539
Views
31
CrossRef citations to date
0
Altmetric
Reviews

New nucleoside analogs for patients with hematological malignancies

Pages 343-359 | Published online: 14 Feb 2011

Bibliography

  • Robak T, Korycka A, Lech-Maranda E, Robak P. Current status of older and new purine nucleoside analogues in the treatment of lymphoproliferative diseases. Molecules 2009;14:1183-226
  • Galmarini CM, Popowycz F, Joseph B. Cytotoxic nucleoside analogues: different strategies to improve their clinical efficacy. Curr Med Chem 2008;15:1072-82
  • Faderl S, Gandhi V, Kantarjian HM. Potential role of novel nucleoside analogs in the treatment of acute myeloid leukemia. Curr Opin Hematol 2008;15:101-7
  • Robak T, Jamroziak K, Gora-Tybor J, Comparison of cladribine plus cyclophosphamide with fludarabine plus cyclophosphamide as first-line therapy for chronic lymphocytic leukemia: a phase III randomized study by the Polish Adult Leukemia Group (PALG-CLL3 Study). J Clin Oncol 2010;28:1863-9
  • Parker WB. Enzymology of purine and pyrimidine antimetabolites used in the treatment of cancer. Chem Rev 2009;109:2880-93
  • Korycka A, Lech-Maranda E, Robak T. Novel purine nucleoside analogues for hematological malignancies. Recent Pat Anticancer Drug Discov 2008;3:123-36
  • Lech-Maranda E, Korycka A, Robak T. Pharmacological and clinical studies on purine nucleoside analogs–new anticancer agents. Mini Rev Med Chem 2006;6:575-81
  • Robak T, Lech-Maranda E, Korycka A, Robak E. Purine nucleoside analogs as immunosuppressive and antineoplastic agents: mechanism of action and clinical activity. Curr Med Chem 2006;13:3165-89
  • Lech-Maranda E, Korycka A, Robak T. Clofarabine as a novel nucleoside analogue approved to treat patients with haematological malignancies: mechanism of action and clinical activity. Mini Rev Med Chem 2009;9:805-12
  • Bonate PL, Arthaud L, Cantrell WR Jr, Discovery and development of clofarabine: a nucleoside analogue for treating cancer. Nat Rev Drug Discov 2006;5:855-63
  • Jeha S, Gandhi V, Chan KW, Clofarabine, a novel nucleoside analog, is active in pediatric patients with advanced leukemia. Blood 2004;103:784-9
  • Jeha S, Gaynon PS, Razzouk BI, Phase II study of clofarabine in pediatric patients with refractory or relapsed acute lymphoblastic leukemia. J Clin Oncol 2006;24:917-23
  • Burnett AK, Baccarani M, Johnson P, Effectiveness of clofarabine in elderly AML patients with adverse cytogenetics unfit for intensive chemotherapy [abstract 1985]. Blood 2006;108(Suppl):562a
  • Faderl S, Verstovsek S, Cortes J, Clofarabine and cytarabine combination as induction therapy for acute myeloid leukemia (AML) in patients 50 years of age or older. Blood 2006;108:45-51
  • Faderl S, Ravandi F, Huang X, A randomized study of clofarabine versus clofarabine plus low-dose cytarabine as front-line therapy for patients aged 60 years and older with acute myeloid leukemia and high-risk myelodysplastic syndrome. Blood 2008;112:1638-45
  • Kantarjian HM, Jeha S, Gandhi V, Clofarabine: past, present, and future. Leuk Lymphoma 2007;48:1922-30
  • Faderl S, Garcia-Manero G, Ravandi F, Oral (po) and Intravenous (iv) Clofarabine for Patients (pts) with Myelodysplastic Syndrome (MDS). ASH Annual Meeting Abstracts. Blood 2008;112:222
  • Roecker AM, Allison JC, Kisor DF. Nelarabine: efficacy in the treatment of clinical malignancies. Future Oncol 2006;2:441-8
  • Rodriguez COJ, Mitchell BS, Ayres M, Arabinosylguanine is phosphorylated by both cytoplasmic deoxycytidine kinase and mitochondrial deoxyguanosine kinase. Cancer Res 2002;62:3100-5
  • Ghandi V, Plunkett W. Clofarabine and nelarabine: two new purine nucleoside analogs. Curr Opini Oncol 2006;18:584-90
