Skip to main content
Log in

New Antiherpesvirus Agents

Their Targets and Therapeutic Potential

  • Review Article
  • Published:
Drugs Aims and scope Submit manuscript

Summary

Of the large number of agents under development for the treatment of herpes virus infections [herpes simplex virus types 1 and 2 (HSV-1 and HSV-2), varicella zoster virus (VZV), cytomegalovirus (CMV), Epstein-Barr virus (EBV)], only ten have apparently reached clinical development.

Aciclovir was approved for the treatment of HSV infections over 10 years ago, and it remains an important and reliable antiviral agent. Recent approvals in some countries of valaciclovir for VZV infection and famciclovir for both HSV and VZV infections demonstrate the rapidity of change in this field.

Intravenous ganciclovir and foscarnet are approved for the treatment of CMV infection in the immunocompromised patient. Five of the antiherpetic drugs under current clinical development are nucleoside analogues or their prodrugs; another is a phosphorylated nucleoside (nucleotide). Four of the nucleoside agents — penciclovir, famciclovir, valaciclovir and lobucavir — are being developed for the management of HSV and VZV infections. Valaciclovir is also being developed for the prevention of CMV infections and famciclovir and lobucavir for the treatment of hepatitis virus infection. Oral ganciclovir, lobucavir, ISIS 2922 and cidofovir are being developed for the suppression of CMV infections in immunocompromised patients. Sorivudine has been studied in VZV infections.

n-Docosanol is under development for HSV infections, and cidofovir is being developed for both HSV and CMV infections, as well as for treatment of other viral diseases.

Traditionally, the adverse effects associated with anti-CMV compounds have been more difficult to manage and are acceptable clinically only because of the severity of the underlying infection and lack of safer therapeutic alternatives. In general, toxicity issues continue to be problematic in the anti-CMV arena, although newer agents have improved the situation to some extent. In contrast, the safety of anti-HSV compounds has traditionally been excellent, establishing a safety standard that must be met by newer agents entering the field.

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

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

References

  1. Schaeffer HJ, Beauchamp L, de Miranda P, et al. 9-(2-hydroxyethoxymethyl)guanine activity against viruses of the herpes group. Nature 1978; 272: 583–5

    Article  PubMed  CAS  Google Scholar 

  2. de Miranda P, Blum MR. Pharmacokinetics of acyclovir after intravenous and oral administration. J Antimicrob Chemother 1983; 12 Suppl.B: 29–37

    Article  PubMed  Google Scholar 

  3. Gnann Jr JW. New an ti virais with activity against varicella-zoster virus. Ann Neurol 1994; 34: 569–72

    Google Scholar 

  4. Faulds D, Heel RC. Ganciclovir: a review of its antiviral activity, pharmacokinetic properties and therapeutic efficacy in cytomegalovirus infections. Drugs 1990; 39: 597–638

    Article  PubMed  CAS  Google Scholar 

  5. Markham A, Faulds D. Ganciclovir: an update of its therapeutic use in cytomegalovirus infection. Drugs 1994; 48: 455–84

    Article  PubMed  CAS  Google Scholar 

  6. Chrisp P, Clissold SP. Foscarnet: a review of its antiviral activity, pharmacokinetic properties and therapeutic use in immunocompromised patients with cytomegalovirus retinitis. Drugs 1991; 41: 104–29

    Article  PubMed  CAS  Google Scholar 

  7. Vere Hodge RA, Cheng Y-C. The mode of action of penciclovir. Antiviral Chem Chemother 1993, 4 Suppl.1: 13–24

    Google Scholar 

  8. Pue MA, Benet LZ. Pharmacokinetics of famciclovir in man. Antiviral Chem Chemother 1993; 4 Suppl.1: 47–55

    CAS  Google Scholar 

  9. Vere Hodge RA. Famciclovir and penciclovir: the mode of action of famciclovir including its conversion to penciclovir. Antiviral Chem Chemother 1993; 4: 67–84

    CAS  Google Scholar 

  10. Harnden MR, Jarvest RJ, Boyd MR, et al. Prodrugs of selective antiherpesvirus agent 9-(4-hydroxyethoxy-3-hydroxy-methylbut-1-yl) guanine (BRL 39123). J Med Chem 1989; 32: 1738–43

