Citation: Qinxue Hu, Robin Shattock. Novel Antiviral Agents Targeting HIV Entry and Transmission .VIROLOGICA SINICA, 2007, 22(6) : 451-461.

Novel Antiviral Agents Targeting HIV Entry and Transmission

  • Corresponding author: Qinxue Hu, qhu@sgul.ac.uk; qhu@wh.iov.cn
  • Received Date: 13 September 2007
    Accepted Date: 09 October 2007
    Available online: 01 December 2007

    Fund Project: NIH AI041346NIH AI065413the 973 Program 2006CB504200

  • Studies of the mechanism of HIV entry and transmission have identified multiple new targets for drug development. A range of inhibitors have demonstrated potent antiretroviral activity by interfering with CD4-gp120 interaction, coreceptor binding or viral-cell fusion in preclinical and clinical studies. One of these agents, fusion inhibitor enfuvirtide, is already in clinical use. Here we review the progress in the development of specific entry inhibitors as novel therapeutics. The potential of entry inhibitors as topical microbicides to block HIV transmission is also discussed.

  • 加载中
    1. Baba M, Nishimura O, Kanzaki N, et al.1999. A small-molecule, nonpeptide CCR5 antagonist with highly potent and selective anti-HIV-1 activity. Proc Natl Acad Sci USA, 96: 5698-5703.
        doi: 10.1073/pnas.96.10.5698

    2. Baba M, Takashima K, Miyake H, et al.2005. TAK-652 inhibits CCR5-mediated human immunodeficiency virus type 1 infection in vitro and has favorable pharmaco-kinetics in humans. Antimicrob Agents Chemother, 49: 4584-4591.
        doi: 10.1128/AAC.49.11.4584-4591.2005

    3. Berger E A, Murphy P M, Farber J M. 1999. Chemokine receptors as HIV-1 coreceptors: roles in viral entry, tropism, and disease. Annu Rev Immunol, 17: 657-700.
        doi: 10.1146/annurev.immunol.17.1.657

    4. Bleul C C, Farzan M, Choe H, et al.1996. The lymphocyte chemoattractant SDF-1 is a ligand for LESTR/ fusin and blocks HIV-1 entry. Nature, 382: 829-833.
        doi: 10.1038/382829a0

    5. Boshoff C, Endo Y, Collins P D, et al.1997. Angiogenic and HIV-inhibitory functions of KSHV-encoded che-mokines. Science, 278: 290-294.
        doi: 10.1126/science.278.5336.290

    6. Chan D C, Chutkowski C T, Kim P S. 1998. Evidence that a prominent cavity in the coiled coil of HIV type 1 gp41 is an attractive drug target. Proc Natl Acad Sci USA, 95: 15613-15617.
        doi: 10.1073/pnas.95.26.15613

    7. Chan D C, Fass D, Berger J M, et al.1997. Core structure of gp41 from the HIV envelope glycoprotein. Cell, 89: 263-273.
        doi: 10.1016/S0092-8674(00)80205-6

    8. Clapham P R, Weber J N, Whitby D, et al.1989. Soluble CD4 blocks the infectivity of diverse strains of HIV and SIV for T cells and monocytes but not for brain and muscle cells. Nature, 337: 368-370.
        doi: 10.1038/337368a0

    9. Cocchi F, DeVico A L, Garzino-Demo A, et al.1995. Identification of RANTES, MIP-1 alpha, and MIP-1 beta as the major HIV-suppressive factors produced by CD8+ T cells [see comments]. Science, 270: 1811-1815.
        doi: 10.1126/science.270.5243.1811

    10. Connor R I, Sheridan K E, Ceradini D, et al.1997.Change in coreceptor use correlates with disease pro-gression in HIV-1--infected individuals. J Exp Med, 185: 621-628.
        doi: 10.1084/jem.185.4.621

    11. Daar E S, Li X L, Moudgil T, et al.1990. High concentrations of recombinant soluble CD4 are required to neutralize primary human immunodeficiency virus type 1 isolates. Proc Natl Acad Sci USA, 87: 6574-6578.
        doi: 10.1073/pnas.87.17.6574

    12. Dalgleish A G, Beverley P C, Clapham P R, et al.1984.The CD4 (T4) antigen is an essential component of the receptor for the AIDS retrovirus. Nature, 312: 763-767.
        doi: 10.1038/312763a0

    13. Deng Y, Zheng Q, Ketas T J, et al.2007. Protein design of a bacterially expressed HIV-1 gp41 fusion inhibitor.Biochemistry, 46: 4360-4369.
        doi: 10.1021/bi7001289

    14. Donzella G A, Schols D, Lin S W, et al.1998. AMD3100, a small molecule inhibitor of HIV-1 entry via the CXCR4 co-receptor. Nat Med, 4: 72-77.
        doi: 10.1038/nm0198-072

    15. Doranz B J, Grovit-Ferbas K, Sharron M P, et al.1997.A small-molecule inhibitor directed against the chemokine receptor CXCR4 prevents its use as an HIV-1 coreceptor. J Exp Med, 186: 1395-1400.
        doi: 10.1084/jem.186.8.1395

    16. Dragic T, Trkola A, Thompson D A, et al.2000. A binding pocket for a small molecule inhibitor of HIV-1 entry within the transmembrane helices of CCR5. Proc Natl Acad Sci USA, 97: 5639-5644.
        doi: 10.1073/pnas.090576697

    17. Elias C J, Coggins C. 1996. Female-controlled methods to prevent sexual transmission of HIV. Aids, 10 Suppl 3: S43-51.

