells carrying both CDA variant alleles. A shorter survival was also observed for the cN-II variant allele, but only in FLT3-ITD-negative patients (25 vs. 31 months, P = 0.075). Our results indicate that polymorphisms in genes related to nucleoside analog drug metabolism may serve as prognostic markers in de novo NK-AML.
Collapse
Affiliation(s)
- Ingrid Jakobsen Falk
- Clinical Pharmacology, Department of Medical and Health Sciences, Faculty of Health SciencesLinköping UniversityLinköping Sweden
| | - Anna Fyrberg
- Faculty of Health Sciences, Centre for Biomedical ResourcesLinköping UniversityLinköping Sweden
| | - Esbjörn Paul
- Department of Medicine Division of HematologyKarolinska InstitutetHuddinge Stockholm Sweden
| | - Hareth Nahi
- Department of Medicine Division of HematologyKarolinska InstitutetHuddinge Stockholm Sweden
| | - Monica Hermanson
- Department of Immunology, Genetics and Pathology, Rudbeck LaboratoryUppsala UniversityUppsala Sweden
| | - Richard Rosenquist
- Department of Immunology, Genetics and Pathology, Rudbeck LaboratoryUppsala UniversityUppsala Sweden
| | - Martin Höglund
- Division of Hematology Department of Medical SciencesUppsala UniversityUppsala Sweden
| | - Lars Palmqvist
- Department of Clinical Chemistry and Transfusion MedicineInstitute of BiomedicineUniversity of GothenburgGothenburg Sweden
| | - Dick Stockelberg
- Department of internal medicine, Section for Hematology and CoagulationSahlgrenska University HospitalGothenburg Sweden
| | - Yuan Wei
- Department of internal medicine, Section for Hematology and CoagulationSahlgrenska University HospitalGothenburg Sweden
| | - Henrik Gréen
- Clinical Pharmacology, Department of Medical and Health Sciences, Faculty of Health SciencesLinköping UniversityLinköping Sweden
- Science for Life LaboratoryKTH Royal Institute of TechnologySchool of BiotechnologySolna Sweden
- Department of Forensic Genetics and Forensic Toxicology, National Board of Forensic MedicineLinköping Sweden
| | - Kourosh Lotfi
- Clinical Pharmacology, Department of Medical and Health Sciences, Faculty of Health SciencesLinköping UniversityLinköping Sweden
- Department of HematologyLinköping University HospitalLinköping Sweden
| |
Collapse
|
7
|
Wan X, Zhang W, Li L, Xie Y, Li W, Huang N. A new target for an old drug: identifying mitoxantrone as a nanomolar inhibitor of PIM1 kinase via kinome-wide selectivity modeling. J Med Chem 2013; 56:2619-29. [PMID: 23442188 DOI: 10.1021/jm400045y] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The rational design of selective kinase inhibitors remains a great challenge. Here we describe a physics-based approach to computationally modeling the kinase inhibitor selectivity profile. We retrospectively assessed this protocol by computing the binding profiles of 17 well-known kinase inhibitors against 143 kinases. Next, we predicted the binding profile of the chemotherapy drug mitoxantrone, and chose the predicted top five kinase targets for in vitro kinase assays. Remarkably, mitoxantrone was shown to possess low nanomolar inhibitory activity against PIM1 kinase and to inhibit the PIM1-mediated phosphorylation in cancer cells. We further determined the crystal complex structure of PIM1 bound with mitoxantrone, which reveals the structural and mechanistic basis for a novel mode of PIM1 inhibition. Although mitoxantrone's mechanism of action had been originally thought to act through DNA intercalation and type II topoisomerase inhibition, we hypothesize that PIM1 kinase inhibition might also contribute to mitoxantrone's therapeutic efficacy and specificity.
