1
|
Novel antimicrobial ciprofloxacin-pyridinium quaternary ammonium salts with improved physicochemical properties and DNA gyrase inhibitory activity. Med Chem Res 2021. [DOI: 10.1007/s00044-021-02798-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
2
|
Molecular mechanisms of apoptosis induced by a novel synthetic quinolinone derivative in HL-60 human leukemia cells. Chem Biol Interact 2020; 320:109005. [DOI: 10.1016/j.cbi.2020.109005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/11/2020] [Accepted: 02/17/2020] [Indexed: 11/23/2022]
|
3
|
Bruno PM, Lu M, Dennis KA, Inam H, Moore CJ, Sheehe J, Elledge SJ, Hemann MT, Pritchard JR. The primary mechanism of cytotoxicity of the chemotherapeutic agent CX-5461 is topoisomerase II poisoning. Proc Natl Acad Sci U S A 2020; 117:4053-4060. [PMID: 32041867 PMCID: PMC7049172 DOI: 10.1073/pnas.1921649117] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Small molecules can affect many cellular processes. The disambiguation of these effects to identify the causative mechanisms of cell death is extremely challenging. This challenge impacts both clinical development and the interpretation of chemical genetic experiments. CX-5461 was developed as a selective RNA polymerase I inhibitor, but recent evidence suggests that it may cause DNA damage and induce G-quadraplex formation. Here we use three complimentary data mining modalities alongside biochemical and cell biological assays to show that CX-5461 exerts its primary cytotoxic activity through topoisomerase II poisoning. We then show that acquired resistance to CX-5461 in previously sensitive lymphoma cells confers collateral resistance to the topoisomerase II poison doxorubicin. Doxorubicin is already a frontline chemotherapy in a variety of hematopoietic malignancies, and CX-5461 is being tested in relapse/refractory hematopoietic tumors. Our data suggest that the mechanism of cell death induced by CX-5461 is critical for rational clinical development in these patients. Moreover, CX-5461 usage as a specific chemical genetic probe of RNA polymerase I function is challenging to interpret. Our multimodal data-driven approach is a useful way to detangle the intended and unintended mechanisms of drug action across diverse essential cellular processes.
Collapse
Affiliation(s)
- Peter M Bruno
- Howard Hughes Medical Institute, Brigham and Women's Hospital, Boston, MA 02115
- Division of Genetics, Brigham and Women's Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
| | - Mengrou Lu
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802
| | - Kady A Dennis
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802
| | - Haider Inam
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802
| | - Connor J Moore
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802
| | - John Sheehe
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802
| | - Stephen J Elledge
- Howard Hughes Medical Institute, Brigham and Women's Hospital, Boston, MA 02115;
- Division of Genetics, Brigham and Women's Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
| | - Michael T Hemann
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142;
| | - Justin R Pritchard
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802;
- Huck Institute for the Life Sciences, The Pennsylvania State University, University Park, PA 16802
| |
Collapse
|
4
|
Mohammed HH, Abuo-Rahma GEDA, Abbas SH, Abdelhafez ESM. Current Trends and Future Directions of Fluoroquinolones. Curr Med Chem 2019; 26:3132-3149. [DOI: 10.2174/0929867325666180214122944] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 10/16/2017] [Accepted: 02/08/2018] [Indexed: 12/12/2022]
Abstract
Fluoroquinolones represent an interesting synthetic class of antimicrobial agents with broad spectrum and potent activity. Since the discovery of nalidixic acid, the prototype of quinolones, several structural modifications to the quinolone nucleus have been carried out for improvement of potency, spectrum of activity, and to understand their structure activity relationship (SAR). The C-7 substituent was reported to have a major impact on the activity. Accordingly, Substitution at C-7 or its N-4-piperazinyl moiety was found to affect potency, bioavailability, and physicochemical properties. Also, it can increase the affinity towards mammalian topoisomerases that may shift quinolones from antibacterial to anticancer candidates. Moreover, the presence of DNA topoisomerases in both eukaryotic and prokaryotic cells makes them excellent targets for chemotherapeutic intervention in antibacterial and anticancer therapies. Based on this concept, several fluoroquionolones derivatives have been synthesized and biologically evaluated as antibacterial, antituberculosis, antiproliferative, antiviral and antifungal agents. This review is an attempt to focus on the therapeutic prospects of fluoroquinolones with an updated account on their atypical applications such as antitubercular and anticancer activities.
