Skip to main content

New Reverse Transcriptase Inhibitors in Development

  • Chapter
Reverse Transcriptase Inhibitors in HIV/AIDS Therapy

Part of the book series: Infectious Disease ((ID))

  • 750 Accesses

Abstract

Since the identification of HIV as the etiological agent of AIDS, the HIV reverse transcriptase (RT) enzyme has been considered an ideal selective antiretroviral drug (ARV) target. Although several polymerases are found in human cells, none are comparable to HIV RT in their ability to catalyze the synthesis of new DNA from an RNA template. A selective RT inhibitor should, therefore, exert limited toxicity. The RT enzyme plays a crucial role in the early stages of the viral life cycle and can be competitively inhibited by chain-terminating nucleoside analogs (several of which were synthesized for basic research use well before the discovery of HIV). Therefore, it is not surprising that nucleoside RT inhibitors (NRTIs) were the first ARVs to be made available for the treatment of HIV infection. There now exist two additional classes of RT inhibitors, with independent modes of action; the non-nucleoside RT inhibitors (NNRTIs), first discovered in the early 1990s (1, and the nucleotide RT inhibitors. As described in detail in the earlier chapters of this volume Chapter 11 to Chapter 14 the NNRTIs act by noncompetitive binding to a hydrophobic pocket on the p66 subunit of the RT enzyme. The consequence of NNRTI binding to this unique site within the RT alters the ability of the enzyme to carry out its function. The current generation of NNRTIs selectively inhibits HIV-1 replication, reflecting their very high specificity for RT. They even have no significant activity against HIV-2, simian immunodeficiency virus, or other studied retroviruses. Unlike the NRTIs and the nucleotide RT inhibitors, NNRTIs do not require intracellular activation through phosphorylation.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 169.00
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 249.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 219.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Preview

Unable to display preview. Download preview PDF.

Unable to display preview. Download preview PDF.

References

  1. Pauwels R, Andries K, Desmyter J, et al. Potent and selective inhibition of HIV-1 replication in vitro by a novel series of TIBo derivatives. Nature 1990;343: 470–474.

    Article  PubMed  CAS  Google Scholar 

  2. Palella FJ Jr, Delaney KM, Moorman AC, et al. Declining morbidity and mortality among patients with advanced human immunodeficiency virus infection.N Engl J Med 1998;338:853–860.

    Article  PubMed  Google Scholar 

  3. Mallal SA, John M, Moore CB, James IR, McKinnon EJ. Contribution of nucleoside analogue reverse transcriptase inhibitors to subcutaneous fat wasting in patients with HIV infection. AIDS 2000;14:1309–1316.

    Article  PubMed  CAS  Google Scholar 

  4. Vigouroux C, Gharakhanian S, Salhi Y, et al. Adverse metabolic disorders during highly active antiretroviral treatments (HAART) of HIV disease. Diabetes Metab 1999;25:383–392.

    PubMed  CAS  Google Scholar 

  5. ter Hofstede HJ, de Marie S, Foudraine NA, Danner SA, Brinkman K. Clinical features and risk factors of lactic acidosis following long-term antiretroviral therapy:4 fatal cases. Int J STD AIDS 2000;11:611–616.

    Article  PubMed  Google Scholar 

  6. Nunez M, Lana R, Mendoza JL, Martin-Carbonero L, Soriano V. Risk factors for severe hepatic injury after introduction of highly active antiretroviral therapy.J Acquir Immune Defic Syndr 2001;27:426–431.

    PubMed  CAS  Google Scholar 

  7. Welch KJ, Morse A. Association between efavirenz and selected psychiatric and neurological conditions. J Infect Dis 2002;185:268–269.

    Article  PubMed  Google Scholar 

  8. Simon V, Vanderhoeven J, Hurley A, et al. Evolving patterns of HIV-1 resistance to antiretroviral agents in newly infected individuals. AIDS 2002;16: 1511–1519

    Article  PubMed  Google Scholar 

  9. Grant RM, Hecht FM, Warmerdam M, et al. Time trends in primary HIV-1 drug resistance among recently infected persons. JAMA 2002;288:1811–1883.

    Article  Google Scholar 

  10. Analysis of prevalence of HIV-1 drug resistance in primary infections in the United Kingdom. BMJ 2001;322:1087–1088.

    Google Scholar 

  11. Briones C, Perez-olmeda M, Rodriguez C, del Romero J, Hertogs K, Soriano V. Primary genotypic and phenotypic HIV-1 drug resistance in recent seroconverters in Madrid. J Acquir Immune Defic Syndr 2001;26:145–150.