Collapse
Affiliation(s)
- Xiaobo Wan
- Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | | | | | | | | | | |
Collapse
|
8
|
Physicochemical Mechanisms of Synergistic Biological Action of Combinations of Aromatic Heterocyclic Compounds. ACTA ACUST UNITED AC 2013. [DOI: 10.1155/2013/278143] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The mechanisms of synergistic biological effects observed in the simultaneous use of aromatic heterocyclic compounds in combination are reviewed, and the specific biological role of heteroassociation of aromatic molecules is discussed.
Collapse
|
9
|
Association of ABCB1 polymorphisms with survival and in vitro cytotoxicty in de novo acute myeloid leukemia with normal karyotype. THE PHARMACOGENOMICS JOURNAL 2010; 12:111-8. [PMID: 20938465 DOI: 10.1038/tpj.2010.79] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
10
|
Paul E, Paul E, Uggla B, Deneberg S, Bengtzen S, Hermansson M, Dahlman I, Rosenquist R, Wiman KG, Nahi H. Low p14ARF expression inde novoacute myeloid leukemia with normal karyotype is associated with poor survival. Leuk Lymphoma 2009; 50:1512-8. [DOI: 10.1080/10428190903111914] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
11
|
Bozdağ S, Capan Y, Vural I, Dalkara T, Dogan AL, Guc D, Hincal AA, Deluca PP. In vitrocytotoxicity of mitoxantrone-incorporated albumin microspheres on acute promyelocytic leukaemia cells. J Microencapsul 2008; 21:751-60. [PMID: 15799225 DOI: 10.1080/02652040400000553] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
In the present study, the preparation and characterization of bovine serum albumin (BSA) microspheres and the evaluation of the in vitro cytotoxicity of these microspheres on acute promyelocytic leukaemia (HL-60) cells were described. Mitoxantrone (MTZ)-incorporated microspheres were evaluated for particle size, drug loading, release characteristics and surface morphology. The biological effect of MTZ released from BSA microspheres was determined on an in vitro cultured HL-60 cell line, showing that, after encapsulation, MTZ still retains cytotoxic activity. For this purpose, methyl-thiazol-tetrazolium (MTT) assay was used to evaluate the in vitro cytotoxicity of MTZ-loaded microspheres. Particle size of BSA microspheres was determined between 17.61-20.38 microm and they were smooth and spherical in shape. Encapsulation efficiency of the drug-loaded microspheres was between 22.26-60.50%. For MTZ-containing microspheres, the cell death ratios were greater than 80% for all formulations. This study demonstrate that BSA microspheres were well suited for the controlled release of MTZ and were promising for anti-cancer chemotherapy.
Collapse
Affiliation(s)
- S Bozdağ
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Hacettepe University, Ankara, Turkey
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Nahi H, Lehmann S, Bengtzen S, Jansson M, Möllgård L, Paul C, Merup M. Chromosomal aberrations in 17p predict in vitro drug resistance and short overall survival in acute myeloid leukemia. Leuk Lymphoma 2008; 49:508-16. [PMID: 18297528 DOI: 10.1080/10428190701861645] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Chromosomal aberrations are important prognostic parameters in acute myeloid leukemia (AML). Indicators of poor prognosis include del(5q)/-5, del(7q)/-7, abnormal 3q or complex karyotype. In recent years, it has become clear that aberrations in 17p represent one of the indicators of poor prognosis in haematological malignancies. In AML, deletions in 17p have been shown to indicate a dismal prognosis; genetic aberrations in 9p have also been discussed as influencing long-term survival in AML. In this study, we correlated genetic abnormalities in chromosomes 9 and 17 in patients with de novo AML to in vitro cytotoxicity of conventional anti-leukemic drugs, and long-term overall survival. Blast cells were isolated from 387 patients diagnosed with AML. Chromosomal analysis was successful in 336 cases. All samples were tested for in vitro cytotoxicity against fludarabine, amsacrine, mitoxantrone, etoposide, daunorubicin and Ara-C after being cultured for 4 days, using an ATP assay. Among the 336 patients, five main groups were identified. Abnormal chromosome 17 (n = 22), abnormal 9p (n = 13), monosomy 7 or deletion 7q (n = 35), complex karyotype (n = 52) and normal karyotype (n = 132). Patients with abnormalities of chromosome 17 showed significantly greater resistance to all drugs tested and significantly shorter overall survival compared with patients with normal and complex karyotypes (p = 0.0001 and 0.041, respectively). All patients with abnormalities of chromosome 17 died within 11 months of diagnosis. A tendency towards shorter overall survival and greater drug resistance was also noted when comparing chromosome 17 abnormalities with del(7q)/-7, but the differences did not reach statistical significance. Patients with abnormal 9p showed significantly shorter overall survival but did not differ significantly as regards in vitro drug resistance compared with patients presenting with a normal karyotype. Chromosomal abnormalities affecting the p53 pathway have a significant impact on cytostatic drug resistance and survival in AML. Developing new drugs targeting the p53 pathway could be a way to improve treatment of AML.