    18. Fichorova R N, Tucker L D, Anderson D J. 2001. The molecular basis of nonoxynol-9-induced vaginal inflame-mation and its possible relevance to human immunode-ficiency virus type 1 transmission. J Infect Dis, 184: 418-428.
        doi: 10.1086/jid.2001.184.issue-4

    19. Gallo S A, Finnegan C M, Viard M, et al.2003. The HIV Env-mediated fusion reaction. Biochim Biophys Acta, 1614: 36-50.
        doi: 10.1016/S0005-2736(03)00161-5

    20. Geijtenbeek T B, Kwon D S, Torensma R, et al.2000.DC-SIGN, a dendritic cell-specific HIV-1-binding protein that enhances trans-infection of T cells. Cell, 100:587-597.
        doi: 10.1016/S0092-8674(00)80694-7

    21. Hatse S, Princen K, De Clercq E, et al.2005. AMD3465, a monomacrocyclic CXCR4 antagonist and potent HIV entry inhibitor. Biochem Pharmacol, 70: 752-761.
        doi: 10.1016/j.bcp.2005.05.035

    22. Ho H T, Fan L, Nowicka-Sans B, et al.2006. Envelope conformational changes induced by human immunode-ficiency virus type 1 attachment inhibitors prevent CD4 binding and downstream entry events. J Virol, 80: 4017-4025.
        doi: 10.1128/JVI.80.8.4017-4025.2006

    23. Hu Q, Barry A P, Wang Z, et al.2000. Evolution of the human immunodeficiency virus type 1 envelope during infection reveals molecular corollaries of specificity for coreceptor utilization and AIDS pathogenesis. J Virol, 74: 11858-11872.
        doi: 10.1128/JVI.74.24.11858-11872.2000

    24. Hu Q, Frank I, Williams V, et al.2004. Blockade of attachment and fusion receptors inhibits HIV-1 infection of human cervical tissue. J Exp Med, 199: 1065-1075.
        doi: 10.1084/jem.20022212

    25. Hu Q, Mahmood N, Shattock R J. 2007. High-mannose-specific deglycosylation of HIV-1 gp120 induced by resistance to cyanovirin-N and the impact on antibody neutralization. Virology, 368: 145-154.
        doi: 10.1016/j.virol.2007.06.029

    26. Hu Q, Napier K B, Trent J O, et al.2005. Restricted variable residues in the C-terminal segment of HIV-1 V3 loop regulate the molecular anatomy of CCR5 utilization. J Mol Biol, 350: 699-712.
        doi: 10.1016/j.jmb.2005.05.024

    27. Hu Q, Trent J O, Tomaras G D, et al.2000. Identification of Env determinants in V3 that influence the molecular anatomy of CCR5 utilization. J Mol Biol, 302: 359-375.
        doi: 10.1006/jmbi.2000.4076

    28. Hu Q, Younson J, Griffin G E, et al.2006. Pertussis toxin and its binding unit inhibit HIV-1 infection of human cervical tissue and macrophages involving a CD14 pathway. J Infect Dis, 194: 1547-1556.
        doi: 10.1086/jid.2006.194.issue-11

    29. Hussey R E, Richardson N E, Kowalski M, et al.1988. A soluble CD4 protein selectively inhibits HIV replication and syncytium formation. Nature, 331:78-81.
        doi: 10.1038/331078a0

    30. Ichiyama K, Yokoyama-Kumakura S, Tanaka Y, et al. 2003. A duodenally absorbable CXC chemokine receptor 4 antagonist, KRH-1636, exhibits a potent and selective anti-HIV-1 activity. Proc Natl Acad Sci USA, 100: 4185-4190.
        doi: 10.1073/pnas.0630420100

    31. Jacobson J M, Lowy I, Fletcher C V, et al.2000.Single-dose safety, pharmacology, and antiviral activity of the human immunodeficiency virus (HIV) type 1 entry inhibitor PRO 542 in HIV-infected adults.J Infect Dis, 182: 326-329.
        doi: 10.1086/jid.2000.182.issue-1

    32. Jiang S, Zhao Q, Debnath A K. 2002. Peptide and non-peptide HIV fusion inhibitors. Curr Pharm Des, 8: 563-580.
        doi: 10.2174/1381612024607180

    33. Kadow J, Wang H G, Lin P F. 2006. Small-molecule HIV-1 gp120 inhibitors to prevent HIV-1 entry: an emerging opportunity for drug development. Curr Opin Investig Drugs, 7: 721-726.

    34. Kawamura T, Cohen S S, Borris D L, et al.2000.Candidate microbicides block HIV-1 infection of human immature Langerhans cells within epithelial tissue explants. J Exp Med, 192: 1491-1500.
        doi: 10.1084/jem.192.10.1491

    35. Kawamura T, Gulden F O, Sugaya M, et al.2003. R5 HIV productively infects Langerhans cells, and infection levels are regulated by compound CCR5 polymorphisms.Proc Natl Acad Sci USA, 100: 8401-8406.
        doi: 10.1073/pnas.1432450100

    36. Kilby J M, Hopkins S, Venetta T M, et al.1998. Potent suppression of HIV-1 replication in humans by T-20, a peptide inhibitor of gp41-mediated virus entry. Nat Med, 4: 1302-1307.
        doi: 10.1038/3293

    37. Klatzmann D, Champagne E, Chamaret S, et al.1984. T-lymphocyte T4 molecule behaves as the receptor for human retrovirus LAV. Nature, 312: 767-768.
        doi: 10.1038/312767a0

