Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Strategies in the design of antiviral drugs

Key Points

  • Greater understanding of viral life cycles has resulted in the discovery and validation of several targets for therapeutic intervention, and an increase in the number of antiviral drugs from 5 in 1990 to over 30 in 2001. However, there is considerable room for improvement, as these compounds are not always efficacious (owing to virus resistance) or well tolerated.

  • Antiviral drug design could, in principle, be targeted at either viral proteins or cellular proteins. The first approach is likely to yield more specific, less toxic compounds, with a narrow spectrum of antiviral activity and a higher likelihood of resistance developing. The second approach might afford antiviral compounds with a broader activity spectrum and less chance of resistance developing, but higher likelihood of toxicity.

  • Some stages in the viral life cycle that can, or could, be targeted by drugs include virus adsorption, virus–cell fusion, viral DNA or RNA synthesis and viral enzymes, such as HIV protease and influenza virus neuraminidase. Two host cellular enzymes, inosine 5′-monophosphate dehydrogenase and S-adenosylhomocysteine hydrolase, could also be targets for certain classes of antiviral agents.

  • Licensed drugs include:

  • Inhibitors of viral DNA polymerases: nucleoside analogues such as acyclovir, and acyclic nucleoside phosphonates such as cidofovir.

  • Inhibitors of HIV reverse transcriptase: nucleoside reverse transcriptase inhibitors such as azidothymidine, non-nucleoside reverse transcriptase inhibitors such as nevirapine, and acyclic nucleoside phosphonates such as tenofovir and adefovir.

  • Inhibitors of HIV protease, such as indinavir.

  • Inhibitors of influenza virus neuraminidase, such as zanamivir.

  • IMP dehydrogenase inhibitors, such as ribavirin and mycophenolic acid.

  • Drugs in development that are not in the above classes include:

  • Inhibitors of virus adsorption, such as polyanions.

  • Inhibitors of virus–cell fusion, such as AMD3100, TAK779 and T20.

  • Inhibitors of human rhinovirus proteases, such as AG7088.

Abstract

A decade ago, just five drugs were licensed for the treatment of viral infections. Since then, greater understanding of viral life cycles, prompted in particular by the need to combat human immunodeficiency virus, has resulted in the discovery and validation of several targets for therapeutic intervention. Consequently, the current antiviral repertoire now includes more than 30 drugs. But we still lack effective therapies for several viral infections, and established treatments are not always effective or well tolerated, highlighting the need for further refinement of antiviral drug design and development. Here, I describe the rationale behind current and future drug-based strategies for combating viral infections.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: The viral life cycle, as exemplified by HIV.
Figure 2: Basic (skeletal) pharmacophores or prototypic compounds of the classes of antiviral agents described in this review.
Figure 3: Interaction of CCR5 with TAK779.
Figure 4: Examples of antiviral nucleoside analogues acting by a chain termination mechanism.
Figure 5: Interaction of HIV-1 RT with UC781.
Figure 6: Interaction of HIV protease with KNI272.
Figure 7: Interaction of influenza neuraminidase with oseltamivir.

Similar content being viewed by others

References

  1. De Clercq, E. Antiviral drugs: current state of the art. J. Clin. Virol. 22, 73–89 (2001).

    CAS  PubMed  Google Scholar 

  2. Gallaher, W. R., Ball, J. M., Garry, R. F., Martin-Amedee, A. M. & Montelaro, R. C. A general model for the surface glycoproteins of HIV and other retroviruses. AIDS Res. Hum. Retroviruses 11, 191–202 (1995).

    CAS  PubMed  Google Scholar 

  3. Shukla, D. et al. A novel role for 3-O-sulfated heparan sulfate in herpes simplex virus 1 entry. Cell 99, 13–22 (1999).

    CAS  PubMed  Google Scholar 

  4. Chen, Y. et al. Dengue virus infectivity depends on envelope protein binding to target cell heparan sulfate. Nature Med. 3, 866–871 (1997).