Collapse
Affiliation(s)
- Hareth Nahi
- Department of Haematology, Karolinska Institute at Huddinge University Hospital, Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|
13
|
Quantitation of the molecular mechanisms of biological synergism in a mixture of DNA-acting aromatic drugs. Biophys Chem 2008; 132:148-58. [DOI: 10.1016/j.bpc.2007.11.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2007] [Revised: 11/04/2007] [Accepted: 11/05/2007] [Indexed: 01/01/2023]
|
14
|
Nahi H, Merup M, Lehmann S, Bengtzen S, Möllgård L, Selivanova G, Wiman KG, Paul C. PRIMA-1 induces apoptosis in acute myeloid leukaemia cells with p53 gene deletion. Br J Haematol 2006; 132:230-6. [PMID: 16398657 DOI: 10.1111/j.1365-2141.2005.05851.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The p53 tumour suppressor gene located on chromosome 17p13 is the most frequently mutated gene in human tumours. About 5-8% of cases with acute myeloid leukaemia (AML) carry the p53 mutation. Recently, the compound p53-dependent reactivation and induction of massive apoptosis (PRIMA-1) has been shown to induce cytotoxic effects and apoptosis in human tumour cells by restoration of the transcriptional activity of mutated p53. This is believed to be mediated by a change in the conformation of mutated p53 protein, restoring DNA binding and activation of p53 target genes. We studied the effects of PRIMA-1 and commonly used antileukaemic drugs on AML cells from 62 patients. Cells were obtained from peripheral blood or bone marrow and were exposed to PRIMA-1, cytarabine, daunorubicin, chlorodeoxyadenosine and fludarabine. This study showed that PRIMA-1 had cytotoxic effects on AML cells. Conventional AML drugs were less effective in cells with hemizygous p53 deletion. Interestingly, our data indicated that PRIMA-1 was more effective in this subgroup of patients compared with patients with normal chromosome 17. Our data suggest that the concept of restoration of p53 protein by PRIMA-1 or a PRIMA-1-based new drug may increase the efficacy of AML treatment in patients with p53 mutations.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- Adult
- Aged
- Aged, 80 and over
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Aza Compounds/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Cell Survival/drug effects
- Dose-Response Relationship, Drug
- Drug Resistance, Neoplasm
- Female
- Gene Deletion
- Genes, p53
- Humans
- In Situ Hybridization, Fluorescence
- Karyotyping
- Leukemia, Myeloid/genetics
- Leukemia, Myeloid/metabolism
- Leukemia, Myeloid/pathology
- Male
- Middle Aged
- Neoplasm Proteins/metabolism
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- H Nahi
- Department of Haematology, Karolinska University Hospital at Huddinge, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Potter AJ, Rabinovitch PS. The cell cycle phases of DNA damage and repair initiated by topoisomerase II-targeting chemotherapeutic drugs. Mutat Res 2005; 572:27-44. [PMID: 15790488 DOI: 10.1016/j.mrfmmm.2004.11.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2004] [Revised: 11/09/2004] [Accepted: 11/22/2004] [Indexed: 10/25/2022]
Abstract