  • Gandhi V, Tam C, O'Brien S, Phase I trial of nelarabine in indolent Leukemias. J Clin Oncol 2008;26:1098-105
  • Cohen MH, Johnson JR, Justice R, Pazdur R. FDA drug approval summary: nelarabine (Arranon) for the treatment of T-cell lymphoblastic leukemia/lymphoma. Oncologist 2008;13:709-14
  • Kurtzberg J, Ernst TJ, Keating MJ, Phase I study of 506U78 administered on a consecutive 5-day schedule in children and adults with refractory hematologic malignancies. J Clin Oncol 2005;23:3396-403
  • Berg SL, Blaney SM, Devidas M, Phase II study of nelarabine (compound 506U78) in children and young adults with refractory T-cell malignancies: a report from the children's oncology group. J Clin Oncol 2005;23:3376-403
  • DeAngelo DJ, Yu D, Johnson JL, Nelarabine induces complete remissions in adults with relapsed or refractory T-lineage acute lymphoblastic leukemia or lymphoblastic lymphoma: cancer and leukemia Group B study 19801. Blood 2007;109:5136-42
  • Czuczman MS, Porcu P, Johnson J, Results of a phase II study of 506U78 in cutaneous T-cell lymphoma and peripheral T-cell lymphoma: CALGB 59901. Leuk Lymphoma 2007;48:97-103
  • Goy A, Bleyer A, Hagemeister F, Phase II study of compound GW506U78 (araG) for patients with indolent B-cell or peripheral T-cell lymphoma previously treated with chemotherapy [abstract 2359]. Blood 2003;102:639a
  • Korycka A, Błonski JZ, Robak T. Forodesine (BCX-1777, Immucillin H)—a new purine nucleoside analogue: mechanism of action and potential clinical application. Mini Rev Med Chem 2007;7:976-83
  • Bantia S, Miller PJ, Parker CD, Purine nucleoside phosphorylase inhibitor BCX-1777 (Immucillin-H)–a novel potent and orally active immunosuppressive agent. Int Immunopharmacol 2001;1:1199-210
  • Bantia S, Ananth SL, Parker CD, Mechanism of inhibition of T-acute lymphoblastic leukemia cells by PNP inhibitor–BCX- 1777. Int Immunopharmacol 2003;3:879-87
  • Kicska GA, Tyler PC, Evans GB, Purine-less death in plasmodium falciparum induced by immucillin-H, a transition state analogue of purine nucleoside phosphorylase. J Biol Chem 2002;277:3226-31
  • Balakrishnan K, Nimmanapalli R, Ravandi F, Forodesine, an inhibitor of purine nucleoside phosphorylase, induces apoptosis in chronic lymphocytic leukemia cells. Blood 2006;108:2392-8
  • Alonso R, Lopez-Guerra M, Upshaw R, Forodesine has high antitumor activity in chronic lymphocytic leukemia and activates p53-independent mitochondrial apoptosis by induction of p73 and BIM. Blood 2009;114:1563-75
  • Al-Kali A, Gandhi V, Ayoubi M, Forodesine: review of preclinical and clinical data. Future Oncol 2010;6:1211-17
  • Balakrishnan K, Burger JA, Quiroga MP, Influence of bone marrow stromal microenvironment on forodesine-induced responses in CLL primary cells. Blood 2010;116:1083-91
  • Balakrishnan K, Verma D, O'Brien S, Phase 2 and pharmacodynamic study of oral forodesine in patients with advanced, fludarabine-treated chronic lymphocytic leukemia. Blood 2010;116:886-92
  • Duvic M, Forero-Torres A, Foss F, Response to oral forodesine in refractory cutaneous T-cell lymphoma: interim results of a phase I/II study [abstract 122]. Blood 2007;110:44a
  • Furman RR, Lia Gore L, Ravandi F, Hoelzer D. Forodesine IV (Bcx-1777) is clinically active in relapsed/refractory T-cell leukemia: results of a phase II study (Interim Report) [abstract 1851]. Blood 2006;108:524a
  • Bieghs L, Caers J, De Bruyne E, The effects of forodesine in murine and human multiple myeloma cells. Adv Hematol 2010;2010:131895