    CAS  PubMed  Google Scholar 

  5. Herold, B. C. et al. Poly(sodium 4-styrene sulfonate): an effective candidate topical antimicrobial for the prevention of sexually transmitted diseases. J. Infect. Dis. 181, 770–773 (2000).

    CAS  PubMed  Google Scholar 

  6. Esté, J. A. et al. Development of resistance of human immunodeficiency virus type 1 to dextran sulfate associated with the emergence of specific mutations in the envelope gp120 glycoprotein. Mol. Pharmacol. 52, 98–104 (1997).

    PubMed  Google Scholar 

  7. Esté, J. A. et al. Human immunodeficiency virus glycoprotein gp120 as the primary target for the antiviral action of AR177 (zintevir). Mol. Pharmacol. 53, 340–345 (1998).

    PubMed  Google Scholar 

  8. Cabrera, C. et al. Resistance of the human immunodeficiency virus to the inhibitory action of negatively charged albumins on virus binding to CD4. AIDS Res. Hum. Retroviruses 15, 1535–1543 (1999).

    CAS  PubMed  Google Scholar 

  9. Schols, D. et al. Inhibition of T-tropic HIV strains by selective antagonization of the chemokine receptor CXCR4. J. Exp. Med. 186, 1383–1388 (1997).Describes, for the first time, the specific antagonistic effect of the bicyclam AMD3100 against the binding of T-lymphotropic (X4) HIV strains with their co-receptor CXCR4, an essential step for entry of these viruses into the cells.

    CAS  PubMed  PubMed Central  Google Scholar 

  10. De Clercq, E. Inhibition of HIV infection by bicyclams, highly potent and specific CXCR4 antagonists. Mol. Pharmacol. 57, 833–839 (2000).

    CAS  PubMed  Google Scholar 

  11. Baba, M. et al. A small-molecule, nonpeptide CCR5 antagonist with highly potent and selective anti-HIV-1 activity. Proc. Natl Acad. Sci. USA 96, 5698–5703 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Dragic, T. et al. 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 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Hatse, S. et al. Mutation of Asp171 and Asp262 of the chemokine receptor CXCR4 impairs its coreceptor function for human immunodeficiency virus-1 entry and abrogates the antagonistic activity of AMD3100. Mol. Pharmacol. 60, 164–173 (2001).

    CAS  PubMed  Google Scholar 

  14. Strizki, J. M. et al. 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 (2001).The most recent report on a potential anti-HIV drug candidate that blocks HIV infection through an antagonistic effect on the CCR5 co-receptor, which is used by macrophage-tropic (R5) HIV strains to enter cells.

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Root, M. J., Kay, M. S. & Kim, P. S. Protein design of an HIV-1 entry inhibitor. Science 291, 884–888 (2001).

    CAS  PubMed  Google Scholar 

  16. Eckert, D. M., Malashkevich, V. N., Hong, L. H., Carr, P. A. & Kim, P. S. Inhibiting HIV-1 entry: discovery of d-peptide inhibitors that target the gp41 coiled–coil pocket. Cell 99, 103–115 (1999).

    CAS  PubMed  Google Scholar 

  17. Kilby, J. M. et al. Potent suppression of HIV-1 replication in humans by T-20, a peptide inhibitor of gp41-mediated virus entry. Nature Med. 4, 1302–1307 (1998).

    CAS  PubMed  Google Scholar 

  18. Lambert, D. M. et al. Peptides from conserved regions of paramyxovirus fusion (F) proteins are potent inhibitors of viral fusion. Proc. Natl Acad. Sci. USA 93, 2186–2191 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Schwartz, J. A., Lium, E. K. & Silverstein, S. J. Herpes simplex virus type 1 entry is inhibited by the cobalt chelate complex CTC-96. J. Virol. 75, 4117–4128 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. McGuigan, C. et al. Potent and selective inhibition of varicella-zoster virus (VZV) by nucleoside analogues with an unusual bicyclic base. J. Med. Chem. 42, 4479–4484 (1999).