    Article  PubMed  Google Scholar 

  11. Winton CF, Fowles SE, Vere Hodge RA, et al. Assay of famciclovir and its metabolites, including the antiherpes agent penciclovir, in plamsa and urine of rats dog and man. In: Reid E., Wilson ID, editors. Analysis of drugs and metabolites. Cambridge: Royal Soc Chemistry, 1990: 163–71

    Google Scholar 

  12. Vere Hodge RA. Famciclovir and penciclovir: the mode of action of famciclovir including its conversion to penciclovir. Antiviral Chem Chemother 1993: 4: 67–84

    CAS  Google Scholar 

  13. Boyd MR, Bacon TH, Sutton D, et al. Antiherpesvirus activity of 9-(4-hydroxy-3-hydroxymethylbut-1-yl)guanine (BRL 39123) in cell culture. Antimicrob Agents Chemother 1987; 31: 1238–42

    Article  PubMed  CAS  Google Scholar 

  14. Sacks SL. Genital herpes simplex virus infection and treatment. In: Sacks SL, Straus S, Whitley R, et al., editors. Clinical management of herpes viruses. Amsterdam: IOS Press, 1995

    Google Scholar 

  15. Boyd MR, Safrin S, Kern ER. Penciclovir: a review of its spectrum of activity, selectivity and cross-resistance pattern. Anti-vir Chem Chemother 1993; 4 Suppl.1: 3–11

    CAS  Google Scholar 

  16. Chatis PA, Crumpacker CS. Resistance of herpesviruses to antiviral drugs. Antimicrob Agents Chemother 1992; 36: 1589–95

    Article  PubMed  CAS  Google Scholar 

  17. Degreef H. Famciclovir, a new oral antiherpes drug: results of the first controlled clinical study demonstrating its efficacy and safety in the treatment of uncomplicated herpes zoster in immune competent patients. Int J Antimicrob Agents 1994: 4: 241–6

    Article  PubMed  CAS  Google Scholar 

  18. Tyring S, Nahlik J, Cummingham A, et al. The Collaborative Famciclovir Herpes Zoster Study Group: efficacy and safety of famciclovir in the treatment of patients with herpes zoster [abstract no. 1540]. Abstracts of the 33rd ICAAC; 1993: New Orleans (LA). Washington (DC): American Society for Microbiology, 1993

    Google Scholar 

  19. Tyring S, Barbarash RA, Nahlik JE, et al. Famciclovir for the treatment of acute herpes zoster: effects on acute disease and postherpetic neuralgia. A randomized, double-blind, placebo-controlled trial. Ann Intern Med 1995 Jul 15; 123: 89–96

    PubMed  CAS  Google Scholar 

  20. Sacks SL, Aoki FY, Diaz-Mitoma F, et al. Patient-initiated treatment of recurrent genital herpes with oral famciclovir: a Canadian multicentre, placebo-controlled, dose-ranging study [abstract no. H4]. Abstracts of 34th ICAAC; 1994 Oct 4–7: Orlando (FL). Washington (DC): American Society for Microbiology, 1994

    Google Scholar 

  21. Sacks SL, Martel A, Aoki F, et al. Early, clinic-initiated treatment of recurrent genital herpes using famciclovir: results of a Canadian, multicenter study. Baltimore: American Federation of Clinical Research, 1994

    Google Scholar 

  22. Mertz GJ, Loveless MO, Kraus SJ, et al. Famciclovir for suppression of recurrent genital herpes [abstract no. H3]. Abstracts of 34th ICAAC; 1994 Oct 4-7: Orlando (FL). Washington (DC): American Society for Microbiology, 1994

    Google Scholar 

  23. Daniels S, Schentag JJ. Drug interaction studies and safety of famciclovir in healthy volunteers. Antiviral Chem Chemother 1993; 4 Suppl.1: 57–64

    CAS  Google Scholar 

  24. Saltzman R, Jurewicz R, Boon R. Safety of famciclovir in patients with herpes zoster and genital herpes. Antimicrob Agents Chemother 1994; 38: 2454–7

    Article  PubMed  CAS  Google Scholar 

  25. Sacks SL, Varner T, Macintosh M. Double-blind, randomised, placebo-controlled, lesional, clinic-initiated treatment of culture positive recurrent genital herpes with intravenous penciclovir: clinical benefits of rapid-onset antiviral activity [abstract 135]. 3rd Congress of the European Academy of Dermatology and Venereology; 1993: Copenhagen

    Google Scholar 

  26. Lavender EA, Laroche JP, Gill CR, et al. Dermal irritancy potential of penciclovir cream compared with acyclovir 5% cream in health volunteers [abstract no. 130]. 3rd Congress of the European Academy of Dermatology and Venereology; 1993: Copenhagen