Although cytostasis and cytotoxicity induced by cancer chemotherapy drugs targeting topoisomerase II (topoII) arise in specific cell cycle phases, it is unknown whether the drug-initiated DNA damage triggering these responses, or the repair (reversal) of this damage, differs between cell cycle phases or between drug classes. Accordingly, we used a flow cytometric alkaline unwinding assay to measure DNA damage (strand breakage (SB)) and SB repair in each cell cycle compartment of human cancer cell lines treated with clinically relevant concentrations of doxorubicin, daunomycin, etoposide, and mitoxantrone. We found that treated HeLa and A549 cells exhibited the greatest SB in G2/M phase, the least in G1 phase, and generally an intermediate amount in S phase. The cell cycle phase specificity of the DNA damage appeared to be predictive of the cell cycle phase of growth arrest. Furthermore, it appeared to be dependent on topoIIalpha expression as the extent of SB did not differ between cell cycle compartments in topoIIalpha-diminished A549(VP)28 cells. HeLa cells were apparently unable to repair doxorubicin-initiated SB. The rate of repair of etoposide-initiated SB in HeLa cells and of mitoxantrone-initiated SB in HeLa and A549 cells was similar in each cell cycle compartment. In A549 cells, the rate of repair of doxorubicin and etoposide-initiated SB differed between cell cycle phases. Overall, these results indicate that the cell cycle phase specificity of cytostasis and cytotoxicity induced in tumor cells by topoII-targeting drugs may be directly related to the cell cycle phase specificity of the drug-initiated DNA damage. Analysis by cell cycle compartment appears to clarify some of the intercellular heterogeneity in the extent of drug-initiated DNA damage and cytotoxicity previously observed in cancer cells analyzed as a single population; this approach might be useful in resolving inconsistent results reported in investigations of tumor cell topoII content versus response to topoII-targeting drugs.
Collapse
Affiliation(s)
- Alan J Potter
- Department of Pathology, Box 357705, University of Washington, Seattle, WA 98195, USA
| | | |
Collapse
|
16
|
Nahi H, Lehmann S, Mollgard L, Bengtzen S, Selivanova G, Wiman KG, Paul C, Merup M. Effects of PRIMA-1 on chronic lymphocytic leukaemia cells with and without hemizygous p53
deletion. Br J Haematol 2004; 127:285-91. [PMID: 15491287 DOI: 10.1111/j.1365-2141.2004.05210.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The tumour suppressor gene p53 is the most commonly mutated gene in solid tumours. Although less common in haematological malignancies, 10-15% of B-cell chronic lymphocytic leukaemia (B-CLL) cases carry a p53 mutation. Recently, the compound P53-dependent reactivation and induction of massive apoptosis (PRIMA-1) has been shown to induce cytotoxic effects and apoptosis in human tumour cells by restoration of the transcriptional activity of mutated p53. This is believed to be mediated by a change in the conformation of mutated p53 protein, restoring DNA binding and activation of p53 target genes. We studied the effects of PRIMA-1 and commonly used anti-leukaemic drugs on B-CLL cells from 14 patients with and without hemizygous p53 deletion. Cells obtained from peripheral blood or bone marrow were exposed to PRIMA-1 and fludarabine alone or in combination. PRIMA-1 showed cytotoxic effects on B-CLL cells from samples with and without hemizygous p53 deletion. Furthermore, conventional B-CLL drugs were less effective in cell samples with hemizygous p53 deletion and the response depended on the size of the p53 deleted clone. Finally, we found evidence for synergistic and additive effects of PRIMA-1 in combination with fludarabine.