    CAS  PubMed  Google Scholar 

  21. McGuigan, C. et al. Highly potent and selective inhibition of varicella-zoster virus by bicyclic furopyrimidine nucleosides bearing an aryl side chain. J. Med. Chem. 43, 4993–4997 (2000).References 20 and 21 provide an example of how classical medicinal chemistry can lead to the discovery of an entirely new class of nucleoside analogues with exquisite antiviral activity (that is, inhibition of varicella-zoster virus replication at subnanomolar concentrations).

    CAS  PubMed  Google Scholar 

  22. Iwayama, S. et al. Antiherpesvirus activities of (1′S,2′R)-9-[[1′,2′-bis(hydroxymethyl)-cycloprop-1′-yl]methyl] (A-5021) in cell culture. Antimicrob. Agents Chemother. 42, 1666–1670 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Ono, N. et al. Mode of action of (1′S,2′R)-9-[[1′,2′-bis(hydroxymethyl)cycloprop-1′-yl]methyl]guanine (A-5021) against herpes simplex virus type 1 and type 2 and varicella-zoster virus. Antimicrob. Agents Chemother. 42, 2095–2102 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Wang, J. et al. The cyclohexene ring system as a furanose mimic: synthesis and antiviral activity of both enantiomers of cyclohexenylguanine. J. Med. Chem. 43, 736–745 (2000).

    CAS  PubMed  Google Scholar 

  25. Siddiqui, A. Q. et al. Design and synthesis of lipophilic phosphoramidate d4T-MP prodrugs expressing high potency against HIV in cell culture: structural determinants for in vitro activity and QSAR. J. Med. Chem. 42, 4122–4128 (1999).

    CAS  PubMed  Google Scholar 

  26. Saboulard, D. et al. Characterization of the activation pathway of phosphoramidate triester prodrugs of stavudine and zidovudine. Mol. Pharmacol. 56, 693–704 (1999).

    CAS  PubMed  Google Scholar 

  27. Meier, C., Lorey, M., De Clercq, E. & Balzarini, J. cycloSal-2′,3′-dideoxy-2′,3′-didehydrothymidine monophosphate (cycloSal-d4TMP): synthesis and antiviral evaluation of a new d4TMP delivery system. J. Med. Chem. 41, 1417–1427 (1998).

    CAS  PubMed  Google Scholar 

  28. Balzarini, J. et al. Cyclosaligenyl-2′,3′-didehydro-2′,3′-dideoxythymidine monophosphate: efficient intracellular delivery of d4TMP. Mol. Pharmacol. 58, 928–935 (2000).

    CAS  PubMed  Google Scholar 

  29. De Clercq, E. The role of non-nucleoside reverse transcriptase inhibitors (NNRTIs) in the therapy of HIV-1 infection. Antiviral Res. 38, 153–179 (1998).

    CAS  PubMed  Google Scholar 

  30. Jonckheere, H., Anné, J. & De Clercq, E. The HIV-1 reverse transcription (RT) process as target for RT inhibitors. Med. Res. Rev. 20, 129–154 (2000).

    CAS  PubMed  Google Scholar 

  31. Esnouf, R. M., Stuart, D. I., De Clercq, E., Schwartz, E. & Balzarini, J. Models which explain the inhibition of reverse transcriptase by HIV-1-specific (thio)carboxanilide derivatives. Biochem. Biophys. Res. Commun. 234, 458–464 (1997).

    CAS  PubMed  Google Scholar 

  32. Ren, J. 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. 275, 14316–14320 (2000).Reference 32 emphasizes that future design of new non-nucleoside HIV-1 reverse transcriptase inhibitors could be rationally guided by resilience to drug-resistance mutations.

    CAS  PubMed  Google Scholar 

  33. 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. 57, 954–960 (2000).