    38. Kledal T N, Rosenkilde M M, Coulin F, et al.1997. A broad-spectrum chemokine antagonist encoded by Kaposi's sarcoma-associated herpesvirus. Science, 277: 1656-1659.
        doi: 10.1126/science.277.5332.1656

    39. Lalezari J P, Henry K, O'Hearn M, et al.2003.Enfuvirtide, an HIV-1 fusion inhibitor, for drug-resistant HIV infection in North and South America.N Engl J Med, 348: 2175-2185.
        doi: 10.1056/NEJMoa035026

    40. Lazzarin A, Clotet B, Cooper D, et al.2003. Efficacy of enfuvirtide in patients infected with drug-resistant HIV-1 in Europe and Australia. N Engl J Med, 348:2186-2195.
        doi: 10.1056/NEJMoa035211

    41. Lederman M M, Veazey R S, Offord R, et al.2004.Prevention of vaginal SHIV transmission in rhesus macaques through inhibition of CCR5. Science, 306: 485-487.
        doi: 10.1126/science.1099288

    42. Lehner T. 2002. The role of CCR5 chemokine ligands and antibodies to CCR5 coreceptors in preventing HIV infection. Trends Immunol, 23: 347-351.
        doi: 10.1016/S1471-4906(02)02252-4

    43. Lehner T, Doyle C, Wang Y, et al.2001. Immunogenicity of the extracellular domains of C-C chemokine receptor 5 and the in vitro effects on simian immunodeficiency virus or HIV infectivity. J Immunol, 166: 7446-7455.
        doi: 10.4049/jimmunol.166.12.7446

    44. Lin P F, Blair W, Wang T, et al.2003. A small molecule HIV-1 inhibitor that targets the HIV-1 envelope and inhibits CD4 receptor binding. Proc Natl Acad Sci USA, 100: 11013-11018.
        doi: 10.1073/pnas.1832214100

    45. Liu S, Jing W, Cheung B, et al.2007. HIV gp41 C-terminal heptad repeat contains multifunctional domains. Relation to mechanisms of action of anti-HIV peptides. J Biol Chem, 282: 9612-9620.
        doi: 10.1074/jbc.M609148200

    46. Liu S, Lu H, Niu J, et al. 2005. Different from the HIV fusion inhibitor C34, the anti-HIV drug Fuzeon (T-20) inhibits HIV-1 entry by targeting multiple sites in gp41 and gp120. J Biol Chem, 280: 11259-11273.
        doi: 10.1074/jbc.M411141200

    47. Ma D, Yu S, Li B, et al.2007. Synthesis and Biological Evaluation of 1, 3, 3, 4-Tetrasubstituted Pyrrolidine CCR5 Receptor Antagonists. Discovery of a Potent and Orally Bioavailable Anti-HIV Agent. ChemMedChem, 2: 187-193.
        doi: 10.1002/(ISSN)1860-7187

    48. Mack M, Luckow B, Nelson P J, et al.1998.Aminooxypentane-RANTES induces CCR5 internalization but inhibits recycling: a novel inhibitory mechanism of HIV infectivity. J Exp Med, 187: 1215-1224.
        doi: 10.1084/jem.187.8.1215

    49. McKnight A, Weiss R A. 2003. Blocking the docking of HIV-1. Proc Natl Acad Sci USA, 100:10581-10582.
        doi: 10.1073/pnas.2035071100

    50. McMichael A J, Hanke T. 2003. HIV vaccines 1983-2003. Nat Med, 9: 874-880.
        doi: 10.1038/nm0703-874

    51. Moench T R, Chipato T, Padian N S. 2001. Preventing disease by protecting the cervix: the unexplored promise of internal vaginal barrier devices. Aids, 15: 1595-1602.
        doi: 10.1097/00002030-200109070-00001

    52. Moore J P, Doms R W. 2003. The entry of entry inhibitors: a fusion of science and medicine. Proc Natl Acad Sci USA, 100: 10598-10602.
        doi: 10.1073/pnas.1932511100

    53. Moore J P, Stevenson M. 2000. New targets for inhibitors of HIV-1 replication. Nature Reviews, Molecular Cell Biology, 1: 40-49.
        doi: 10.1038/35036060

    54. Mosier D E, Picchio G R, Gulizia R J, et al.1999. Highly potent RANTES analogues either prevent CCR5-using human immunodeficiency virus type 1 infection in vivo or rapidly select for CXCR4-using variants. J Virol, 73: 3544-3550.

    55. Munch J, Standker L, Adermann K, et al.2007.Discovery and optimization of a natural HIV-1 entry inhibitor targeting the gp41 fusion peptide. Cell, 129: 263-275.
        doi: 10.1016/j.cell.2007.02.042

    56. Murakami T, Nakajima T, Koyanagi Y, et al.1997. A small molecule CXCR4 inhibitor that blocks T cell line-tropic HIV-1 infection. J Exp Med, 186: 1389-1393.
        doi: 10.1084/jem.186.8.1389

    57. O'Hara B M, Olson W C. 2002. HIV entry inhibitors in clinical development. Curr Opin Pharmacol, 2:523-528.
        doi: 10.1016/S1471-4892(02)00196-0

    58. Oberlin E, Amara A, Bachelerie F, et al.1996. The CXC chemokine SDF-1 is the ligand for LESTR/fusin and prevents infection by T-cell-line-adapted HIV-1. Nature, 382: 833-835.
        doi: 10.1038/382833a0

    59. Olson W C, Rabut G E, Nagashima K A, et al.1999.Differential inhibition of human immunodeficiency virus type 1 fusion, gp120 binding, and CC-chemokine activity by monoclonal antibodies to CCR5. J Virol, 73:4145-4155.