  • Kamath VP, Xue J, Juarez-Brambila JJ. Synthesis of analogs of forodesine HCl, a human purine nucleoside phosphorylase inhibitor-Part II. Bioorg Med Chem Lett 2009;19:2627-9
  • Gandhi V, Chen W, Ayres M, Plasma and cellular pharmacology of 8-chloro-adenosine in mice and rats. Cancer Chemother Pharmacol 2002;50:85-94
  • Stellrecht CM, Rodriguez CO Jr, Ayres M, Gandhi V. RNA-directed actions of 8-chloro-adenosine in multiple myeloma cells. Cancer Res 2003;63:7968-74
  • Chen LS, Sheppard TL. Chain termination and inhibition of saccharomyces cerevisiae poly(A) polymerase by C-8-modified ATP analogs. J Biol Chem 2004;279:40405-11
  • Balakrishnan K, Stellrecht CM, Genini D, Cell death of bioenergetically compromised and transcriptionally challenged CLL lymphocytes by chlorinated ATP. Blood 2005;105:4455-62
  • Stellrecht CM, Phillip CJ, Cervantes-Gomez F, Gandhi V. Multiple myeloma cell killing by depletion of the MET receptor tyrosine kinase. Cancer Res 2007;67:9913-20
  • Dennison JB, Balakrishnan K, Gandhi V. Preclinical activity of 8-chloroadenosine with mantle cell lymphoma: roles of energy depletion and inhibition of DNA and RNA synthesis. Br J Haematol 2009;147:297-307
  • Available from: WWW.ClinicalTrials.Gov. NCT00714103. A Phase I Study of 8-Chloro-Adenosine in Previously Treated Patients With Chronic Lymphocytic Leukemia Verified by M.D. Anderson Cancer Center, July 2010
  • Mowery YM, Weinberg JB, Kennedy MN, LMP-420: a novel purine nucleoside analog with potent cytotoxic effects for CLL cells and minimal toxicity for normal hematopoietic cells. Leukemia 2010;24:1580-7
  • Hale LP, Cianciolo G. Treatment of experimental colitis in mice with LMP-420, an inhibitor of TNF transcription. J Inflamm (Lond) 2008;5:4
  • Haraguchi S, Day NK, Kamchaisatian W, LMP-420, a small-molecule inhibitor of TNF-alpha, reduces replication of HIV-1 and Mycobacterium tuberculosis in human cells. AIDS Res Ther 2006;3:8
  • Saiki JH, McCredie KB, Vietti TJ, 5-azacytidine in acute leukemia. Cancer 1978;42:2111-14
  • Garcia-Manero G. Demethylating agents in myeloid malignancies. Curr Opin Oncol 2008;20:705-10
  • Kiziltepe T, Hideshima T, Catley L, 5-Azacytidine, a DNA methyltransferase inhibitor, induces ATR-mediated DNA double-strand break responses, apoptosis, and synergistic cytotoxicity with doxorubicin and bortezomib against multiple myeloma cells. Mol Cancer Ther 2007;6:1718-27
  • Schmelz K, Wagner M, Dorken B, Tamm I. 5-Aza-2′-deoxycytidine induces p21WAF expression by demethylation of p73 leading to p53-independent apoptosis in myeloid leukemia. Int J Cancer 2005;114:683-95
  • Hollenbach PW, Nguyen AN, Brady H, A comparison of azacitidine and decitabine activities in acute myeloid leukemia cell lines. LoS One 2010;5:e9001
  • Fenaux P, Mufti GJ, Hellstrom-Lindberg E, Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomised, open-label, phase III study. Lancet Oncol 2009;10:223-32
  • Sudan N, Rossetti JM, Shadduck RK, Treatment of acute myelogenous leukemia with outpatient azacitidine. Cancer 2006;107:1839-43
  • Thepot S, Itzykson R, Seegers V, Azacytidine (Aza) as first line therapy in AML: results of the French ATU program [abstract 843]. Blood 2009;114:347
  • Blum W, Klisovic RB, Hackanson B, Phase I study of decitabine alone or in combination with valproic acid in acute myeloid leukemia. J Clin Oncol 2007;25:3884-91
  • Garcia-Manero G, Kantarjian HM, Sanchez-Gonzalez B, Phase 1/2 study of the combination of 5-aza-2′-deoxycytidine with valproic acid in patients with leukemia. Blood 2006;108:3271-9
  • Available from: http://www.supergen.com/investors/pressrelease.html?pressReleaseId=896
  • Wang Y, Liu X, Matsuda A, Plunkett W. Repair of 2′-C-cyano-2′-deoxy-1-beta-D-arabino-pentofuranosylcytosine-induced DNA single-strand breaks by transcription-coupled nucleotide excision repair. Cancer Res 2008;68:3881-9
  • Hanaoka K, Suzuki M, Kobayashi T, Antitumor activity and novel DNA-self-strand-breaking mechanism of CNDAC (1-(2-C-cyano-2-deoxy-beta-D-arabino-pentofuranosyl) cytosine) and its N4-palmitoyl derivative (CS-682). Int J Cancer 1999;82:226-36
  • Azuma A, Huang P, Matsuda A, Plunkett W. 2′-C-cyano-2′-deoxy-1-beta-D-arabino-pentofuranosylcytosine:a novel anticancer nucleoside analog that causes both DNA strand breaks and G(2) arrest. Mol Pharmacol 2001;59:725-31
  • Liu X, Matsuda A, Plunkett W. Ataxia-telangiectasia and Rad3-related and DNA-dependent protein kinase cooperate in G2 checkpoint activation by the DNA strand-breaking nucleoside analogue 2′-C-cyano-2′-deoxy-1-beta-D-arabino-pentofuranosylcytosine. Mol Cancer Ther 2008;7:133-42
  • Liu X, Guo Y, Li Y, Molecular basis for G2 arrest induced by 2′-C-cyano-2′-deoxy-1-beta-D-arabino-pentofuranosylcytosine and consequences of checkpoint abrogation. Cancer Res 2005;65:6874-81
  • Serova M, Galmarini CM, Ghoul A, Antiproliferative effects of sapacitabine (CYC682), a novel 2′-deoxycytidine-derivative, in human cancer cells. Br J Cancer 2007;97:628-36
  • Green SR, Choudhary AK, Fleming IN. Combination of sapacitabine and HDAC inhibitors stimulates cell death in AML and other tumour types. Br J Cancer 2010;103:1391-9
  • Kantarjian H, Garcia-Manero G, O'Brien S, Phase I clinical and pharmacokinetic study of oral sapacitabine in patients with acute leukemia and myelodysplastic syndrome. J Clin Oncol 2010;28:285-91
  • Swords R, Giles F. Troxacitabine in acute leukemia. Hematology 2007;12:219-27
  • Gourdeau H, Clarke ML, Ouellet F, Mechanisms of uptake and resistance to troxacitabine, a novel deoxycytidine nucleoside analogue, in human leukemic and solid tumor cell lines. Cancer Res 2001;61:7217-24
  • Moore LE, Boudinot FD, Chu CK. Preclinical pharmacokinetics of beta-L- dioxolane-cytidine, a novel anticancer agent, in rats. Cancer Chemother Pharmacol 1997;39:532-6
  • Siu LL, Attardo G, Izbicka E, Activity of (-)-2′-deoxy-3′- oxacytidine (BCH-4556) against human tumor colony-forming units. Ann Oncol 1998;9:885-91
  • Giles FJ, Cortes JE, Baker SD, Troxacitabine, a novel dioxolane nucleoside analog, has activity in patients with advanced leukemia. J Clin Oncol 2001;19:762-71
  • Giles FJ, Faderl S, Thomas DA, Randomized phase I/II study of troxacitabine combined with cytarabine, idarubicin, or topotecan in patients with refractory myeloid leukemias. J Clin Oncol 2003;21:1050-6
  • Giles FJ, Kantarjian HM, Cortes JE, Adaptive randomized study of idarubicin and cytarabine versus troxacitabine and cytarabine versus troxacitabine and idarubicin in untreated patients 50 years or older with adverse karyotype acute myeloid leukemia. J Clin Oncol 2003;21:1722-7
  • Roboz GJ, Giles FJ, Ritchie EK, Phase I/II study of continuous-infusion troxacitabine in refractory acute myeloid leukemia. J Clin Oncol 2007;25:10-5
  • Parker WB, Shaddix SC, Rose LM, Waud WR. Metabolism of 4′-thio-beta-D-arabinofuranosylcytosine in CEM cells. Biochem Pharmacol 2000;60:1925-32
  • Tiwari KN, Shortnacy-Fowler AT, Cappellacci L, Synthesis of 4′-thio-beta-D-arabinofuranosylcytosine (4′-thio-ara-C) and comparison of its anticancer activity with that of ara-C. Nucleosides Nucleotides Nucleic Acids 2000;19:329-40
  • Blajeski AL, Richardson KA, Emerson DL, OSI-7836 (4′-thio-beta-D-arabinofuranosylcytosine: a novel anticancer nucleoside analog that inhibits DNA polymerases and induces G2 arrest. Proc Am Assoc Cancer Res 2002;43: abstract 4779