    CAS  PubMed  Google Scholar 

  34. Xiong, X., Smith, J. L. & Chen, M. S. Effect of incorporation of cidofovir into DNA by human cytomegalovirus DNA polymerase on DNA elongation. Antimicrob. Agents Chemother. 41, 594–599 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Balzarini, J., Hao, Z., Herdewijn, P., Johns, D. G. & De Clercq, E. Intracellular metabolism and mechanism of anti-retrovirus action of 9-(2-phosphonylmethoxyethyl)adenine, a potent anti-human immunodeficiency virus compound. Proc. Natl Acad. Sci. USA 88, 1499–1503 (1991).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. De Clercq, E. et al. A novel selective broad-spectrum anti-DNA virus agent. Nature 323, 464–467 (1986).

    CAS  PubMed  Google Scholar 

  37. De Clercq, E. Therapeutic potential of HPMPC as an antiviral drug. Rev. Med. Virol. 3, 85–96 (1993).

    CAS  Google Scholar 

  38. De Clercq, E. Vaccinia virus inhibitors as a paradigm for the chemotherapy of poxvirus infections. Clin. Microbiol. Rev. 14, 382–397 (2001).Describes several compounds that are effective against vaccinia virus, and that could, therefore, be considered for the therapy of poxvirus infections, including smallpox, should the need arise. The prominent candidate is cidofovir, which has already proved to be effective in the treatment of poxvirus infections, such as molluscum contagiosum, in humans.

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Pommier, Y, Pilon, A. A., Bajaj, K., Mazumder, A. & Neamati, N. HIV-1 integrase as a target for antiviral drugs. Antiviral Chem. Chemother. 8, 463–483 (1997).

    CAS  Google Scholar 

  40. Pommier, Y., Marchand, C. & Neamati, N. Retroviral integrase inhibitors year 2000: update and perspectives. Antiviral Res. 47, 139–148 (2000).

    CAS  PubMed  Google Scholar 

  41. Pluymers, W. et al. Viral entry as the primary target for the anti-HIV activity of chicoric acid and its tetra-acetyl esters. Mol. Pharmacol. 58, 641–648 (2000).References 41 and 48 show that caution should be exercised in identifying the primary molecular target in the mode of action of antiviral compounds, in particular, anti-HIV agents, which are capable of interacting at several steps in the viral replication cycle.

    CAS  PubMed  Google Scholar 

  42. Hazuda, D. J. et al. Inhibitors of strand transfer that prevent integration and inhibit HIV-1 replication in cells. Science 287, 646–650 (2000).

    CAS  PubMed  Google Scholar 

  43. Wai, J. S. et al. 4-Aryl-2,4-dioxobutanoic acid inhibitors of HIV-1 integrase and viral replication in cells. J. Med. Chem. 43, 4923–4926 (2000).

    CAS  PubMed  Google Scholar 

  44. Daelemans, D., Vandamme, A.-M. & De Clercq, E. Human immunodeficiency virus gene regulation as a target for antiviral chemotherapy. Antiviral Chem. Chemother. 10, 1–14 (1999).

    CAS  Google Scholar 

  45. Baba, M. et al. Inhibition of human immunodeficiency virus type 1 replication and cytokine production by fluoroquinoline derivatives. Mol. Pharmacol. 53, 1097–1103 (1998).

    CAS  PubMed  Google Scholar 

  46. Turpin, J. A. et al. Inhibition of acute-, latent-, and chronic-phase human immunodeficiency virus type 1 (HIV-1) replication by a bistriazoloacridone analog that selectively inhibits HIV-1 transcription. Antimicrob. Agents Chemother. 42, 487–494 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Hamy, F. et al. An inhibitor of the Tat/TAR RNA interaction that effectively suppresses HIV-1 replication. Proc. Natl Acad. Sci. USA 94, 3548–3553 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Daelemans, D. et al. A second target for the peptoid Tat/transactivation response element inhibitor CGP64222: inhibition of human immunodeficiency virus replication by blocking CXC-chemokine receptor 4-mediated virus entry. Mol. Pharmacol. 57, 116–124 (2000).