    60. Pantophlet R, Burton D R. 2006. GP120: target for neutralizing HIV-1 antibodies. Annu Rev Immunol, 24: 739-769.
        doi: 10.1146/annurev.immunol.24.021605.090557

    61. Pastore C, Picchio G R, Galimi F, et al.2003. Two mechanisms for human immunodeficiency virus type 1 inhibition by N-terminal modifications of RANTES. Antimicrob Agents Chemother, 47: 509-517.
        doi: 10.1128/AAC.47.2.509-517.2003

    62. Reimann K A, Khunkhun R, Lin W, et al.2002. A humanized, nondepleting anti-CD4 antibody that blocks virus entry inhibits virus replication in rhesus monkeys chronically infected with simian immunodeficiency virus.AIDS Res Hum Retroviruses, 18: 747-755.
        doi: 10.1089/08892220260139486

    63. Rodriguez-Frade J M, Del Real G, Serrano A, et al.2004.Blocking HIV-1 infection via CCR5 and CXCR4 receptors by acting in trans on the CCR2 chemokine receptor. Embo J, 23: 66-76.
        doi: 10.1038/sj.emboj.7600020

    64. Root M J, Kay M S, Kim P S. 2001. Protein design of an HIV-1 entry inhibitor. Science, 291: 884-888.
        doi: 10.1126/science.1057453

    65. Rowland-Jones S L. 2003. Timeline: AIDS pathogenesis: what have two decades of HIV research taught us? Nat Rev Immunol, 3: 343-348.
        doi: 10.1038/nri1058

    66. Schols D, Struyf S, Van Damme J, et al. 1997. Inhibition of T-tropic HIV strains by selective antagonization of the chemokine receptor CXCR4. J Exp Med, 186: 1383-1388.
        doi: 10.1084/jem.186.8.1383

    67. Schon A, Madani N, Klein J C, et al. 2006.Thermodynamics of binding of a low-molecular-weight CD4 mimetic to HIV-1 gp120. Biochemistry, 45: 10973-10980.
        doi: 10.1021/bi061193r

    68. Shattock R J, Moore J P. 2003. Inhibiting sexual transmission of HIV-1 infection. Nat Rev Micro, 1: 25-34.
        doi: 10.1038/nrmicro729

    69. Shearer W T, Israel R J, Starr S, et al.2000.Recombinant CD4-IgG2 in human immunodeficiency virus type 1-infected children: phase 1/2 study. The Pediatric AIDS Clinical Trials Group Protocol 351 Study Team. J Infect Dis, 182: 1774-1779.
        doi: 10.1086/jid.2000.182.issue-6

    70. Sia S K, Kim P S. 2003. Protein grafting of an HIV-1-inhibiting epitope. Proc Natl Acad Sci USA, 100: 9756-9761.
        doi: 10.1073/pnas.1733910100

    71. Simmons G, Clapham P R, Picard L, et al.1997. Potent inhibition of HIV-1 infectivity in macrophages and lymphocytes by a novel CCR5 antagonist. Science, 276: 276-279.
        doi: 10.1126/science.276.5310.276

    72. Stone A. 2002. Microbicides: a new approach to preventing HIV and other sexually transmitted infections. Nat Rev Drug Discov, 1: 977-985.
        doi: 10.1038/nrd959

    73. Stone N D, Dunaway S B, Flexner C W, et al.2007.Multiple Dose Escalation Study of the Safety, Pharma-cokinetics, and Biologic Activity of Oral AMD070, a selective CXCR4 Receptor Inhibitor, in Human Subjects (ACTG A5191). Antimicrob Agents Chemother, 51:2351-2358.
        doi: 10.1128/AAC.00013-07

    74. Strizki J M, Tremblay C, Xu S, et al.2005. Discovery and characterization of vicriviroc (SCH 417690), a CCR5 antagonist with potent activity against human immunodefi-ciency virus type 1. Antimicrob Agents Chemother, 49: 4911-4919.
        doi: 10.1128/AAC.49.12.4911-4919.2005

    75. Strizki J M, Xu S, Wagner N E, et al.2001. SCH-C (SCH 351125), an orally bioavailable, small molecule antagonist of the chemokine receptor CCR5, is a potent inhibitor of HIV-1 infection in vitro and in vivo. Proc Natl Acad Sci USA, 98: 12718-12723.
        doi: 10.1073/pnas.221375398

    76. Sutor G C, Dreikhausen U, Vahning U, et al.1992.Neutralization of HIV-1 by anti-idiotypes to monoclonal anti-CD4. Potential for idiotype immunization against HIV. J Immunol, 149: 1452-1461.

    77. Takashima K, Miyake H, Kanzaki N, et al. 2005. Highly potent inhibition of human immunodeficiency virus type 1 replication by TAK-220, an orally bioavailable small-molecule CCR5 antagonist. Antimicrob Agents Che-mother, 49: 3474-3482.
        doi: 10.1128/AAC.49.8.3474-3482.2005

    78. Tamamura H, Xu Y, Hattori T, et al.1998. A low-molecular-weight inhibitor against the chemokine receptor CXCR4: a strong anti-HIV peptide T140. Biochem Biophys Res Commun, 253: 877-882.
        doi: 10.1006/bbrc.1998.9871

    79. Trkola A, Ketas T J, Nagashima K A, et al. 2001. Potent, broad-spectrum inhibition of human immunodeficiency virus type 1 by the CCR5 monoclonal antibody PRO 140. J Virol, 75: 579-588.
        doi: 10.1128/JVI.75.2.579-588.2001