    Google Scholar 

  27. First approval for SB’s famciclovir. Scrip 1993; 1883: 17

  28. Famciclovir for herpes zoster. Med Lett Drugs Ther 1994; 36: 97-8

    Google Scholar 

  29. Beauchamp LM, Orr GF, de Miranda P, et al. Amino-acid ester prodrugs of acyclovir. Antivir Chem Chemother 1992; 3: 157–64

    CAS  Google Scholar 

  30. de Miranda P, Krasny HC, Page DA, et al. The dispositions of acyclovir in different species. J Pharmacol Exp Ther 1981; 19: 309–15

    Google Scholar 

  31. Burnette, TC, de Miranda P. Metabolic disposition of BW256U, the L-Valyl ester of acyclovir, in the rat [abstract]. Antiviral Res 1992; 17 Suppl.1: 188

    Google Scholar 

  32. Darby G. Acyclovir … and beyond. J Int Med Res 1994; 22 Suppl.1: 33A–42A

    PubMed  Google Scholar 

  33. de Miranda P, Burnette TC. Metabolism and pharmacokinetics of the acyclovir prodrug BW 256U 87 in cynomolgus monkeys [abstract]. Antiviral Res 1992; 17 Suppl.1: 53

    Google Scholar 

  34. Burnette TC, de Miranda P. Purification and characterization of an enzyme from rat liver that hydrolyzes 25 GU 87, the L-valyl ester prodrug of acyclovir [abstract]. Antiviral Res 1993; 20 Suppl.1: 119

    Google Scholar 

  35. Collins, P. The spectrum of antiviral activities of acyclovir in vitro and in vivo. J Antimicrob Chemother 1983; 12 Suppl.B: 19–27

    Article  PubMed  CAS  Google Scholar 

  36. Wagstaff AG, Faulds D, Goa KL. Acyclovir: a reappraisal of its antiviral activity, pharmacokinetic properties and therapeutic efficacy. Drugs 1994; 47: 153–205

    Article  PubMed  CAS  Google Scholar 

  37. Elion GB. Acyclovir: discovery, mechanism of action, and selectivity. J Med Virol 1993; Suppl. 1: 2–6

    Article  PubMed  Google Scholar 

  38. Nusinoff-Lehrman S, Smiley L, Szczech G. Update on acyclovir prodrugs. In: Mills J, Corey L, editors. Antiviral chemotherapy: new directions for application and clinical research. Englewood Cliffs (NJ): Prentice Hall, 1993: 97–104

    Google Scholar 

  39. Weiler S, Blum MR, Doucette M, et al. Pharmacokinetics of the acyclovir prodrug valaciclovir after escalating single and multiple-dose administration to normal volunteers. Clin Pharmacol Ther 1993; 54: 595–605

    Article  Google Scholar 

  40. Blum MR, Soul-Lawton J, Smith CM, et al. Increased bioavailability of acyclovir from oral valaciclovir in healthy volunteers [abstract]. Antiviral Res 1994; 23 Suppl.1: 74

    Google Scholar 

  41. Laskin OL. Clinical pharmacokinetics of acyclovir. Clin Pharmacokinet 1983; 8: 187–201

    Article  PubMed  CAS  Google Scholar 

  42. Weiler S, Blum MR, Smiley ML. Phase I pharmacokinetics of the acyclovir prodrug, valaciclovir. Antiviral Res 1993; 20 Suppl.1: 44

    Google Scholar 

  43. Beutner K, International Valaciclovir Zoster Study Group. The efficacy and safety of Valtrex™ (valaciclovir) for treatment of herpes zoster [abstract]. American Academy of Dermatology Annual Meeting; 1994 Jul 30–Aug 4: San Francisco (CA)

    Google Scholar 

  44. International Valaciclovir HSV Study Group, Smiley ML. Valaciclovir and acyclovir for the treatment of recurrent genital herpes simplex virus infection [abstract no. 1290]. Abstracts of the 33rd ICAAC; 1993: New Orleans (LA). Washington (DC): American Society for Microbiology, 1993

    Google Scholar 

  45. Balfour Jr HH, Chase A, Stapleton JT, et al. A randomized placebo-controlled trial of oral acyclovir for the prevention of cytomegalovirus disease in recipients of renal allograft. N Engl J Med 1989; 320: 1381–7