    CAS  PubMed  Google Scholar 

  49. Chao, S. H. et al. Flavopiridol inhibits P-TEFb and blocks HIV-1 replication. J. Biol. Chem. 275, 28345–28348 (2000).

    CAS  PubMed  Google Scholar 

  50. Dymock, B. W., Jones, P. S. &, Wilson, F. X. Novel approaches to the treatment of hepatitis C virus infection. Antiviral Chem. Chemother. 11, 79–96 (2000).

    CAS  Google Scholar 

  51. Paolini, C., De Francesco, R. & Gallinari, P. Enzymatic properties of hepatitis C virus NS3-associated helicase. J. Gen. Virol. 81, 1335–1345 (2000).

    CAS  PubMed  Google Scholar 

  52. Carroll, S. S. et al. Only a small fraction of purified hepatitis C RNA-dependent RNA polymerase is catalytically competent: implications for viral replication and in vitro assays. Biochemistry 39, 8243–8249 (2000).

    CAS  PubMed  Google Scholar 

  53. Patick, A. K. & Potts, K. E. Protease inhibitors as antiviral agents. Clin. Microbiol. Rev. 11, 614–627 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Erickson, J. W. & Burt, S. K. Structural mechanisms of HIV drug resistance. Annu. Rev. Pharmacol. Toxicol. 36, 545–571 (1996).

    CAS  PubMed  Google Scholar 

  55. Turner, S. R. et al. Tipranavir (PNU-140690): a potent, orally bioavailable nonpeptidic HIV protease inhibitor of the 5,6-dihydro-4-hydroxy-2-pyrone sulfonamide class. J. Med. Chem. 41, 3467–3476 (1998).

    CAS  PubMed  Google Scholar 

  56. Hagen, S. E. et al. 4-Hydroxy-5,6-dihydropyrones as inhibitors of HIV protease: the effect of heterocyclic substituents at C-6 on antiviral potency and pharmacokinetic parameters. J. Med. Chem. 44, 2319–2332 (2001).

    CAS  PubMed  Google Scholar 

  57. Waxman, L. & Darke, P. L. The herpesvirus proteases as targets for antiviral chemotherapy. Antiviral Chem. Chemother. 11, 1–22 (2000).

    CAS  Google Scholar 

  58. Jarvest, R. L. et al. Inhibition of herpes proteases and antiviral activity of 2-substituted thieno-[2,3-d]oxazinones. Bioorg. Med. Chem. Lett. 9, 443–448 (1999).

    CAS  PubMed  Google Scholar 

  59. Smith, D. G. et al. The inhibition of human cytomegalovirus (hCMV) protease by hydroxylamine derivatives. Bioorg. Med. Chem. Lett. 9, 3137–3142 (1999).

    CAS  PubMed  Google Scholar 

  60. Ogilvie, W. W. et al. Synthesis and antiviral activity of monobactams inhibiting the human cytomegalovirus protease. Bioorg. Med. Chem. 7, 1521–1531 (1999).

    CAS  PubMed  Google Scholar 

  61. Borthwick, A. D. et al. Design and synthesis of pyrrolidine-5,5-trans-lactams (5-oxo-hexahydro-pyrrolo[3,2-b]pyrroles) as novel mechanism-based inhibitors of human cytomegalovirus protease. 1. The α-methyl-trans-lactam template. J. Med. Chem. 43, 4452–4464 (2000).

    CAS  PubMed  Google Scholar 

  62. Matsumoto, M., Misawa, S., Chiba, N., Takaku, H. & Hayashi, H. Selective nonpeptidic inhibitors of herpes simplex virus type 1 and human cytomegalovirus proteases. Biol. Pharm. Bull. 24, 236–241 (2001).