    80. Trkola A, Kuhmann S E, Strizki J M, et al. 2002. HIV-1 escape from a small molecule, CCR5-specific entry inhibitor does not involve CXCR4 use. Proc Natl Acad Sci USA, 99: 395-400.
        doi: 10.1073/pnas.012519099

    81. Tsamis F, Gavrilov S, Kajumo F, et al.2003. Analysis of the mechanism by which the small-molecule CCR5 antagonists SCH-351125 and SCH-350581 inhibit human immunodeficiency virus type 1 entry. J Virol, 77: 5201-5208.
        doi: 10.1128/JVI.77.9.5201-5208.2003

    82. Turpin J A. 2002. Considerations and development of topical microbicides to inhibit the sexual transmission of HIV. Expert Opin Investig Drugs, 11: 1077-1097.
        doi: 10.1517/13543784.11.8.1077

    83. UNAIDS. 2006. Joint United Nations Programme on HIV/AIDS (UNAIDS and World Health Organization (WHO). AIDS Epidemic Updated-December 2006 (www.unaids.org/en/HIV_data/epi2006/default.asp).

    84. Veazey R S, Klasse P J, Ketas T J, et al. 2003. Use of a Small Molecule CCR5 Inhibitor in Macaques to Treat Simian Immunodeficiency Virus Infection or Prevent Simian-Human Immunodeficiency Virus Infection.J Exp Med, 198: 1551-1562.
        doi: 10.1084/jem.20031266

    85. Veazey R S, Klasse P J, Schader S M, et al. 2005.Protection of macaques from vaginal SHIV challenge by vaginally delivered inhibitors of virus-cell fusion. Nature, 438: 99-102.
        doi: 10.1038/nature04055

    86. Vermeire K, Princen K, Hatse S, et al. 2004. CADA, a novel CD4-targeted HIV inhibitor, is synergistic with various anti-HIV drugs in vitro. Aids, 18: 2115-2125.
        doi: 10.1097/00002030-200411050-00003

    87. Vermeire K, Zhang Y, Princen K, et al. 2002. CADA inhibits human immunodeficiency virus and human herpesvirus 7 replication by down-modulation of the cellular CD4 receptor. Virology, 302: 342-353.
        doi: 10.1006/viro.2002.1624

    88. Walker D K, Abel S, Comby P, et al. 2005. Species differences in the disposition of the CCR5 antagonist, UK-427, 857, a new potential treatment for HIV. Drug Metab Dispos, 33: 587-595.
        doi: 10.1124/dmd.104.002626

    89. Watson C, Jenkinson S, Kazmierski W, et al. 2005. The CCR5 receptor-based mechanism of action of 873140, a potent allosteric noncompetitive HIV entry inhibitor. Mol Pharmacol, 67: 1268-1282.
        doi: 10.1124/mol.104.008565

    90. Weissenhorn W, Dessen A, Harrison S C, et al.1997.Atomic structure of the ectodomain from HIV-1 gp41 [see comments]. Nature, 387: 426-430.
        doi: 10.1038/387426a0

    91. Wilkinson D, Tholandi M, Ramjee G, et al.2002.Nonoxynol-9 spermicide for prevention of vaginally acquired HIV and other sexually transmitted infections: systematic review and meta-analysis of randomised controlled trials including more than 5000 women. Lancet Infect Dis, 2: 613-617.
        doi: 10.1016/S1473-3099(02)00396-1

    92. Yang Q E, Stephen A G, Adelsberger J W, et al.2005.Discovery of small-molecule human immunodeficiency virus type 1 entry inhibitors that target the gp120-binding domain of CD4. J Virol, 79: 6122-6133.
        doi: 10.1128/JVI.79.10.6122-6133.2005

    93. Zaitseva M, Peden K, Golding H. 2003. HIV coreceptors: role of structure, posttranslational modifications, and internalization in viral-cell fusion and as targets for entry inhibitors. Biochim Biophys Acta, 1614: 51-61.
        doi: 10.1016/S0005-2736(03)00162-7

    94. Zhao Q, Ma L, Jiang S, et al. 2005. Identification of N-phenyl-N'-(2, 2, 6, 6-tetramethyl-piperidin-4-yl)-oxalamid es as a new class of HIV-1 entry inhibitors that prevent gp120 binding to CD4. Virology, 339: 213-225.
        doi: 10.1016/j.virol.2005.06.008

  • 加载中

Figures(1) / Tables(1)

Article Metrics

Article views(4049) PDF downloads(18) Cited by()

Related
Proportional views

    Novel Antiviral Agents Targeting HIV Entry and Transmission

      Corresponding author: Qinxue Hu, qhu@sgul.ac.uk; qhu@wh.iov.cn
    • 1. Center for Infection, Department of Cellular and Molecular Medicine, St George's University of London, London SW17 0RE, UK
    • 2. State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
    Fund Project:  NIH AI041346NIH AI065413the 973 Program 2006CB504200

    Abstract: Studies of the mechanism of HIV entry and transmission have identified multiple new targets for drug development. A range of inhibitors have demonstrated potent antiretroviral activity by interfering with CD4-gp120 interaction, coreceptor binding or viral-cell fusion in preclinical and clinical studies. One of these agents, fusion inhibitor enfuvirtide, is already in clinical use. Here we review the progress in the development of specific entry inhibitors as novel therapeutics. The potential of entry inhibitors as topical microbicides to block HIV transmission is also discussed.