Collapse
Affiliation(s)
- Hamada H.H. Mohammed
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | | | - Samar H. Abbas
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | | |
Collapse
|
5
|
Zhao XL, Yu CZ. Vosaroxin induces mitochondrial dysfunction and apoptosis in cervical cancer HeLa cells: Involvement of AMPK/Sirt3/HIF-1 pathway. Chem Biol Interact 2018; 290:57-63. [DOI: 10.1016/j.cbi.2018.05.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 05/14/2018] [Accepted: 05/21/2018] [Indexed: 12/16/2022]
|
6
|
Gravina GL, Mancini A, Mattei C, Vitale F, Marampon F, Colapietro A, Rossi G, Ventura L, Vetuschi A, Di Cesare E, Fox JA, Festuccia C. Enhancement of radiosensitivity by the novel anticancer quinolone derivative vosaroxin in preclinical glioblastoma models. Oncotarget 2018; 8:29865-29886. [PMID: 28415741 PMCID: PMC5444710 DOI: 10.18632/oncotarget.16168] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 03/03/2017] [Indexed: 12/24/2022] Open
Abstract
Purpose Glioblastoma multiforme (GBM) is the most aggressive brain tumor. The activity of vosaroxin, a first-in-class anticancer quinolone derivative that intercalates DNA and inhibits topoisomerase II, was investigated in GBM preclinical models as a single agent and combined with radiotherapy (RT). Results Vosaroxin showed antitumor activity in clonogenic survival assays, with IC50 of 10−100 nM, and demonstrated radiosensitization. Combined treatments exhibited significantly higher γH2Ax levels compared with controls. In xenograft models, vosaroxin reduced tumor growth and showed enhanced activity with RT; vosaroxin/RT combined was more effective than temozolomide/RT. Vosaroxin/RT triggered rapid and massive cell death with characteristics of necrosis. A minor proportion of treated cells underwent caspase-dependent apoptosis, in agreement with in vitro results. Vosaroxin/RT inhibited RT-induced autophagy, increasing necrosis. This was associated with increased recruitment of granulocytes, monocytes, and undifferentiated bone marrow–derived lymphoid cells. Pharmacokinetic analyses revealed adequate blood-brain penetration of vosaroxin. Vosaroxin/RT increased disease-free survival (DFS) and overall survival (OS) significantly compared with RT, vosaroxin alone, temozolomide, and temozolomide/RT in the U251-luciferase orthotopic model. Materials and Methods Cellular, molecular, and antiproliferative effects of vosaroxin alone or combined with RT were evaluated in 13 GBM cell lines. Tumor growth delay was determined in U87MG, U251, and T98G xenograft mouse models. (DFS) and (OS) were assessed in orthotopic intrabrain models using luciferase-transfected U251 cells by bioluminescence and magnetic resonance imaging. Conclusions Vosaroxin demonstrated significant activity in vitro and in vivo in GBM models, and showed additive/synergistic activity when combined with RT in O6-methylguanine methyltransferase-negative and -positive cell lines.