    CAS  PubMed  Google Scholar 

  63. Dragovich, P. S. et al. Structure-based design, synthesis, and biological evaluation of irreversible human rhinovirus 3C protease inhibitors. 1. Michael acceptor structure–activity studies. J. Med. Chem. 41, 2806–2818 (1998).

    CAS  PubMed  Google Scholar 

  64. Dragovich, P. S. et al. Structure-based design, synthesis, and biological evaluation of irreversible human rhinovirus 3C protease inhibitors. 2. Peptide structure–activity studies. J. Med. Chem. 41, 2819–2834 (1998).

    CAS  PubMed  Google Scholar 

  65. Dragovich, P. S. et al. Structure-based design, synthesis, and biological evaluation of irreversible human rhinovirus 3C protease inhibitors. 3. Structure–activity studies of ketomethylene-containing peptidomimetics. J. Med. Chem. 42, 1203–1212 (1999).

    CAS  PubMed  Google Scholar 

  66. Dragovich, P. S. et al. Structure-based design, synthesis, and biological evaluation of irreversible human rhinovirus 3C protease inhibitors. 4. Incorporation of P1 lactam moieties as l-glutamine replacements. J. Med. Chem. 42, 1213–1224 (1999).

    CAS  PubMed  Google Scholar 

  67. Matthews, D. A. et al. Structure-assisted design of mechanism-based irreversible inhibitors of human rhinovirus 3C protease with potent antiviral activity against multiple rhinovirus serotypes. Proc. Natl Acad. Sci. USA 96, 11000–11007 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Patick, A. K. et al. In vitro antiviral activity of AG7088, a potent inhibitor of human rhinovirus 3C protease. Antimicrob. Agents Chemother. 43, 2444–2450 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Tautz, N., Kaiser, A. & Thiel, H.-J. NS3 serine protease of bovine viral diarrhea virus: characterization of active site residues, NS4A cofactor domain, and protease-cofactor interactions. Virology 273, 351–363 (2000).

    CAS  PubMed  Google Scholar 

  70. Llinàs-Brunet, M. et al. Highly potent and selective peptide-based inhibitors of the hepatitis C virus serine protease: towards smaller inhibitors. Bioorg. Med. Chem. Lett. 10, 2267–2270 (2000).

    PubMed  Google Scholar 

  71. Bennett, J. M. et al. The identification of α-ketoamides as potent inhibitors of hepatitis C virus NS3–4A proteinase. Bioorg. Med. Chem. Lett. 11, 355–357 (2001).

    CAS  PubMed  Google Scholar 

  72. Sing, W. T., Lee, C. L., Yeo, S. L., Lim, S. P. & Sim, M. M. Arylalkylidene rhodanine with bulky and hydrophobic functional group as selective HCV NS3 protease inhibitor. Bioorg. Med. Chem. Lett. 11, 91–94 (2001).

    CAS  PubMed  Google Scholar 

  73. Von Itzstein, M. et al. Rational design of potent sialidase-based inhibitors of influenza virus replication. Nature 363, 418–423 (1993).The first demonstration of how computer-assisted drug design, based on the crystal structure of the influenza viral neuraminidase, could lead to the development of new antiviral drugs.

    CAS  PubMed  Google Scholar 

  74. Barnett, J. M. et al. Zanamivir susceptibility monitoring and characterization of influenza virus clinical isolates obtained during phase II clinical efficacy studies. Antimicrob. Agents Chemother. 44, 78–87 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Hayden, F. G. et al. Efficacy and safety of the neuraminidase inhibitor zanamivir in the treatment of influenza virus infections. N. Engl. J. Med. 337, 874–880 (1997).

    CAS  PubMed  Google Scholar 

  76. Kim, C. U. et al. Influenza neuraminidase inhibitors possessing a novel hydrophobic interaction in the enzyme active site: design, synthesis, and structural analysis of carbocyclic sialic acid analogues with potent anti-influenza activity. J. Am. Chem. Soc. 119, 681–690 (1997).