    Article  PubMed  CAS  Google Scholar 

  12. Mayers DL, Japour AJ, Arduino JM, et al. Dideoxynucleoside resistance emerges with prolonged zidovudine monotherapy. The RV43 Study Group. Antimicrob Agents Chemother 1994;38:307–314.

    PubMed  CAS  Google Scholar 

  13. Shulman NS, Machekano RA, Shafer RW, et al. Genotypic correlates of a virologic response to stavudine after zidovudine monotherapy. J Acquir Immune Defic Syndr 2001;27:377–380.

    PubMed  CAS  Google Scholar 

  14. Iversen AK, Shafer RW, Wehrly K, et al. Multidrug-resistant human immunodeficiency virus type 1 strains resulting from combination antiretroviral therapy. J Virol 1996;70:1086–1090.

    PubMed  CAS  Google Scholar 

  15. Larder BA Bloor S Kemp SD et al. A family of insertion mutations between codons 67 and 70 of human immunodeficiency virus type 1 reverse transcriptase confer multinucleoside analog resistance. Antimicrob Agents Chemother 1999;43:1961–1967

    Google Scholar 

  16. Bangsberg DR, Perry S, Charlebois ED, et al. Non-adherence to highly active antiretroviral therapy predicts progression to AIDS. AIDS 2001;15:1181–1183.

    Article  PubMed  CAS  Google Scholar 

  17. Knobel H, Guelar A, Carmona A, et al. Virologic outcome and predictors of virologic failure of highly active antiretroviral therapy containing protease inhibitors. AIDS Patient Care STDS 2001;15:193–199.

    Article  PubMed  CAS  Google Scholar 

  18. Landovitz RJ, Sax PE. NRTI-associated mitochondrial toxicity. AIDS Clin Care 2001;13:43–45.

    PubMed  CAS  Google Scholar 

  19. Chen CH, Chen Y-C. The role of cytoplasmic deoxycytidine kinase in the mitochondrial effects of the anti-HIV compound, 2′3′-dideoxy-cytidine. J Biol Chem 1995;276:2859.

    Google Scholar 

  20. Kashman Y, Gustafson KR, Fuller RW, et al. The calanolides, a novel HIVinhibitory class of coumarin derivatives from the tropical rainforest tree, Calophyllum lanigerum. J Med Chem 1992;35:2735–2743.

    Article  PubMed  CAS  Google Scholar 

  21. Hoshi A, Castaner J. Alovudine. Drugs Future 1994;19:221–224.

    Google Scholar 

  22. Kim EY, Vrang L, oberg B, Merigan TC. Anti-HIV type 1 activity of 3′-fluoro-3′-deoxythymidine for several different multidrug-resistant mutants. AIDS Res Hum Retroviruses 2001;17:401–407.

    Article  PubMed  CAS  Google Scholar 

  23. Gu Z, Nguyen-Ba N, Ren J, et al. BCH-10618, a new heterosubstituted nucleoside analogue against HIV-1 infection. Antivir Ther 2001;6:11.

    Google Scholar 

  24. Fujiwara T, Sato A, el Farrash M, et al. S-1153 inhibits replication of known drug-resistant strains of human immunodeficiency virus type 1. Antimicrob Agents Chemother 1998;42:1340–1345.

    PubMed  CAS  Google Scholar 

  25. Ren J, Nichols C, Bird LE, et al. Binding of the second generation non-nucleoside inhibitor S-1153 to HIV-1 reverse transcriptase involves extensive main chain hydrogen bonding. J Biol Chem 2000;275:14-316–14-320.

    CAS  Google Scholar 

  26. Andries K, Azijn H, Thielemans T, et al. TMC125, a novel next generation nonnucleoside reverse transcriptase inhibitor active against non-nucleoside reverse transcriptase inhibitor-resistant HIV-1. Antimicrob Agents Chemother 2004;48:4680–4686.

    Article  PubMed  CAS  Google Scholar 

  27. DeBethune MP, Hertogs K, Azijn H, et al. R165335-TMC125, a third generation non nucleoside reverse transcriptase inhibitor (NNRTI), inhibits 98% of more than 2,000 recombinant HIV clinical isolates at 100 nM [abstract 1841]. 40th Interscience Conference on Antimicrobial Agents and Chemotherapy; Toronto, Canada; Sept 17–20, 2000.

    Google Scholar 

  28. Pelemans H, Esnouf R, De Clercq E, Balzarini J. Mutational analysis of trp-229 of human immunodeficiency virus type 1 reverse transcriptase (RT) identifies this amino acid residue as a prime target for the rational design of new non-nucleoside RT inhibitors. Mol Pharmacol 2000;57:954–960.

    PubMed  CAS  Google Scholar 

  29. Saboulard D, Naesens L, Cahard D, et al. Characterization of the activation pathway of phosphoramidate triester prodrugs of stavudine and zidovudine. Mol Pharmacol 1999;56:693–704.

    PubMed  CAS  Google Scholar 

  30. Gu Z, Wainberg MA, Nguyen-Ba P, L’Heureux L, de Muys JM, Rando RF. Anti-HIV-1 activities of 1,3-dioxolane guanine and 2,6-diaminopurine dioxolane. Nucleosides Nucleotides 1999;18:891–892.

    Article  PubMed  CAS  Google Scholar 

  31. Tanaka H, Baba M, Hayakawa H, et al. A new class of HIV-1-specific 6-substituted acyclouridine derivatives: synthesis and anti-HIV-1 activity of 5-or 6-substituted analogues of 1-[(2-hydroxyethoxy)methyl]-6-(phenylthio)thymine (HEPT). J Med Chem 1991;34:349–357.

    Article  PubMed  CAS  Google Scholar 

  32. Reliquest V, Peytavin G, Allavena C, et al. The pharmacokinetics and tolerance of once daily nevirapine and twice daily emirivine when used in combination. 40th Interscience Conference on Antimicrobial Agents and Chemotherapy; Toronto, Canada; 2000.

    Google Scholar 

  33. Hicks C, Harmon J, Anderson J, Makhuli K, Blum R, Miralles GD. Dual NNRTI therapy (MKC-442, emivirine (EMV) and efavirenz (EFV)) for patients failing PI regimens: pharmacokinetics and short-term efficacy. 7th Conference on Retroviruses and opportunistic Infections [abstract 670]; San Francisco, CA; 2000, Jan 30–Feb 2.

    Google Scholar 

  34. Kim Ho, Schinazi RF, Nampalli S, et al. 1,3-Dioxolanylpurine nucleosides (2R,4R) and (2R,4S) with selective anti-HIV-1 activity in human lymphocytes. J Med Chem 1993;36:30–37.

    Article  PubMed  CAS  Google Scholar 

  35. Furman PA, Jeffrey J, Kiefer LL, et al. Mechanism of action of 1-beta-D-2,6-diaminopurine dioxolane, a prodrug of the human immunodeficiency virus type 1 inhibitor 1-beta-D-dioxolane guanosine. Antimicrob Agents Chemother 2001;45: 158–165.

    Article  PubMed  CAS  Google Scholar 

  36. Triangle Pharmaceuticals licenses anti-AIDS compounds from Emory Univ. Emerging Pharmaceuticals 1996;5:6.

    Google Scholar 

  37. Bazmi HZ, Hammond JL, Cavalcanti SC, Chu CK, Schinazi RF, Mellors JW. In vitro selection of mutations in the human immunodeficiency virus type 1 reverse transcriptase that decrease susceptibility to (-)-beta-D-dioxolane-guanosine and suppress resistance to 3′-azido-3′-deoxythymidine. Antimicrob Agents Chemother 2000;44:1783–1788.

    Article  PubMed  CAS  Google Scholar 

  38. Gu Z, Wainberg MA, Nguyen-Ba N, et al. Mechanism of action and in vitro activity of 1′,3′-dioxolanylpurine nucleoside analogues against sensitive and drug-resistant human immunodeficiency virus type 1 variants. Antimicrob Agents Chemother 1999;43:2376–2382.

    PubMed  CAS  Google Scholar 

  39. Mewshaw JP, Myrick FT, Wakefield DA, et al. Dioxolane guanosine, the active form of the prodrug diaminopurine dioxolane, is a potent inhibitor of drug-resistant HIV-1 isolates from patients for whom standard nucleoside therapy fails. J Acquir Immune Defic Syndr 2002;29:11–20.

    PubMed  CAS  Google Scholar 

  40. Jeffrey K, Borroto-Esoda K, Feng J, et al. Amdoxovir, a nucleoside reverse transcriptase inhibitor, is active against HIV mutants resistant to standard nucleoside therapy. Antivir Ther 2001;6:14.

    Google Scholar 

  41. Harris J, Borroto-Esoda K, Myrick FT, Painter GR. The effect of mycophenolic acid and ribavirin on the anti-HIV activity of amdoxovir (DAPD) [abstract WePeB5900]. 14th International AIDS Conference;Barcelona, Spain; July 7–12, 2002.

    Google Scholar 

  42. Deeks S, Kessler H, Eron J, et al. Short-term monotherapy of DAPD in HIVinfected patients. Antivir Ther 2000;5:7–8.

    Google Scholar 

  43. Eron JJ, Kessler H, Thompson M, Deeks S, Arduino R, Rosseau F. Clinical HIV suppression after short-term monotherapy with DAPD [abstract 690]. 40th Interscience Conference on Antimicrobial Agents and Chemotherapy; Toronto, Canada; 2000.

    Google Scholar 

  44. Thompson M, Richmond G, Kessler H, et al. Preliminary results of amodoxovir in treatment-experienced patients [abstract 554]. 10th Conference on Retroviruses and opportunistic Infections; Boston, MA; February 10–14, 2003.

    Google Scholar 

  45. Gripshover B, Santa J, Ribaudo H, et al. A randomized, placebo-controlled trial of Amdoxovir vs placebo with enfuvirtide plus optimized background therapy for HIV-infected subjects failing current therapy (AACTG 5118) [abstract 553]. 12th Conference on Retroviruses and opportunistic Infections; Boston, MA; February 22–25, 2005.

    Google Scholar 

  46. Lin TS, Luo MZ, Liu MC, et al. Design and synthesis of 2′,3′-dideoxy-2′,3′-didehydro-B-L-cytidine (B-L-d4C) and 2′,3′-dideoxy 2′,3′-didehydro-B-L-5-fluorocytidine (B-L-Fd4C), two exceptionally potent inhibitors of human hepatitis B virus (HBV) and potent inhibitors of human immunodeficiency virus (HIV) in vitro. J Med Chem 1996;39:1757–1759.

    Article  PubMed  CAS  Google Scholar 

  47. Dutschman GE, Bridges EG, Liu SH, et al. Metabolism of 2′, 3′-dideoxy-2′, 3′-didehydro-β -L(—)-5-fluorocytidine and its activity in combination with clinically approved anti-human immunodeficiency virus β -D(+) nucleoside analogs in vitro. Antimicrob Agents Chemother 1998;42:1799-1804.

    PubMed  CAS  Google Scholar 

  48. Chen SH. Comparative evaluation of L-Fd4C and related nucleoside analogs as promising antiviral agents. Curr Med Chem 2002;9:899–912.

    Article  PubMed  CAS  Google Scholar 

  49. Le Guerhier F, Pichoud C, Jamard C, et al. Antiviral activity of B-L-2′,3′-dideoxy-2′,3′-didehydro-5-fluorocytidine in woodchucks chronically infected with woodchuck hepatitis virus. Antimicrob Agents Chemother 2001;45:1065–1077.

    Article  PubMed  Google Scholar 

  50. Le Guerhier F, Pichoud C, Guerret S, et al. Characterization of the antiviral effect of 2′,3′-dideoxy-2′, 3′-didehydro-B-L-5-fluorocytidine in the duck hepatitis B virus infection model. Antimicrob Agents Chemother 2000;44:111–122.

    Article  PubMed  Google Scholar 

  51. Zhu YL, Dutschman DE, Liu SH, Bridges EG, Cheng YC. Anti-hepatitis B virus activity and metabolism of 2′,3′-dideoxy-2′,3′-didehydro-B-L(-)-5-fluorocytidine. Antimicrob Agents Chemother 1998;42:1805–1810.

    PubMed  CAS  Google Scholar 

  52. Dunkle LM, oshana SC, Cheng YC, Hertogs K, Rice WG [abstract 303]. ACH-126, 443: a new nucleoside analogue with potent activity against wild type and resistant HIV-1 and a promising pharmacokinetic and mitochondrial safety profile. 8th Conference on Retroviruses and opportunistic Infections;Chicago, IL; February 4–8, 2001.

    Google Scholar 

  53. Murphy R. New agents in early clinical development [abstract MoorA141]. 14th International AIDS Conference; Barcelona, Spain; July 7–12, 2002.

    Google Scholar 

  54. Dunkle LM, Gathe JC, Pedevillano DE, Robison HG, Rice WG, Pottage JC, the ACH-006 study team. Elvucitabine: potent antiviral activity demonstrated in multi-drug resistant HIV infection. Antivir Ther 2003;8:S5.

    Google Scholar 

  55. Schinazi RF, Mellors J, Bazmi H, et al. DPC 817: a cytidine nucleoside analog with activity against zidovudine-and lamivudine-resistant viral variants.Antimicrob Agents Chemother 2002;46:1394–1401.

    Article  PubMed  CAS  Google Scholar 

  56. Hammond J, Chu CK, Schinazi RF, et al. Structural features of nucleoside analogue reverse transcriptase inhibitors important for selection of resistance mutations in HIV-1 reverse transcriptase. Antivir Ther 2001;6:36.

    Google Scholar 

  57. Ma L, Hurwitz SJ, Shi j, et al. Pharmacokinetics of the antiviral agent B-D-2′,3′-didehydro-2′,3′-dideoxy-5-fluorocytidine in rhesus monkeys. Antimicrob Agents Chemother 1999;43:381–384.

    PubMed  CAS  Google Scholar 

  58. Stuyver LJ, McBrayer TR, Schrman D, et al. Potent antiretroviral effect of Reverset( in HIV-1 infected adults following a single oral dose. Antivir Ther 2004; 9(4):529–536.

    PubMed  CAS  Google Scholar 

  59. Balzarini J, Baba M, Pauwels R, Herdewijn P, De Clercq E. Anti-retrovirus activity of 3′-fluoro-and 3′-azido-substituted pyrimidine 2′,3′-dideoxynucleoside analogues. Biochem Pharmacol 1988;37:2847–2856.

    Article  PubMed  CAS  Google Scholar 

  60. Kong XB, Zhu QY, Vidal PM, et al. Comparisons of anti-human immunodeficiency virus activities, cellular transport, and plasma and intracellular pharmacokinetics of 3′-fluoro-3′-deoxythymidine and 3′-azido-3′-deoxythymidine. Antimicrob Agents Chemother 1992;36:808–818.

    PubMed  CAS  Google Scholar 

  61. Lundgren B, Bottiger D, Ljungdahl-Stahle E, et al. Antiviral effects of 3′-fluorothymidine and 3′-azidothymidine in cynomolgus monkeys infected with simian immunodeficiency virus. J Acquir Immune Defic Syndr 1991;4:489–498.

    PubMed  CAS  Google Scholar 

  62. Bazin H, Chattopadhyaya J, Datema R, et al. An analysis of the inhibition of replication of HIV and MuLV by some 3′-blocked pyrimidine analogs. Biochem Pharmacol 1989;38:109–119.

    Article  PubMed  CAS  Google Scholar 

  63. Vrang L, Zhang H, Palmer S, Kim E-Y, Merigan TC, oberg B. In vitro effects of MIV-310 (alovudine, 3′-fluorodeoxythymidine, FLT) against HIV mutants. Antivir Ther 2002;7:S18–S19.

    Google Scholar 

  64. Calvez V, Tubiana R, Ghosn J, et al. MIV-310 reduces markedly viral load in patients with virological failure despite multiple-drug therapy: results from a 4-week Phase II study. Antivir Ther 2002;7:S4.

    Google Scholar 

  65. Katlama C, Ghosn J, Tubiana R et al. MIV-310 reduces HIV viral load in patients failing multiple antiretroviral therapy: results from a 4-week phase II study. AIDS 2004;18:1299–1304.

    Article  PubMed  CAS  Google Scholar 

  66. de Muys JM, Gourdeau H, Nguyen-Ba N, et al. Anti-human immunodeficiency virus type 1 activity, intracellular metabolism, and pharmacokinetic evaluation of 2′-deoxy-3′-oxa-4′-thiocytidine. Antimicrob Agents Chemother 1999;43:1835–1844.

    PubMed  Google Scholar 

  67. Bedard J, Wainberg TB, Mansour T. Characterization of the anti-HIV properties,resistance profile and safety of doTC (2′-deoxy-3′-oxa-4′ thiocytidine) [abstract 41196]. 12th International AIDS Conference; Geneva, Switzerland; June 28–July 3, 1998.

    Google Scholar 

  68. Smith PF, Forrest A, Ballow CH, Martin DE, Proulx L. Absolute bioavailability and disposition of (-) and (+) 2′-deoxy-3′-oxa-4′-thiocytidine (doTC) following single intravenous and oral doses of racemic doTC in humans. Antimicrob Agents Chemother 2000;44:1609–1615.

    Article  PubMed  CAS  Google Scholar 

  69. Smith PF, Forrest A, Ballow CH, Martin DE, Proulx L. Safety, tolerability, and pharmacokinetics of single oral doses of BCH-10652 in healthy adult males. Antimicrob Agents Chemother 2000;44:2816–2823.

    Article  PubMed  CAS  Google Scholar 

  70. Mansour T, Wainberg M, Salomon H, et al. Resistance profile, selectivity and cellular uptake studies of BCH-10618, BCH-10619 and BCH-10652. Abstract 78 Antivir Res 1998;37:A62.

    Google Scholar 

  71. Cahn P, Lange J, Cassetti I, et al. Anti-HIV-1 activity of SPD 754, a new NRTI:Results of a 10 day monotherapy study in treatment naive HIV patients [abstractLB15]. 2nd IAS Conference on HIV Pathogenesis and Treatment;Paris, France; July 13–16, 2003.

    Google Scholar 

  72. Bethell R, Adams J, De Muys J, et al. Pharmacological evaluation of a dual deoxycytidine analogue combination: 3TC and SPD574 [abstract 138]. 11th Conferenceon Retroviruses and opportunistic Infections;San Francisco, CA; Feb 8–11, 2004.

    Google Scholar 

  73. Potts KE, Fujiwara T, Sato A, et al. Resistance profile of AG1549, a novel nonnucleoside reverse transcriptase inhibitor. Antivir Ther 1999;4:10.

    Google Scholar 

  74. Dezube BJ, Jacobs MS. A second generation non-nucleoside reverse transcript aseinhibitor, AG-1549, in patients infected with HIV-1: a phase I study [abstract 200]. 7th European Conference on Clinical Aspects and Treatment of HIV Infection; Lisbon, Portugal; october 23–27, 1999.

    Google Scholar 

  75. Hernandez J, Amador L, Amantea M, Chao H, Hawley P, Paradiso L. Short-coursemonotherapy with AG1549, a novel nonnucleoside reverse transcriptase inhibitor (NNRTI), in antiretroviral naive patients [abstract 699]. 7th Conference on Retroviruses and opportunistic Infections; San Francisco, CA; Jan 30–Feb 2, 2000.

    Google Scholar 

  76. Jacobs M, Leoung G, Dezube B, et al. Pharmacokinetic (PK) interaction of AG1549, a novel nonnucleoside reverse transcriptase inhibitor (NNRTI), with protease inhibitors (PI) [abstract 83]. 7th Conference on Retroviruses and opportunistic Infections; San Francisco, CA; Jan 30–Feb 2, 2000.

    Google Scholar 

  77. Wolfe P, Hawley P, Boccia G, et al. Safety and efficacy of Capravine versus placebo in HIV-infected patients failing a non nucleoside-reverse-transcriptaseinhibitor-containing regimen: results of a phase II, double-blind placebo-controlled study [abstract 323]. 8th Conference on Retroviruses and opportunistic Infections; Chicago, IL; Feb 4–8, 2001.

    Google Scholar 

  78. Kulju K, Beall MA, Whysall C. 2001 Company Update: Pfizer—sustaining industry-adding earnings and growth performance. Credit Suisse First BostonCorporation; December 21, 2001.

    Google Scholar 

  79. Pesano R, Piraino S, Hawley P, et al, and the 1002 Study Group. 24-week safety, tolerability, and efficacy of capravirine as add-on therapy to nelfinavir and 2 nucleoside reverse transcriptase inhibitors in patients failing a non-nucleoside reverse transcriptase inhibitor-based regimen [abstract 555]. 12th Conference on Retroviruses and opportunistic Infections; Boston, MA; Feb 22–25, 2005.

    Google Scholar 

  80. Pauwels R. New non-nucleoside reverse transcriptase inhibitors (NNRTIs) in development for the treatment of HIV infections. Curr opin Pharmacol 2004;4:437–446.

    Article  PubMed  CAS  Google Scholar 

  81. Janssen PAJ, Lewi P, Arnold E, et al. In search of a novel anti-HIV drug: multidisciplinary coordination in the discovery of 4-[[4-[[4-[(1E)-2-cyanoethenyl]-2,6-dimethylphenyl]amino]-2-pyrimidinyl]amino]-benzonitrile (R278474, rilpivirine). J Med Chem 2005; 48:1901–1909.

    Article  PubMed  CAS  Google Scholar 

  82. Jonckheere H, Azjin H, Roobaert F, et al. A new and accelerated screening strategy to efficiently select novel drug candidates for HIV therapy [abstract S34]. 3rd European Symposium on the Clinical Implications of HIV Drug Resistance; Frankfurt, Germany; Feb 23–25, 2001.

    Google Scholar 

  83. de Kerpel J, Wigerinck P, Kukla MJ, et al. Structural characteristics of the binding of TMC125, a potent next-generation NNRTI, to wild type, single and double HIV mutants [abstract 560279]. 224th National American Chemical Society Meeting; Boston, MA; August 18–22, 2002.

    Google Scholar 

  84. Lewi PJ, de Jonge M, Daeyaert F, et al. on the detection of multiple-binding modes of ligands to proteins, from biological, structural, and modelling data [abstract 101]. 224th National American Chemical Society Meeting; Boston, MA; Aug 18–22, 2002.

    Google Scholar 

  85. Das K, Clark AD Jr, Lewi PJ, et al. Roles of conformational and positional adaptabilityin structure-based design of TMC125-R165335 (etravirine) and related non-nucleoside reverse transcriptase inhibitors that are highly potent and effective against wild-type and drug-resistant HIV-1 variants. J Med Chem 2004;47: 2250–2560.

    Article  CAS  Google Scholar 

  86. DeBethune M, Azjin H, Andries K, Janssen P, Pauwels R. In vitro selection experiments demonstrate reduces development of resistance with TMC 120 and TMC 125 compared with first generation non-nucleoside reverse transcriptase inhibitors. Antivir Ther 2002;6:6.

    Google Scholar 

  87. Vingerhoets J, Azjin H, Fransen E, Andries K, Pauwels R. TMC125 can suppress the selection of resistant HIV from a virus population carrying the K103N or the Y181C mutation. Antivir Ther 2002;7:S8.

    Google Scholar 

  88. Gruzdev B, Rakhmanova A, de Dier K, Comhaire S, Baede-vanDijk P, van’t Klooster G. TMC125 is a highly potent non-nucleoside reverse transcriptase inhibitor (NNRTI) in antiretroviral therapy (ART)-naive, HIV-1 infected subjects [abstract I-668].41st Interscience Conference on Antimicrobial Agents and Chemotherapy; Chicago, IL; Dec 16–19, 2001.

    Google Scholar 

  89. Sankatsing S, Weverling G, van’t Klooster G, Prins J, Lange J. TMC125 monotherapy for 1 week results in a similar initial rate of decline of HIV-1 RNA s therapy with a 5-drug regimen [abstract 6]. 9th Conference on Retroviruses and opportunistic Infections;Seattle, WA; Feb 24–28, 2002.

    Google Scholar 

  90. Gruzdev B, Horban A, Boron-Kaczmarska A, Gille D, van’t Klooster G, Pauwels R. TMC120, a new non-nucleoside reverse transcriptase inhibitor, is a potent antiretroviral in treatment naive, HIV-1 infected subjects [abstract 105]. 8th Conference on Retroviruses and opportunistic Infections; Chicago, IL; Feb 2–8, 2001.

    Google Scholar 

  91. Di Fabio S, van Roey J, Giannini G, et al. Inhibition of vaginal transmission of HIV-1 in hu-PBL-SCID mice by intravaginal gel containing an NNRTI-TMC120[abstract WeorD1315]. 14th International AIDS Conference; Barcelona, Spain; July 7–12, 2002.

    Google Scholar 

  92. de Béthune MP, Andries K, Azijn H, et al. TMC278, a new potent NNRTI; with an increased barrier to resistance and favourable pharmacokinetic profile [abstract 556]. 12th Conference on Retroviruses and opportunistic Infections; Boston, MA; Feb 22–25, 2005.

    Google Scholar 

  93. Goebel F, Yakovlev A, Pozniak A, et al. TMC278: potent anti-HIV activity in antiretroviral therapy-naive patients [abstract 160]. 12th Conference on Retroviruses and opportunistic Infections;Boston, MA; Feb 22–25, 2005.

    Google Scholar 

  94. Flavin MT, Rizzo JD, Khilevich A, et al. Synthesis, chromatographic resolution, and anti-human immunodeficiency virus activity of (+/-)-calanolide A and its enantiomers. J Med Chem 1996;39:1303–1313.