Collapse
Affiliation(s)
- Giovanni Luca Gravina
- Department of Biotechnological and Applied Clinical Sciences, Division of Radiotherapy, University of L'Aquila, L'Aquila, Italy.,Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy
| | - Andrea Mancini
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy
| | - Claudia Mattei
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Neurosciences, University of L'Aquila, L'Aquila, Italy
| | - Flora Vitale
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Neurosciences, University of L'Aquila, L'Aquila, Italy
| | - Francesco Marampon
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy
| | - Alessandro Colapietro
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy
| | - Giulia Rossi
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy
| | - Luca Ventura
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Neurosciences, University of L'Aquila, L'Aquila, Italy
| | - Antonella Vetuschi
- Department of Biotechnological and Applied Clinical Sciences, Chair of Human Anatomy, University of L'Aquila, L'Aquila, Italy
| | - Ernesto Di Cesare
- Department of Biotechnological and Applied Clinical Sciences, Division of Radiotherapy, University of L'Aquila, L'Aquila, Italy
| | - Judith A Fox
- Sunesis Pharmaceuticals Inc., South San Francisco, CA, USA
| | - Claudio Festuccia
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
7
|
Baize N, Monnet I, Greillier L, Quere G, Kerjouan M, Janicot H, Vergnenegre A, Auliac JB, Chouaid C. Second-line treatments of small-cell lung cancers. Expert Rev Anticancer Ther 2017; 17:1033-1043. [DOI: 10.1080/14737140.2017.1372198] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Nathalie Baize
- UTTIOM (Unité Transversale de Thérapeutiques Innovantes en Oncologie Médicale), CHU Angers, France
| | - Isabelle Monnet
- Department of Pulmonology, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| | - Laurent Greillier
- Service d’Oncologie Multidisciplinaire et Innovations Thérapeutiques, AP-HM, Aix-Marseille Université, Marseille, France
| | - Gilles Quere
- Respiratory Disease Department, Brest University Brest, Brest, France
| | - Mallorie Kerjouan
- Respiratory Disease Department, Pontchaillou University Hospital, Rennes, France
| | - Henri Janicot
- Service de pneumologie, CHU Clermont-Ferrand, Clermont Ferrand, France
| | - Alain Vergnenegre
- UOTC (Unité d’Oncologie Thoracique et Cutanée), CHU Limoges, Limoges, France
| | | | - Christos Chouaid
- Department of Pulmonology, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| |
Collapse
|
8
|
Kukowska M. Amino acid or peptide conjugates of acridine/acridone and quinoline/quinolone-containing drugs. A critical examination of their clinical effectiveness within a twenty-year timeframe in antitumor chemotherapy and treatment of infectious diseases. Eur J Pharm Sci 2017; 109:587-615. [PMID: 28842352 DOI: 10.1016/j.ejps.2017.08.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 08/16/2017] [Accepted: 08/19/2017] [Indexed: 01/10/2023]
Abstract
Acridines/acridones, quinolines/quinolones (chromophores) and their derivatives constitute extremely important family of compounds in current medicine. Great significance of the compounds is connected with antimicrobial and antitumor activities. Combining these features together in one drug seems to be long-term benefit, especially in oncology therapy. The attractiveness of the chromophore drugs is still enhanced by elimination their toxicity and improvement not only selectivity, specificity but also bioavailability. The best results are reached by conjugation to natural peptides. This paper highlights significant advance in the study of amino acid or peptide chromophore conjugates that provide highly encouraging data for novel drug development. The structures and clinical significance of amino acid or peptide chromophore conjugates are widely discussed.
Collapse
Affiliation(s)
- Monika Kukowska
- Chair & Department of Chemical Technology of Drugs, Faculty of Pharmacy with Subfaculty of Laboratory Medicine, Medical University of Gdansk, Al. Gen. J. Hallera 107, 80-416 Gdansk, Poland.
| |
Collapse
|
9
|
Benton CB, Ravandi F. Targeting acute myeloid leukemia with TP53-independent vosaroxin. Future Oncol 2016; 13:125-133. [PMID: 27615555 DOI: 10.2217/fon-2016-0300] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Vosaroxin is a quinolone compound that intercalates DNA and induces TP53-independent apoptosis, demonstrating activity against acute myeloid leukemia (AML) in Phase I-III trials. Here, we examine vosaroxin's mechanism of action and pharmacology, and we review its use in AML to date, focusing on details of individual clinical trials. Most recently, when combined with cytarabine in a randomized Phase III trial (VALOR), vosaroxin improved outcomes versus cytarabine alone for relapsed/refractory AML in patients older than 60 years and for patients in early relapse. We consider its continued role in the context of a multifaceted strategy against AML, including its current use in clinical trials. Prospective use will define its role in the evolving landscape of AML therapy.