    CAS  PubMed  Google Scholar 

  77. Mendel, D. B. et al. Oral administration of a prodrug of the influenza virus neuraminidase inhibitor GS 4071 protects mice and ferrets against influenza infection. Antimicrob. Agents Chemother. 42, 640–646 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Hayden, F. G. et al. Use of the oral neuraminidase inhibitor oseltamivir in experimental human influenza: randomized controlled trials for prevention and treatment. JAMA 282, 1240–1246 (1999).

    CAS  PubMed  Google Scholar 

  79. Nicholson, K. G. et al. Efficacy and safety of oseltamivir in treatment of acute influenza: a randomised controlled trial. Lancet 355, 1845–1850 (2000).

    CAS  PubMed  Google Scholar 

  80. Smee, D. F., Huffman, J. H., Morrison, A. C., Barnard, D. L. & Sidwell, R. W. Cyclopentane neuraminidase inhibitors with potent in vitro anti-influenza virus activities. Antimicrob. Agents Chemother. 45, 743–748 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Sidwell, R. W. et al. In vivo influenza virus-inhibitory effects of the cyclopentane neuraminidase inhibitor RWJ-270201. Antimicrob. Agents Chemother. 45, 749–757 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Bantia, S. et al. Comparison of the anti-influenza virus activity of RWJ-270201 with those of oseltamivir and zanamivir. Antimicrob. Agents Chemother. 45, 1162–1167 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Sintchak, M. D. et al. Structure and mechanism of inosine monophosphate dehydrogenase in complex with the immunosuppressant mycophenolic acid. Cell 85, 921–930 (1996).

    PubMed  Google Scholar 

  84. De Clercq, E. et al. Antiviral activities of 5-ethynyl-1-β-d-ribofuranosylimidazole-4-carboxamide and related compouinds. Antimicrob. Agents Chemother. 35, 679–684 (1991).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Markland, W., McQuaid, T. J., Jain, J. & Kwong, A. D. Broad-spectrum antiviral activity of the IMP dehydrogenase inhibitor VX-497: a comparison with ribavirin and demonstration of antiviral additivity with α-interferon. Antimicrob. Agents Chemother. 44, 859–866 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Neyts, J., Andrei, G. & De Clercq, E. The novel immunosuppressive agent mycophenolate mofetil markedly potentiates the antiherpesvirus activities of acyclovir, ganciclovir, and penciclovir in vitro and in vivo. Antimicrob. Agents Chemother. 42, 216–222 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Margolis, D. et al. Abacavir and mycophenolic acid, an inhibitor of inosine monophosphate dehydrogenase, have profound and synergistic anti-HIV activity. J. Acquir. Immune Defic. Syndr. 21, 362–370 (1999).

    CAS  PubMed  Google Scholar 

  88. Saracco, G. et al. A randomized 4-arm multicenter study of interferon-α-2b plus ribavirin in the treatment of patients with chronic hepatitis C not responding to interferon alone. Hepatology 34, 133–138 (2001).

    CAS  PubMed  Google Scholar 

  89. Crotty, S. et al. The broad-spectrum antiviral ribonucleoside ribavirin is an RNA virus mutagen. Nature Med. 6, 1375–1379 (2000).

    CAS  PubMed  Google Scholar 

  90. Crotty, S., Cameron, C. E. & Andino, R. RNA virus error catastrophe: direct molecular test by using ribavirin. Proc. Natl Acad. Sci. USA 98, 6895–6900 (2001).References 89 and 90 afford a rather provocative account of how mutagens that induce 'error catastrophe' could be used in antiviral strategies, at least for some RNA viruses.