Collapse
Affiliation(s)
- H Nahi
- Department of Haematology, Karolinska University Hospital at Huddinge, Stockholm, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Lehmann S, Bengtzen S, Paul A, Christensson B, Paul C. Effects of arsenic trioxide (As2O3) on leukemic cells from patients with non-M3 acute myelogenous leukemia: studies of cytotoxicity, apoptosis and the pattern of resistance. Eur J Haematol 2001; 66:357-64. [PMID: 11488934 DOI: 10.1034/j.1600-0609.2001.066006357.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Arsenic oxide (As2O3) has recently been reported to induce remission in a high percentage of patients with acute promyelocytic leukemia (APL). The aim of this study was to investigate the effects of As2O3 at therapeutic concentrations on cell viability and apoptosis on leukemic cells from patients with non-M3 acute myelogenous leukemia (AML) and to study the resistance profile compared to conventional AML drugs. Cells from 20 patients were exposed to therapeutic concentrations of As2O3 continuously for 96 h. As2O3 reduced the viability in blast cells from all the 20 tested patients compared to unexposed controls (p-value: 0.02 at 0.05 microM; <0.005 at 1.0 microM and higher concentrations). An increase in the apoptotic rate was also seen after incubation with As2O3. Parallel to the incubation with arsenic the in vitro sensitivity to a number of chemotherapeutic agents commonly used in AML was studied. Correlation coefficients for the in vitro sensitivity were highly significant between the conventional AML drugs except for Ara-C. For As2O3, all the correlation coefficients were negative and ranged between -0.05 and -0.51. Furthermore, increased P-gp expression in a multidrug resistant HL-60 cell line did not decrease the sensitivity to As2O3 as compared to the parental cell line. Neither did a P-gp-transfected variant of the K562 cell line show decreased sensitivity to As2O3. We conclude that As2O3 at therapeutic concentrations induces apoptosis and cytotoxic effects in blast cells from patients with non-M3 AML, and that As2O3 differs from conventional AML drugs with respect to the mechanisms that confer resistance to the drugs.
Collapse
Affiliation(s)
- S Lehmann
- Department of Hematology, Huddinge University Hospital, Karolinska Institute, Huddinge, Sweden.
| | | | | | | | | |
Collapse
|
18
|
Skladanowski A, Konopa J. Mitoxantrone and ametantrone induce interstrand cross-links in DNA of tumour cells. Br J Cancer 2000; 82:1300-4. [PMID: 10755405 PMCID: PMC2374491 DOI: 10.1054/bjoc.1999.1095] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We show here that mitoxantrone and ametantrone induce interstrand DNA cross-links in HeLa S3 cells. These cross-links were observed only in cellular system suggesting that metabolism of the drugs is a necessary step leading to DNA cross-linking. Biologically inactive analogue of mitoxantrone, compound NSC 321458, did not induce cross-links in DNA of tumour cells which suggests that DNA cross-linking is associated with the cytotoxic and anti-tumour activity of these compounds.
Collapse
Affiliation(s)
- A Skladanowski
- Department of Pharmaceutical Technology and Biochemistry, Technical University of Gdanśk, Poland
| | | |
Collapse
|
19
|
Möllgård L, Tidefelt U, Sundman-Engberg B, Löfgren C, Lehman S, Paul C. High single dose of mitoxantrone and cytarabine in acute non-lymphocytic leukemia: a pharmacokinetic and clinical study. Ther Drug Monit 1998; 20:640-5. [PMID: 9853980 DOI: 10.1097/00007691-199812000-00010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
In a phase I-II study, the authors evaluated the intracellular pharmacokinetics, toxicity, and efficiency of a high dose of mitoxantrone given as first induction in acute non-lymphocytic leukemia. Twenty-two patients with previously untreated de novo ANLL were included and received 30 or 40 mg/m2 mitoxantrone on day 1 by intravenous infusion over 1 hour and 500 mg/m2 ara-C twice a day for 5 days. If there was no complete remission (CR), a second induction with ara-C, etoposide, and amsacrine was given. The CR rate after two courses with this regimen was 77%. Median duration of severe neutropenia was 18 days in the 30-mg/m2 group and 25 days in the 40-mg/m2 group. Two patients had fatal lung complications probably unrelated to mitoxantrone. A third patient had a possible mitoxantrone-induced reversible lung complication. In the leukemic cells, we found a high accumulation of mitoxantrone which, in contrast to the plasma concentration, remained stable during the 48 hours studied. Compared with previous results with 12 mg/m2 mitoxantrone, the AUC for intracellular concentrations versus time for the first 20 hours studied was increased by 150% to 0.638 nmol/mg cell protein x hour with 30 mg/m2 mitoxantrone and by 260% to 1.103 nmol/mg cell protein x hour with 40 mg/m2 mitoxantrone. In conclusion, a high dose of mitoxantrone results in a high intracellular exposure of the leukemic cells, which may be an advantage in improving survival of these patients.