Collapse
Affiliation(s)
- Christopher B Benton
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
10
|
Madaan A, Verma R, Kumar V, Singh AT, Jain SK, Jaggi M. 1,8-Naphthyridine Derivatives: A Review of Multiple Biological Activities. Arch Pharm (Weinheim) 2015; 348:837-60. [DOI: 10.1002/ardp.201500237] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 10/10/2015] [Accepted: 10/14/2015] [Indexed: 12/16/2022]
Affiliation(s)
- Alka Madaan
- Cell Biology Lab; Dabur Research Foundation; Sahibabad, Ghaziabad Uttar Pradesh India
| | - Ritu Verma
- Cell Biology Lab; Dabur Research Foundation; Sahibabad, Ghaziabad Uttar Pradesh India
| | - Vivek Kumar
- Chemical Research Lab; Dabur Research Foundation; Sahibabad, Ghaziabad Uttar Pradesh India
| | - Anu T. Singh
- Cell Biology Lab; Dabur Research Foundation; Sahibabad, Ghaziabad Uttar Pradesh India
| | - Swatantra K. Jain
- Department of Medical Biochemistry, HIMSR and Department of Biotechnology; Jamia Hamdard; New Delhi India
| | - Manu Jaggi
- Cell Biology Lab; Dabur Research Foundation; Sahibabad, Ghaziabad Uttar Pradesh India
| |
Collapse
|
11
|
Mjos KD, Cawthray JF, Jamieson G, Fox JA, Orvig C. Iron(III)-binding of the anticancer agents doxorubicin and vosaroxin. Dalton Trans 2015; 44:2348-58. [PMID: 25534904 DOI: 10.1039/c4dt02934h] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The Fe(iii)-binding constant of vosaroxin, an anticancer quinolone derivative, has been determined spectrophotometrically and compared with the analogous Fe(iii) complex formed with doxorubicin. The in vivo metabolic stability and iron coordination properties of the quinolones compared to the anthracylines may provide significant benefit to cardiovascular safety. The mechanism of action of both molecules target the topoisomerase II enzyme. Both doxorubicin (Hdox, log βFeL3 = 33.41, pM = 17.0) and vosaroxin (Hvox, log βFeL3 = 33.80(3), pM = 15.9) bind iron(iii) with comparable strength; at physiological pH however, [Fe(vox)3] is the predominant species in contrast to a mixture of species observed for the Fe:dox system. Iron(iii) nitrate and gallium(iii) nitrate at a 1 : 3 ratio with vosaroxin formed stable tris(vosaroxacino)-iron(iii) and tris(vosaroxino)gallium(iii) complexes that were isolated and characterized. Their redox behavior was studied by CV, and their stereochemistry was further explored in temperature dependent (1)H NMR studies. The molecular pharmacology of their interaction with iron(iii) may be one possible differentiation in the safety profile of quinolones compared to anthracyclines in relation to cardiotoxicity.
Collapse
Affiliation(s)
- Katja Dralle Mjos
- Medicinal Inorganic Chemistry Group, University of British Columbia, Department of Chemistry, 2036 Main Mall, Vancouver, British Columbia V6 T 1Z1, Canada
| | | | | | | | | |
Collapse
|
12
|
Lara PN, Moon J, Redman MW, Semrad TJ, Kelly K, Allen JW, Gitlitz BJ, Mack PC, Gandara DR. Relevance of platinum-sensitivity status in relapsed/refractory extensive-stage small-cell lung cancer in the modern era: a patient-level analysis of southwest oncology group trials. J Thorac Oncol 2015; 10:110-5. [PMID: 25490004 PMCID: PMC4320001 DOI: 10.1097/jto.0000000000000385] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Extensive-stage small-cell lung cancer (SCLC) patients who progress after platinum-based chemotherapy are traditionally categorized as platinum sensitive (progression ≥ 90 days from last platinum dose) or refractory (progression < 90 days), a practice arising from seminal observations of worse survival in refractory patients. Subsequent trials accounted for platinum sensitivity, resulting in higher sample sizes and increased resource use. METHODS To assess whether platinum-sensitivity status remains associated with outcomes, patient-level data from recent Southwest Oncology Group trials in second- and/or third-line extensive-stage SCLC were pooled. Hazard ratios (HRs) for progression-free survival (PFS) and overall survival (OS) accounting for platinum sensitivity were calculated using unadjusted and adjusted Cox Proportional Hazard models. Recursive partitioning was performed to define prognostic risk groups. RESULTS Of 329 patients, 151 were platinum sensitive and 178 refractory. HRs from unadjusted Cox PFS and OS models for refractory versus sensitive disease were 1.0 (95% confidence interval, 0.81-1.25; p = 0.98) and 1.24 (0.99-1.57; p = 0.06), respectively. Adjusted Cox models showed that only elevated serum lactate dehydrogenase (HR, 2.04; p < 0.001), males (HR, 1.36; p = 0.04), performance status of 1 (HR, 1.25; p = 0.02), and weight loss greater than or equal to 5% (1.53, p = 0.01) were independently associated with OS. Platinum-sensitivity status was not associated with PFS (HR, 1.11; p = 0.49) or OS (HR, 1.25; p = 0.14), except in a model that excluded 36 patients who received more than one prior chemotherapy regimen (HR, 1.34; p = 0.049). Prognostic groups with differential OS outcomes (high, intermediate, and poor risk) were identified. CONCLUSIONS Platinum-sensitivity status may no longer be strongly associated with PFS or OS in at least one multivariate model. Validation of prognostic risk groups identified here is warranted. These data have critical implications in the design of future SCLC trials.