    • The ideal strategy for viral prevention is an effective vaccine. Unfortunately, the development of a prophylactical vaccine against HIV-1 has been complicated by a number of factors such as the virus genomic variability, lack of a good animal model and lack of satisfactory surrogate markers of protective immunity, as well as the persistent nature of infection. Most importantly, HIV-1 has evolved astute mechani-sms to escape antibody and CTL responses (50, 60, 65). Despite intensive research since HIV-1 was disco-vered more than 25 years ago, there has been no success in developing a protective AIDS vaccine. In two large clinical trails finished in 2003 and 2007, respectively, neither VaxGen's gp 120 vaccine nor Merck's T cell-based vaccine showed protection. Cur-rently, multidrug combination therapies are the main strategy of HIV-1 management. Among these, protease and reverse transcriptase inhibitors, which target HIV-1 at postentry level, have had a major impact on HIV disease progression. However, the combined therapies are limited by their failure to eradicate virus, signifi-cant toxicities and the emergence of drug-resistant viral variants. Moreover, these drugs are not widely avai-lable in the developing world due to their high cost and the lack of adequate infrastructure for delivery. Thus, there is a need to identify and develop new classes of drugs to target additional stages of HIV-1 replication. Three major steps are involved in HIV-1 entry (Fig. 1): interaction of HIV-1 gp120 with cell surface CD4, gp120-CD4 complex binding with a coreceptor and membrane fusion mediated by the HIV-1 transmem-brane gp41 (3, 26, 27). Theoretically, compounds against these three steps can be considered as potential inhibitors of HIV-1 infection (Fig. 1 and Table 1) (24, 52, 68).

      Figure 1.  Potential targets for HIV-1 entry inhibition. T20 (enfuvirtide) is in clinical use. TNX-355, PRO-542, UK-427857 (maraviroc), SCH-D (vicriviroc) and AMD070 are currently in phase Ⅱ clinical trials.

      Table 1.  Entry inhibitors and the mechanisms of inhibitions

    • CD4 has been targeted for blocking HIV-1 entry since it was identified as the primary receptor of HIV-1 (12, 37). Antibodies to CD4 are among the most potent anti-HIV-1 compounds identified, but potential immunosuppressive effects limit their use for therapy (76). Nonetheless, a humanized anti-CD4 mAb, termed TNX355 (Tanox-Biogen, Houston, TX, USA), is now in clinical trials and seems not to cause immunosuppression (62). Other HIV inhibitors an-tagonizing CD4 pathway include small molecule NSC-13778 which binds to the N-terminal two-domain of CD4 (92) and CADA which down-regulates the CD4 receptor expression at post-translational level (86, 87).

    • Because CD4 has nanomolar affinity for gp120 and micromolar or lower affinity for MHC class Ⅱ, soluble mimics of CD4 represent attractive candidates for therapy. However, soluble CD4, which acts as a receptor decoy to prevent HIV-1 envelope glyco-protein (Env) engaging with CD4, showed no signi-ficant antiviral action in vivo despite its efficient neutralization in vitro (8, 11, 29). Nonetheless, PRO-542 (CD4-IgG2; Progenics Pharmaceuticals, Tarrytown, NY, USA), a tetrameric fusion protein between CD4 and immunoglobulin-G, is much more potent in vitro inhibitory activity than the parental monomer (31). When administered intravenously to HIV-1-infected adults and children at single and multiple doses ranging to 10 mg/kg, PRO-542 demon-strated excellent tolerability and pharmacology, and mediated statistically significant acute reductions in viral load (31, 69). In addition to PRO-542, other low-molecular-weight CD4 mimics include NBD-556 and NBD-557 (67, 94). A small molecule, named BMS-378806 (Bristol-Myers Squibb, Wallingford, CT, USA), has selective inhibitory activity for HIV-1 but not SIV and HIV-2 (44). Of note, BMS-378806 blocks the gp120-CD4 binding event, representing the first small molecule to inhibit this interaction (49). BMS-488043, an analogue of BMS-378806, demonstrated anti-HIV efficacy when it was orally administered (22, 33).

    • DC-SIGN (CD209), which is expressed on den-dritic cells (DCs), binds to gp120 with high affinity in addition to its natural ligand ICAM-3 (20). The interaction of HIV-1 with DC-SIGN does not result in infection of DCs, but instead enhances infection in trans of target cells that express CD4 and chemokine receptors. Since DCs normally traffic between mucosal surfaces and lymph nodes, it has been proposed that HIV-1 uses DCs as carriers, allowing the virus to access lymphoid tissue. DC-SIGN may play a critical role in mucosal transmission of virus. If so, inhibitors of DC-SIGN (e.g., monoclonal anti-bodies, sDC-SIGN) could serve as topical microbi-cides to bock sexual transmission of HIV-1 (24). In addition, HIV-1 gp120 glycoprotein can bind to heparin sulphate proteoglycans (HSPGs), glycolipid galactocerebroside (GalCer) and several other C-type lectin receptors (e.g., mannose receptor, langerin). HIV-1 can also attach to different cell types using the ancillary, cell-derived proteins that are incorporated into its membrane as the virus buds out of infected cells. Although it is still uncertain whether, and to what extent, these attachment receptors are involved in the transmission of HIV-1 in vivo, these cellular molecules could be potential targets for interrupting HIV-1 transmission (68).