Collapse
Affiliation(s)
- L Möllgård
- Department of Medicine, Orebro Medical Centre Hospital, Sweden
| | | | | | | | | | | |
Collapse
|
20
|
Schopman EM, Van Bree C, Bakker PJ, Kipp JB, Barendsen GW. Hyperthermia-enhanced effectiveness of mitoxantrone in an experimental rat tumour. Int J Hyperthermia 1996; 12:241-54. [PMID: 8926392 DOI: 10.3109/02656739609022512] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The influence of local hyperthermia (HT) on Mitoxantrone (MITOX) effectiveness was studied in an experimental rat tumour. R-1 rhabdomyosarcomas were treated with MITOX (5 mg/kg ip), HT (43 degrees C for 1 h) or combinations applied at various time intervals up to 24 h. Tumour growth delay and tumour cell clonogenicity were assessed in correlation with the pharmacokinetics in blood plasma and with MITOX-concentrations in tumour tissue. Combined treatments were more effective than expected on the basis of simple addition of effects of single treatments. With increasing time intervals between treatments up to 8 h, an increase in effectiveness was observed. Unfortunately, treatment with an 8-h interval resulted in a high mortality: 80% of the rats died with 5-10 days after treatment. Treatment with a 3-h interval between MITOX and HT was the most effective combination resulting in the highest therapeutic ratio. Even local tumour controls (14/18 rats) were observed. These enhanced effects were associated with a higher MITOX-concentration in the fraction of intact cells recovered from tumours. However, no differences were observed in MITOX-concentration in total tumour tissue nor in plasma concentrations. In conclusion, timing between MITOX and HT is important for drug availability, for interaction of the two modalities to increase damage in tumour cells and for limiting the toxicity to normal tissues.
Collapse
Affiliation(s)
- E M Schopman
- Laboratory for Radiobiology, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
21
|
Sundman-Engberg B, Tidefelt U, Paul C. Effect of cytokines on the toxicity of cytostatic drugs on leukemic cells in vitro and in vivo. Eur J Haematol Suppl 1996; 56:1-6. [PMID: 8599980 DOI: 10.1111/j.1600-0609.1996.tb00285.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Most cytostatic drugs exert their effect on cells in active cell cycle. To improve the effect of cytostatic drugs we have tried, prior to treatment in vitro, to recruit tumor cells from G0 with growth factors. Leukemic cells from the bone marrows of 26 patients with AML and CML in blast crisis were incubated with G-CSF, GM-CSF and IL-3 for 24 h prior to incubation with cytostatic drugs. The cells were incubated with mitoxantrone, etoposide or daunorubicin for 1 h, or with Ara-C continuously. Prior to treatment, 4 patients with AML received GM-CSF for 24 h, after which blast cells from bone marrow were incubated with cytostatic drugs. After incubation with the cytostatic drugs, cells were cultured in a suspension culture for 4 d. The drug effect was determined with a bioluminescence ATP method. Leukemic cells were significantly stimulated by all three cytokines compared to an untreated control. GM-CSF and IL-3 increased the amount of cells 3- to 4-fold and G-CSF increased the amount 3 times compared to untreated cells. G-CSF significantly enhanced the cytotoxic effect of daunorubicin, mitoxantrone, etoposide and Ara-C by 20-40%, which GM-CSF and IL-3 showed a significantly increased toxicity for Ara-C only. Although the cytokines induced a higher percentage of cells killed with the cytostatic drugs, proliferation of the remaining cells resulted in an increased total number of cells from 1.5 to 3 times compared to the unstimulated incubations. We conclude that cytokines induce a higher level of toxicity of cytostatic drugs on leukemic cells, but the increased proliferation of the remaining cells may offset the clinical benefit.
Collapse
Affiliation(s)
- B Sundman-Engberg
- Department of Medicine, Karolinska Insitutet, Huddinge Hospital, Sweden
| | | | | |
Collapse
|
22
|
Jönsson K, Dahlberg N, Tidefelt U, Paul C, Andersson G. Characterization of an anthracycline-resistant human promyelocyte leukemia (HL-60) cell line with an elevated MDR-1 gene expression. Biochem Pharmacol 1995; 49:755-62. [PMID: 7702633 DOI: 10.1016/0006-2952(94)00511-j] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Multidrug resistance to a variety of cytotoxic drugs is due to decreased drug accumulation at the intracellular site of drug action. When due to increased energy-dependent drug efflux, this transport change is often associated with increased expression of an efflux pump for various lipophilic compounds, for example the P-glycoprotein which is the product of the MDR-1 gene. However, previously described HL-60 human promyelocytic leukemia cell lines resistant to the cytotoxic effect of anthracyclines have been reported not to express P-glycoprotein. We have isolated, by drug selection, an anthracycline-resistant HL-60 cell line that, in comparison to parental drug sensitive cells, exhibits a multidrug resistant phenotype including diminished intracellular drug retention, cross-resistance to multiple cytotoxic drugs, increased expression of a monoclonal antibody C219-reactive 180 kDa P-glycoprotein detected by Western blot analysis as well as increased expression of MDR-1 mRNA as determined by Northern blot and solution hybridization/RNAse protection analyses. Evidence is presented that the anthracycline-resistant HL-60 cells have amplified the MDR-1 gene.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- Biological Transport
- Cell Separation
- Doxorubicin/metabolism
- Doxorubicin/pharmacology
- Drug Resistance, Multiple/genetics
- Humans
- Leukemia, Promyelocytic, Acute/genetics
- Leukemia, Promyelocytic, Acute/pathology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Tumor Cells, Cultured/drug effects
Collapse
Affiliation(s)
- K Jönsson
- Department of Medicine, Huddinge Hospital, Karolinska Institute, Sweden
| | | | | | | | | |
Collapse
|
23
|
Abstract
An in vitro transcription assay was used to determine the sequence specificity of binding of mitoxantrone to a 497 bp fragment of DNA containing the lac UV5 promoter. Transcriptional blockages of the E. coli RNA polymerase were observed dominantly prior to 5'-CpA sequences (64% occurrence), and to a lesser extent 5'-CpG sequences (29%). Overall, 93% of all blockage sites were prior to pyrimidine (3'-5') purine sequences. An effect of flanking sequences was evident since the blockage sites contained an A/T base pair 5' prior to the consensus CpA and CpG intercalation sites. The consensus sequences for the preferred mitoxantrone intercalation sites are therefore 5'-(A/T)CA and 5'-(A/T)CG. The location of transcriptional blockages one base pair prior to the intercalation site is consistent with the fact that the mitoxantrone side chains lie in the major groove.
Collapse
Affiliation(s)
- C Panousis
- Department of Biochemistry, La Trobe University, Bundoora, Victoria, Australia
| | | |
Collapse
|