Collapse
Affiliation(s)
- Primo N Lara
- *University of California Davis Comprehensive Cancer Center, Sacramento, California; †SWOG Statistical Center and the Fred Hutchinson Cancer Research Center, Seattle, Washington; ‡Humboldt Medical Specialists, Eureka, California; and §Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Williamson EA, Damiani L, Leitao A, Hu C, Hathaway H, Oprea T, Sklar L, Shaheen M, Bauman J, Wang W, Nickoloff JA, Lee SH, Hromas R. Targeting the transposase domain of the DNA repair component Metnase to enhance chemotherapy. Cancer Res 2012; 72:6200-8. [PMID: 23090115 DOI: 10.1158/0008-5472.can-12-0313] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Previous studies have shown that the DNA repair component Metnase (SETMAR) mediates resistance to DNA damaging cancer chemotherapy. Metnase has a nuclease domain that shares homology with the Transposase family. We therefore virtually screened the tertiary Metnase structure against the 550,000 compound ChemDiv library to identify small molecules that might dock in the active site of the transposase nuclease domain of Metnase. We identified eight compounds as possible Metnase inhibitors. Interestingly, among these candidate inhibitors were quinolone antibiotics and HIV integrase inhibitors, which share common structural features. Previous reports have described possible activity of quinolones as antineoplastic agents. Therefore, we chose the quinolone ciprofloxacin for further study, based on its wide clinical availability and low toxicity. We found that ciprofloxacin inhibits the ability of Metnase to cleave DNA and inhibits Metnase-dependent DNA repair. Ciprofloxacin on its own did not induce DNA damage, but it did reduce repair of chemotherapy-induced DNA damage. Ciprofloxacin increased the sensitivity of cancer cell lines and a xenograft tumor model to clinically relevant chemotherapy. These studies provide a mechanism for the previously postulated antineoplastic activity of quinolones, and suggest that ciprofloxacin might be a simple yet effective adjunct to cancer chemotherapy.
Collapse
Affiliation(s)
- Elizabeth A Williamson
- Department of Medicine, University of Florida and Shands Health Care System, Gainesville, Florida, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Abstract
INTRODUCTION The antineoplastic quinolone derivative vosaroxin (SNS-595, Sunesis, South San Francisco, CA, USA) was first described in 2002. It represents a novel class of anticancer drugs and is currently in a Phase III clinical trial for relapsed and refractory acute myeloid leukemia (AML). AML is the most common form of acute leukemia in adults and is increasing in incidence due to the aging of the American population. Despite advances in diagnosis, prognostic prediction, and treatment in younger age groups, there has been little improvement in survival among patients over 60 years of age, who make up the majority of those affected. AREAS COVERED The development of vosaroxin, its mechanism of action, pharmacology, and metabolism, and the preclinical and clinical data to date will be covered. EXPERT OPINION Despite its structural dissimilarity, vosaroxin has mechanisms of action similar to the anthracyclines and anthracenediones already in use for the treatment of AML. However, unlike these agents, vosaroxin is not a P-gp substrate, appears to be unaffected by overexpression of P-gp or TP53 mutations, and may be useful in the treatment of AML, especially in the elderly.
Collapse
Affiliation(s)
- Jonathan A Abbas
- Medical University of South Carolina, Hollings Cancer Center, 96 Jonathon Lucas St, MSC 635 CSB 903, Charleston, 29425, USA
| | | |
Collapse
|
15
|
Bailly C. Contemporary challenges in the design of topoisomerase II inhibitors for cancer chemotherapy. Chem Rev 2012; 112:3611-40. [PMID: 22397403 DOI: 10.1021/cr200325f] [Citation(s) in RCA: 213] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Christian Bailly
- Centre de Recherche et Développement, Institut de Recherche Pierre Fabre, Toulouse, France.
| |
Collapse
|
16
|
|