    • The discovery of chemokine receptors as corecep-tors for HIV-1 has led to the search for compounds to block HIV-1 infection. Due to the importance of CCR5 in transmission and during the asymptomatic phase of HIV-1 infection (10, 23), as well as the limited impact of a loss of CCR5 function, CCR5 inhibitors are likely to be successful anti-HIV drugs. The natural chemokine ligands are candidates for use in therapy and have been shown to repress HIV-1 replication in vitro (4, 9, 58). However, concerns about chemokines as inflammatory cytokines will prevent their clinical use. Accordingly, modified chemokine antagonists and small molecules that bind to the desired receptor without triggering intracellular signalling have been the major goal in the search for coreceptor-targeted inhibitors. The N-terminal modified RANTES, aminooxypentane-RANTES (AOP-RAN TES), was identified and showed potent inhibition of R5 HIV-1 under conditions where RANTES was barely effective (71). Subsequently studies demon-strated that AOP-RANTES induced CCR5 internali-zation and inhibited recycling (48). Additionally, AOP-RANTES also inhibits HIV-1 binding to CCR3. In addition to AOP-RANTES, two modified RANTES, NNY-RANTES and PSC-RANTES have potent anti-HIV activity. The RANTES analogues showed the same rank order (PSC > NNY > AOP) in their capacity to induce prolonged CCR5 internalization, inhibit surface re-expression, and prevent HIV-1 infection (61). However, NNY-RANTES was shown to select for coreceptor switch to CXCR4-using viral variants in vivo in the hu-PBL-SCID mouse system (54). AOP-RANTES and PSC-RANTES are being tested as microbicides (34, 35, 41).

      Thus far, several other small molecules have been reported to inhibit R5 virus replication. The advantage of small molecule-based inhibitors is that they are unlikely to induce signalling and therefore to indirectly augment viral entry or to induce inflam-mation. The fist small molecule inhibitor against CCR5, TAK-779 (Takeda Chemical Industries, Japan), was reported to bind strongly to CCR5 and to a lesser extent CCR2b. TAK-779 inhibits HIV-1 replication at the membrane fusion stage by blocking the interaction between gp120 and CCR5 (1). CCR5 has a hydrop-hobic pocket that fits close to the membrane surface. TAK-779 fits into this pocket and may alter receptor conformation and thereby block virus infection. Alanine scanning mutagenesis of the transmembrane domains reveals that the binding site for TAK-779 on CCR5 is located near the extracellular surface of the receptor, within a cavity formed between transme-mbrane helices 1, 2, 3, and 7 (16). The orally bioavailable versions of TAK, TAK-220 and TAK-652 have shown potent antiviral activity in vitro and are currently under in vivo study (2, 77). The potent CCR5 inhibitor, SCH-C (SCH-351125), is an oxime-piperidine compound with oral bioavailability and potent anti-HIV activity, which has been tested in HIV-1-infected individuals. 25 mg SCH-C was orally administrated twice a day for 10 days. This dose was well tolerated overall and led to > 0.5 log10 reductions in viral load in 10 out of 12 patients (57, 75). The analogous of SCH, SCH-D (SCH-417690 or vicriviroc; Schering-Plough, Kenil-worth, USA) and AD101 (SCH-350581), potently inhibit HIV-1 replication in primary lymphocytes, as well as viral entry and gp 120 binding to cell lines (74, 80, 81). Other CCR5 anta-gonists under in vitro or in vivo studies include UK-427857 (maraviroc; Pfizer, Sandwich, UK), GW-873140 (aplaviroc; Glaxo-SimthKline, London, UK), CMPD167 (Merck Research Labs, San Diego, CA, USA) and 1, 3, 3, 4-tetrasubstituted pyrrolidine deri-vatives (47, 84, 88, 89).

      It has been documented that blocking CCR5-mediated HIV-1 transmission by β chemokines can be enhanced by the complementary effect of anti-CCR5 antibodies (42, 43). Because cross-reactivity with other G protein coupled receptors (GPCRs) can be an important source of toxicity for small-molecule antagonists and modified chemokines, monoclonal antibodies that have exquisite specificity for CCR5 may provide an attractive mode of therapy. A broadly inhibitory monoclonal antibody, PRO-140 (PA14) potently inhibits HIV-1 entry at concentrations that do not affect CCR5's chemokine receptor activity (59, 79). PRO140 does not induce signalling or down-regulation of CCR5. Its antiviral effect is probably exerted through a mechanism involving receptor blockade.

    • The viral component involved in the interaction of HIV-1 with CXCR4 is the V3 loop of the viral Env gp120. The natural ligand for CXCR4, SDF-1, is highly basic with overall charge of +8, just like the V3 loop of gp120, while the corresponding net charge of the extracellular regions of CXCR4 is-9. Several cationic, arginine-rich peptides, ALX40-4C (N-α-acetyl-nona-D-arginine), T-22 and its shortened version T-140 and T-134, bind to CXCR4 and prevent X4 virus infection (15, 56, 78). The small molecule, AMD3100, a bicyclam derivative, binds and antag-onizes signalling via CXCR4. AMD3100 not only strongly inhibits X4 HIV-1 replication but also blocks cell-surface-expressed HIV-1-Env-induced apoptosis of uninfected cells (14, 66). Its analogue AMD3465 is 10-fold more effective as a CXCR4 antagonist alt-hough oral bioavailability is not yet achieved (21). Nevertheless, the derivative AMD070 (AMD11070) shows good oral bioavailability and tolerance (73). KRH-1636 is another CXCR4 inhibitor (30). KRH-1636 can be absorbed through the duodenum and blocks infection of primary cells by primary and laboratory-adapted X4 viruses with similar efficiency to those of AMD3100 (30). Although CXCR4 antagonists have potential for AIDS treatment, challenges to therapies targeting CXCR4 include the wide tissue distribution of CXCR4 and the low prevalence of patients having only X4 virus.