    CAS  PubMed  PubMed Central  Google Scholar 

  91. De Clercq, E. et al. Broad-spectrum antiviral activities of neplanocin A, 3-deazaneplanocin A, and their 5′-nor derivatives. Antimicrob. Agents Chemother. 33, 1291–1297 (1989).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Bray, M., Davis, K., Geisbert, T., Schmaljohn, C. & Huggins, J. A mouse model for evaluation of prophylaxis and therapy of Ebola hemorrhagic fever. J. Infect. Dis. 179 (Suppl. 1), S248–S258 (1999).

    PubMed  Google Scholar 

  93. Huggins, J., Zhang, Z.-X. & Bray, M. Antiviral drug therapy of filovirus infections: S-adenosylhomocysteine hydrolase inhibitors inhibit Ebola virus in vitro and in a lethal mouse model. J. Infect. Dis. 179, (Suppl. 1) S240–S247 (1999).

    CAS  PubMed  Google Scholar 

  94. Bray, M., Driscoll, J. & Huggins, J. W. Treatment of lethal Ebola virus infection in mice with a single dose of an S-adenosyl-l-homocysteine hydrolase inhibitor. Antiviral Res. 45, 135–147 (2000).Most virus infections are amenable to antiviral therapy, and this also holds for such feared viral pathogens as Ebola.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

E.D.C. holds the Professor P. De Somer Chair of Microbiology at the School of Medicine, Katholieke Universiteit Leuven, Belgium, and thanks C. Callebaut for her invaluable editorial assistance.

Author information

Authors and Affiliations

Authors

Supplementary information

Related links

Related links

DATABASES

LocusLink

CCR5

CDK9

CXCR4

IMP dehydrogenase

interferon-α

MIP1

NF-κB

RANTES

SAH hydrolase

SDF1

Medscape DrugInfo

abacavir

acyclovir

amprenavir

cidofovir

delavirdine

didanosine

efavirnez

famciclovir

ganciclovir

indinavir

lamivudine

lopinavir

nelfinavir

nevirapine

oseltamivir

penciclovir

ribavirin

ritonavir

saquinavir

stavudine

valaciclovir

valganciclovir

zanamivir

zalcitabine

zidovudine

Protein Data Bank

GS4071

IMP dehydrogenase

NS3 helicase

NS5B

FURTHER INFORMATION

Encyclopedia of Life Sciences

Antiviral drugs

Glossary

VIRION

A mature infectious virus particle.

V3 LOOP

The gp120 protein has eleven defined loop segments, five of which are termed variable (designated V1–V5).

ENVELOPE

A lipoprotein-bilayer outer membrane of many viruses. Envelope proteins often aid in identifying and attaching the virus to a cell-surface receptor, whereby viral entry can occur.

PRODRUG

A pharmacologically inactive compound that is converted to the active form of the drug by endogenous enzymes or metabolism. It is generally designed to overcome problems associated with stability, toxicity, lack of specificity or limited (oral) bioavailability.

CONGENER

Any member of the same chemical family.

ALLOSTERIC SITES

Two or more topologically distinct binding sites within a protein can interact functionally with each other. So, two sites in different positions can bind ligands (substrates, inhibitors and so on), and binding of a ligand at one site alters the properties of the other(s).

CPK COLOURING

The CPK colour scheme for elements is based on the colours of the popular plastic space-filling models developed by Corey, Pauling and Kultun, and is conventionally used by chemists. In this scheme, carbon is represented in light grey, oxygen in red, nitrogen in blue, sulphur in yellow, hydrogen in white and chlorine in green.

SEROTYPE

Variety of a species (usually bacteria or virus) characterized by its antigenic properties.

PEGYLATION

Addition of poly(ethylene glycol) (PEG) groups to proteins can increase their resistance to proteolytic degradation, improve their water solubility and reduce their antigenicity.

Rights and permissions

Reprints and permissions

About this article

Cite this article

De Clercq, E. Strategies in the design of antiviral drugs. Nat Rev Drug Discov 1, 13–25 (2002). https://doi.org/10.1038/nrd703

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrd703

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing