Voreloxin Is an Anticancer Quinolone Derivative that Intercalates DNA and Poisons Topoisomerase II

Background Topoisomerase II is critical for DNA replication, transcription and chromosome segregation and is a well validated target of anti-neoplastic drugs including the anthracyclines and epipodophyllotoxins. However, these drugs are limited by common tumor resistance mechanisms and side-effect profiles. Novel topoisomerase II-targeting agents may benefit patients who prove resistant to currently available topoisomerase II-targeting drugs or encounter unacceptable toxicities. Voreloxin is an anticancer quinolone derivative, a chemical scaffold not used previously for cancer treatment. Voreloxin is completing Phase 2 clinical trials in acute myeloid leukemia and platinum-resistant ovarian cancer. This study defined voreloxin's anticancer mechanism of action as a critical component of rational clinical development informed by translational research. Methods/Principal Findings Biochemical and cell-based studies established that voreloxin intercalates DNA and poisons topoisomerase II, causing DNA double-strand breaks, G2 arrest, and apoptosis. Voreloxin is differentiated both structurally and mechanistically from other topoisomerase II poisons currently in use as chemotherapeutics. In cell-based studies, voreloxin poisoned topoisomerase II and caused dose-dependent, site-selective DNA fragmentation analogous to that of quinolone antibacterials in prokaryotes; in contrast etoposide, the nonintercalating epipodophyllotoxin topoisomerase II poison, caused extensive DNA fragmentation. Etoposide's activity was highly dependent on topoisomerase II while voreloxin and the intercalating anthracycline topoisomerase II poison, doxorubicin, had comparable dependence on this enzyme for inducing G2 arrest. Mechanistic interrogation with voreloxin analogs revealed that intercalation is required for voreloxin's activity; a nonintercalating analog did not inhibit proliferation or induce G2 arrest, while an analog with enhanced intercalation was 9.5-fold more potent. Conclusions/Significance As a first-in-class anticancer quinolone derivative, voreloxin is a toposiomerase II-targeting agent with a unique mechanistic signature. A detailed understanding of voreloxin's molecular mechanism, in combination with its evolving clinical profile, may advance our understanding of structure-activity relationships to develop safer and more effective topoisomerase II-targeted therapies for the treatment of cancer.

[1]  Y. Pommier DNA topoisomerase I and II in cancer chemotherapy: update and perspectives , 2004, Cancer Chemotherapy and Pharmacology.

[2]  K. Kreuzer,et al.  A unique type II topoisomerase mutant that is hypersensitive to a broad range of cleavage-inducing antitumor agents. , 2002, Biochemistry.

[3]  A. Maxwell,et al.  Quinolone-DNA Interaction: Sequence-Dependent Binding to Single-Stranded DNA Reflects the Interaction within the Gyrase-DNA Complex , 2003, Antimicrobial Agents and Chemotherapy.

[4]  A. Guo,et al.  Delineating the contribution of secretory transporters in the efflux of etoposide using Madin-Darby canine kidney (MDCK) cells overexpressing P-glycoprotein (Pgp), multidrug resistance-associated protein (MRP1), and canalicular multispecific organic anion transporter (cMOAT). , 2002, Drug metabolism and disposition: the biological fate of chemicals.

[5]  D. Gewirtz,et al.  A critical evaluation of the mechanisms of action proposed for the antitumor effects of the anthracycline antibiotics adriamycin and daunorubicin. , 1999, Biochemical pharmacology.

[6]  J. Champoux DNA topoisomerases: structure, function, and mechanism. , 2001, Annual review of biochemistry.

[7]  N. Osheroff,et al.  Etoposide, topoisomerase II and cancer. , 2005, Current medicinal chemistry. Anti-cancer agents.

[8]  J. Doroshow,et al.  Oxidative DNA base modifications in peripheral blood mononuclear cells of patients treated with high-dose infusional doxorubicin. , 2001, Blood.

[9]  D. Taatjes,et al.  Nuclear targeting and retention of anthracycline antitumor drugs in sensitive and resistant tumor cells. , 2001, Current medicinal chemistry.

[10]  N. Osheroff,et al.  DNA topoisomerase II, genotoxicity, and cancer. , 2007, Mutation research.

[11]  N. Osheroff,et al.  DNA topoisomerase protocols , 1999 .

[12]  L. Mitscher Bacterial topoisomerase inhibitors: quinolone and pyridone antibacterial agents. , 2005, Chemical reviews.

[13]  A. Bodley,et al.  DNA topoisomerase II-mediated interaction of doxorubicin and daunorubicin congeners with DNA. , 1989, Cancer research.

[14]  J M Berger,et al.  Recent advances in understanding structure-function relationships in the type II topoisomerase mechanism. , 2005, Biochemical Society transactions.

[15]  JAMES C. Wang,et al.  Cellular roles of DNA topoisomerases: a molecular perspective , 2002, Nature Reviews Molecular Cell Biology.

[16]  Mark F. Lutz,et al.  Monitoring of Anthracycline-Induced Cardiotoxicity , 2008, The Annals of pharmacotherapy.

[17]  S. Cutts,et al.  DNA repair in response to anthracycline-DNA adducts: a role for both homologous recombination and nucleotide excision repair. , 2008, Mutation research.

[18]  J. Wang,et al.  Inducible overexpression, purification, and active site mapping of DNA topoisomerase II from the yeast Saccharomyces cerevisiae. , 1989, The Journal of biological chemistry.

[19]  Xilin Zhao,et al.  Quinolone-Mediated Bacterial Death , 2007, Antimicrobial Agents and Chemotherapy.

[20]  T. Yamaoka,et al.  Amrubicin, a novel 9‐aminoanthracycline, enhances the antitumor activity of chemotherapeutic agents against human cancer cells in vitro and in vivo , 2007, Cancer science.

[21]  M. Griffiths Handbook of cancer chemotherapy , 1996 .

[22]  D. Crothers,et al.  Characterization of covalent adriamycin-DNA adducts. , 1998, Proceedings of the National Academy of Sciences of the United States of America.

[23]  G. Mueller,et al.  Simple procedure for isolation of DNA, RNA and protein fractions from cultured animal cells. , 1975, Analytical biochemistry.

[24]  M. Caligiuri,et al.  Dose escalation studies of cytarabine, daunorubicin, and etoposide with and without multidrug resistance modulation with PSC-833 in untreated adults with acute myeloid leukemia younger than 60 years: final induction results of Cancer and Leukemia Group B Study 9621. , 2004, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[25]  D. J. Clarke,et al.  A topoisomerase II-dependent G2 cycle checkpoint in mammalian cells , 1994, Nature.

[26]  J. Nitiss DNA topoisomerase II and its growing repertoire of biological functions , 2009, Nature Reviews Cancer.

[27]  A. Buzdar,et al.  Early and delayed clinical cardiotoxicity of doxorubicin , 1985, Cancer.

[28]  Yukiko Nakamura,et al.  Efficacy of amrubicin for non-small cell lung cancer after failure of two or more prior chemotherapy regimens. , 2008, Anticancer research.

[29]  D. J. Clarke,et al.  DNA Topoisomerases , 2009, Methods in Molecular Biology™.

[30]  J. Karp,et al.  Phase Ib/II pharmacokinetic/pharmacodynamic (PK/PD) study of combination voreloxin and cytarabine in relapsed or refractory AML patients. , 2009, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[31]  N. Osheroff,et al.  Type II topoisomerases as targets for quinolone antibacterials: turning Dr. Jekyll into Mr. Hyde. , 2001, Current pharmaceutical design.

[32]  L. Liu,et al.  A homogeneous type II DNA topoisomerase from HeLa cell nuclei. , 1981, The Journal of biological chemistry.

[33]  K. Kohn,et al.  Local sequence requirements for DNA cleavage by mammalian topoisomerase II in the presence of doxorubicin. , 1990, Nucleic acids research.

[34]  D. Anderson,et al.  Reactive oxygen species-induced DNA damage and its modification: a chemical investigation. , 1997, Mutation research.

[35]  M. Maris,et al.  A Phase 2 Dose Regimen Optimization Study of Three Schedules of Voreloxin as Single Agent Therapy for Elderly Patients with Newly Diagnosed Acute Myeloid Leukemia. , 2009 .

[36]  A. Nudelman,et al.  Doxorubicin-DNA adducts induce a non-topoisomerase II-mediated form of cell death. , 2006, Cancer research.

[37]  D. Hooper,et al.  Mechanisms of action of antimicrobials: focus on fluoroquinolones. , 2001, Clinical infectious diseases : an official publication of the Infectious Diseases Society of America.

[38]  N. Osheroff,et al.  The geometry of DNA supercoils modulates topoisomerase-mediated DNA cleavage and enzyme response to anticancer drugs. , 2006, Biochemistry.

[39]  M. Palumbo,et al.  Hot-spot consensus of fluoroquinolone-mediated DNA cleavage by Gram-negative and Gram-positive type II DNA topoisomerases , 2007, Nucleic acids research.

[40]  D. Taatjes,et al.  Mass spectrometric measurement of formaldehyde generated in breast cancer cells upon treatment with anthracycline antitumor drugs. , 2000, Chemical research in toxicology.

[41]  E. Kraut,et al.  Analysis of topoisomerase I/DNA complexes in patients administered topotecan. , 1995, Cancer research.

[42]  N. Osheroff,et al.  Topoisomerase II as a target for anticancer drugs: when enzymes stop being nice. , 2000, Progress in nucleic acid research and molecular biology.

[43]  J. Berger,et al.  Structural basis for gate-DNA recognition and bending by type IIA topoisomerases , 2007, Nature.

[44]  W. Earnshaw,et al.  Differential expression of DNA topoisomerases I and II during the eukaryotic cell cycle. , 1988, Proceedings of the National Academy of Sciences of the United States of America.

[45]  L. Gianni,et al.  Anthracyclines: Molecular Advances and Pharmacologic Developments in Antitumor Activity and Cardiotoxicity , 2004, Pharmacological Reviews.

[46]  H. Kim,et al.  Levels of multidrug resistance (MDR1) P-glycoprotein expression by human breast cancer correlate with in vitro resistance to taxol and doxorubicin. , 1998, Clinical cancer research : an official journal of the American Association for Cancer Research.

[47]  J. Nitiss Targeting DNA topoisomerase II in cancer chemotherapy , 2009, Nature Reviews Cancer.

[48]  U. Hoch,et al.  Voreloxin, formerly SNS-595, has potent activity against a broad panel of cancer cell lines and in vivo tumor models , 2009, Cancer Chemotherapy and Pharmacology.