      In addition to CCR5 or CXCR4 specific drugs, development of novel inhibitors targeting both CCR5 and CXCR4 is not impossible. For instance, the viral chemokine vMIP2 can antagonize CCR5 and CXCR4 and hence blocks the infection of both R5 and X4 viruses (5, 38). A recently generated CCR2 antibody, which induces oligomerization of CCR2 with CCR5 or CXCR4, can prevent the infection of HIV-1 through CCR5 and CXCR4 (63).

    • The most widely accepted model describing HIV-1 Env-mediated membrane fusion postulates that CD4 or/and coreceptor binding results in the formation of a coiled-coil in gp41 (7, 90). It induces the exposure of the gp41 fusion peptide, the formation of the six-helix bundle and ultimately membrane fusion. Env regions that are targets for antiviral agents are transiently exposed during the course of fusion. Several synthetic peptides derived from the first helical region (HR1 or NHR) and the second helical region (HR2 or CHR), termed as N-and C-peptides, have anti-fusion activities (19, 32). By far, the most potent fusion inhibitors, including T20 and C34, are C-peptides. T20 (originally called DP178), a 36-mer peptide base on HR2 in gp41, was previously believed to bind to the exposed grooves on the surface of the triple stranded coiled-coil, preventing the formation of six-helix bundle and membrane fusion (36). Of interest, recent studies indicate that T20 inhibits HIV-1 entry by targeting multiple sites in gp41 and gp120 (45, 46). T20 is now a licensed drug (enfuvirtide or fuzeon; Trimeris, Morrisville, NC, USA] (39, 40). T1249, consisting of 39 L-amino acids, is a second generation HR2 peptide mimetic that combines the virtues of T-20 with sequence changes intended to increase its potency and breadth of action. C34, also based on gp41 HR2, was identified by the protein dissection method and comprises 34 amino acids spanning gp41 residues 628-661. It is soluble in aqueous solution and is a highly effective inhibitor of HIV-1 entry and cell-cell fusion (6). More recently, Sia et al has designed a potent, conformationally stable, anti-HIV-1 peptide based on the liner peptide C34, named C34 coil, using protein grafting techniques (70). A converse inhibition strategy that targets the C-terminal heptad repeat region has been employed by Root et al (64). This protein, denoted 5-Helix, comprises the six-helix bundle lacking one C-terminal peptide. The 5-Helix protein binds to the C-peptide region of gp41 and displays potent (nanomolar) inhibitory activity against diverse HIV-1 variants (64). More recently, C52L, a bacterially expressed recombinant peptide inhibitor that includes both the C-peptide and tryptophan-rich region, has demonstrated potent anti-HIV activity in both in vitro and in vitro studies (13, 85). Because C52L can be expressed in bacteria, it might be more economical to manufacture on a large scale than T-20-like peptides produced by chemical synthesis (13). Of great interest, the discovery of a natural HIV-1 fusion inhibitor from human blood, designated as VIRIP (virus-inhibitory peptide), may lead to the development of a new class of antiretroviral drugs (55). Apart from peptide fusion inhibitors, another non-peptide fusion inhibitor, designated ADS-J1, is a phenylazo-naphthalene sulfonic acid derivative, and was found to inhibit the formation of the fusion-driving six-helix bundle and HIV-1 Env-mediated membrane fusion (32).

    • In addition to being used therapeutically, entry inhibitors could be used prophylactically to prevent HIV transmission. Newly acquired HIV-1 infections are largely the result of heterosexual contact. World-wide, women face a growing risk of infection with HIV-1 (83). In the absence of an effective vaccine, mechanical barriers such as condoms can be effective in preventing sexual transmission of HIV-1. However, this method is not always accepted by male partners or is impractical (e.g., contraceptive) for use by woman. Therefore, there is an urgent need for additional interventions to prevent new HIV-1 infections. Vaginal microbicides may represent an additional important method, under the personal control of women, that can be used for the prevention of HIV-1 as well as other sexually transmitted diseases (17, 51, 68).

      To prevent HIV-1 transmission, a microbicide must inactivate the virus (both free and cell-associated) while it is still in the vaginal lumen, prevent the virus from attaching to and fusion with host cells, or prevent the virus from replicating in susceptible cells. Ideally, these topical microbicides should be inex-pensive, easy to use, stable under low pH conditions, colourless, tasteless, and nonirritating to genital mucosal tissues (72, 82). To date, three microbicides tested in phase Ⅲ trials, nonoxynol-9 (N9), Savvy gel and cellulose sulphate, all belong to a'first generation' of products that aim to make the vagina less hospitable to HIV, but the results from these trials were disappointing. For N9, the failure is considered to be related to the toxicity of N9 on the vaginal mucosa, as repeated exposure may lead to inflam-mation and epithelial disruption, which may attract CD4+ T cells and facilitate HIV-1 infection (18, 91). Therefore, non-toxic and specific inhibitors, including those already in clinical trial as new anti-HIV drugs (57, 93), might be more suitable for long-term use intravaginally without destroying the integrity of the epithelium barrier. The virus attachment and fusion stages of the HIV-1 life cycle provide attractive targets for the development of mechanism-based, HIV-1-specfic microbicides. Because the mechanism of HIV-1 entry is the same for all the genetic subtypes, specific inhibitors of HIV-1 attachment, fusion and entry could prevent or hinder vaginal or rectal transmission of HIV-1, when properly formulated and applied to women or men prior to sexual intercourse (24, 53, 68, 85). Apart from specific entry/fusion inhibitors, com-pounds like carbohydrage-binding agents and poly-anions, are also potential microbicide candidates (25, 28, 68).

    Figure (1)  Table (1) Reference (94) Relative (20)

    目录

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return