    Article  PubMed  CAS  Google Scholar 

  95. Galinis DL, Fuller RW, McKee TC, et al. Structure-activity modifications of the HIV-1 inhibitors (+)-calanolide A and (-)-calanolide B. J Med Chem 1996;39:4507–4510.

    Article  PubMed  CAS  Google Scholar 

  96. Zembower DE, Liao S, Flavin MT, et al. Structural analogues of the calanolide anti-HIV agents. Modification of the trans-10,11-dimethyldihydropyran-12-ol ring (ring C). J Med Chem 1997;40:1005–1017.

    Article  PubMed  CAS  Google Scholar 

  97. Hizi A, Tal R, Shaharabany M, et al. Specific inhibition of the reverse transcriptase of human immunodeficiency virus type 1 and the chimeric enzymes of human immunodeficiency virus type 1 and type 2 by nonnucleoside inhibitors. Antimicrob Agents Chemother 1993;37:1037–1042

    PubMed  CAS  Google Scholar 

  98. Currens MJ, Mariner JM, McMahon JB, Boyd MR. Kinetic analysis of inhibition of human immunodeficiency virus type-1 reverse transcriptase by calanolide A. J Pharmacol Exp Ther 1996;279:652–661.

    PubMed  CAS  Google Scholar 

  99. Quan Y, Motakis D, Buckheit R Jr, et al. Sensitivity and resistance to (+)-calanolide A of wild-type and mutated forms of HIV-1 reverse transcriptase. Antivir Ther 1999;4:203–209.

    PubMed  CAS  Google Scholar 

  100. Buckheit RW Jr, Fliakas-Boltz V, Yeagy-Bargo S, et al. Resistance to 1-[(2-hydroxyethoxy)methyl]-6-(phenylthio)thymine derivatives is generated by mutations at multiple sites in the HIV-1 reverse transcriptase. Virology 1995; 20:186–193.

    Article  Google Scholar 

  101. Currens MJ, Gulakowski RJ, Mariner JM, et al. Antiviral activity and mechanism of action of calanolide A against the human immunodeficiency virus type-1. J Pharmacol Exp Ther 1996;279:645–651.

    PubMed  CAS  Google Scholar 

  102. Creagh T, Ruckle JL, Tolbert DT, et al. Safety and pharmacokinetics of single doses of (+)-calanolide a, a novel, naturally occurring nonnucleoside reverse transcriptase inhibitor, in healthy, human immunodeficiency virus-negative human subjects. Antimicrob Agents Chemother 2001;45:1379–1386.

    Article  PubMed  CAS  Google Scholar 

  103. Sherer R, Dutta B, Anderson R et al. A phase 1B study of (+)-calanolide A in HIV-1 infected, antiretroviral-therapy-naive patients [abstract 508]. 7th Conference on Retroviruses and opportunistic Infections; San Francisco, CA; Jan 30-Feb 02, 2000.

    Google Scholar 

  104. Xu Z, Patil SD, Thilagar AK, Frank P. In vitro studies on interaction of (+)-calanolide A with cytochrome P-450 enzymes. 39th Interscience Conference on Antimicrobial Agents and Chemotherapy; San Francisco, CA; Sept 26–29, 1999.

    Google Scholar 

  105. Taylor DL, Ahmed PS, Tyms AS, et al. Drug resistance and drug combination features of the human immunodeficiency virus inhibitor, BCH-10652 [(+/-)-2′-deoxy-3′-oxa-4′-thiocytidine, doTC]. Antivir Chem Chemother 2000;11:291–301.

    PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2006 Humana Press Inc., Totowa, NJ

About this chapter

Cite this chapter

Pauwels, R. (2006). New Reverse Transcriptase Inhibitors in Development. In: St.Georgiev, V., Skowron, G., Ogden, R., Lange, J.M.A. (eds) Reverse Transcriptase Inhibitors in HIV/AIDS Therapy. Infectious Disease. Humana Press. https://doi.org/10.1007/978-1-59745-085-0_16

Download citation

  • DOI: https://doi.org/10.1007/978-1-59745-085-0_16

  • Publisher Name: Humana Press

  • Print ISBN: 978-1-58829-649-8

  • Online ISBN: 978-1-59745-085-0

  • eBook Packages: MedicineMedicine (R0)

Publish with us

Policies and ethics