  • Richardson KA, Vega TP, Richardson FC, Polymerization of the triphosphates of AraC, 2_,2_-difluorodeoxycytidine (dFdC) and OSI-7836 (T-araC) by human DNA polymerase alpha and DNA primase. Biochem Pharmacol 2004;68:2337-46
  • Tomkinson B, Brown E, Henninger D, The antitumor activity of GS7836 (4′-thio-araC), a nucleoside analog, in mouse xenografts: comparison to standard cytotoxic agents and schedule dependence [abstract 5418]. Proc Am Assoc Cancer Res 2002;43:1094
  • Waud WR, Gilbert KS, Shepherd RV, Preclinical antitumor activity of 4V-thio-h-D-arabinofuranosylcytosine (4V-thio-ara-C). Cancer Chemother Pharmacol 2003;51:422-6
  • Parker WB, Shaddix SC, Gilbert KS, Enhancement of the in vivo antitumor activity of clofarabine by 1-beta-D-[4-thio-arabinofuranosyl]-cytosine. Cancer Chemother Pharmacol 2009;64:253-61
  • Goss G, Siu LL, Gauthier I, A phase I, first in man study of OSI-7836 in patients with advanced refractory solid tumors:IND.147, a study of the investigational new drug program of the national cancer institute of canada clinical trials group. Cancer Chemother Pharmacol 2006;58:703-10
  • Lee CP, de Jonge MJ, O'Donnell AE, A phase I study of a new nucleoside analogue, OSI-7836, using two administration schedules in patients with advanced solid malignancies. Clin Cancer Res 2006;12:2841-8
  • Zhou L, Cheng X, Connolly BA, Zebularine: a novel DNA methylation inhibitor that forms a covalent complex with DNA methyltransferases. J Mol Biol 2002;321:591-9
  • Cheng JC, Weisenberger DJ, Gonzales FA, Continuous zebularine treatment effectively sustains demethylation in human bladder cancer cells. Mol Cell Biol 2004;24:1270-8
  • Scott SA, Lakshimikuttysamma A, Sheridan DP, Zebularine inhibits human acute myeloid leukemia cell growth in vitro in association with p15INK4B demethylation and reexpression. Exp Hematol 2007;35:263-73
  • Flotho C, Claus R, Batz C, The DNA methyltransferas inhibitors azacitidine, decitabine and zebularine exert differential effects on cancer gene expression in acute myeloid leukemia cells. Leukemia 2009;23:1019-28
  • Cheng JC, Yoo CB, Weisenberger DJ, Preferential response of cancer cells to zebularine. Cancer Cell 2004;6:151-8
  • Herranz M, Caballero MJ,. Fraga MF, et al. The novel DNA methylation inhibitor zebularine is effective against the development of murine T-cell lymphoma. Blood 2006;107:1174-7
  • Balch C, Yan P, Craft T, Antimitogenic and chemosensitizing effects of the methylation inhibitor zebularine in ovarian cancer. Mol Cancer Ther 2005;4:1505-14
  • Cheng JC, Matsen CB, Gonzales FA, Inhibition of DNA methylation and reactivation of silenced genes by zebularine. J Natl Cancer Inst 2003;95:399-409
  • Rao SP, Rechsteiner MP, Berger C, Zebularine reactivates silenced E- adherin but unlike 5-Azacytidine does not induce switching from latent to lytic pstein-Barr virus nfection in Burkitt's lymphoma Akata cells. Mol Cancer 2007;6:3
  • Marquez VE, Barchi JJ Jr, Kelley JA, Zebularine: a unique molecule for an pigenetically based strategy in cancer chemotherapy. The magic of its hemistry and biology. Nucleosides Nucleotides Nucleic Acids 2005;24:305-18
  • Madhavi Billam M, Sobolewski MD, Davidson NE. Effects of a novel DNA methyltransferase inhibitor zebularine on human breast cancer cells. Breast Cancer Res Treat 2010;120:581-92
  • Robak T, Wierzbowska A. Current and emerging therapies for acute myeloid leukemia. Clin Ther 2009;31(Pt 2):2349-70
  • Secrist JA III. Nucleosides as anticancer agents: from concept to the clinic. Nucleic Acids Symp Ser(Oxf) 2005;49:15-6

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.