    Article  PubMed  Google Scholar 

  46. Meyers JD, Reed EC, Shepp DH, et al. Acyclovir for the prevention of cytomegalovirus infection and diseases in allogeneic marrow transplantation. N Engl J Med 1988; 318: 70–5

    Article  PubMed  CAS  Google Scholar 

  47. Valaciclovir CMV prophylaxis trial halted due to lower survival. Antiviral Agents Bull 1995; 8: 101–2

    Google Scholar 

  48. Collaborative DHPG Treatment Study Group. Treatment of serious cytomegalovirus infection with 9-(1,3-dihydroxy-2-propoxymethyl) guanine in patients with AIDS and other immunodeficiences. N Engl J Med 1986; 314: 801–5

    Article  Google Scholar 

  49. Balfour Jr HH. Management of cytomegalovirus disease with antiviral drugs. Rev Infect Dis 1990; 12 Suppl.7: S849–60

    Article  PubMed  CAS  Google Scholar 

  50. Little E, Stuart AD, Chee MS. Human cytomegalovirus UL97 open reading frame encodes a protein that phospharylates the antiviral analogue ganciclovir. Nature 1992; 358: 160–2

    Article  Google Scholar 

  51. Sullivan V, Talarico CL, Stanat SC, et al. A protein kinase homologue controls phosphorylation of ganciclovir in human cytomegalovirus-infected cells. Nature 1992; 358: 162–4

    Article  PubMed  CAS  Google Scholar 

  52. Boehme RE. Phosphorylation of the antiviral precursor 9-(1,3-dihydroxy-2-propokymethyl) guanine monophosphate by guanglate kinase isozymes. J Biol Chem 1984; 259: 12346–9

    PubMed  CAS  Google Scholar 

  53. Cole NL, Balfour Jr HH. In vitro susceptibility of cytomegalovirus isolates from immunocompromised patients to acyclovir and ganciclovir. Diag Microbiol Infect Dis 1987; 6: 255–61

    Article  CAS  Google Scholar 

  54. Biron KK, Stanat SC, Sonell JB, et al. Metabolic activation of the nucleoside analogue 9-[2-hydroxy-(hydroxymethyl) ethoxylmethyl guanine in hyuman diploid fibroblasts infected with human cytomegalovirus. Proc Nat Acad Sci USA 1985; 82: 2473–7

    Article  PubMed  CAS  Google Scholar 

  55. Plotkin SA, Drew WL, Felsenstein D, et al. Sensitivity of clinical isolates of human cytomegalovirus to 9-(1, 3-dihydroxy-2-propoxymethyl) guanine. J Infect Dis 1985; 152: 833–4

    Article  PubMed  CAS  Google Scholar 

  56. Spector SA, Busch DF, Follansbee S, et al. Pharmacokinetic, safety and antiviral profiles of oral ganciclovir in persons infected with human immunodeficiency virus: a phase I/II study. AIDS Clinical Trials Group and Cytomegalovirus Cooperative Study Group. J Infect Dis 1995; 171: 1431–7

    Article  PubMed  CAS  Google Scholar 

  57. Jacobson MA. Current management of cytomegalovirus disease in patients with AIDS. AIDS Res Human Retrov 1994; 10: 917–23

    Article  CAS  Google Scholar 

  58. Jabs DA, Newman C, de Bustros S, et al. Treatment of cytomegalovirus retinitis with ganciclovir. Ophthalmology 1987; 94: 824–30

    PubMed  CAS  Google Scholar 

  59. Erice A, Jordan MC, Chace BA, et al. Ganciclovir treatment of cytomegalovirus disease in transplant recipient and other immunocompromised host. JAMA 1987; 257: 3082–7

    Article  PubMed  CAS  Google Scholar 

  60. Laskin OL, Cederberg DM, Mills J, et al. Ganciclovir for the treatment and suppression of serious infections caused by cytomegalovirus. Am J Med 1987; 83: 201–7

    Article  PubMed  CAS  Google Scholar 

  61. Laskin OL, Stahl-Baylis CM, Kaiman CM, et al. Use oganciclovir to treat serious cytomegalovirus infections in patients with AIDS. J Infect Dis 1987; 155: 323–7

    Article  PubMed  CAS  Google Scholar 

  62. Buhles Jr WC, Mastre BJ, Tinker AJ, et al. Ganciclovir treatment of life-or sight-threatening cytomegalovirus infection: experience in 314 immunocompromised patients. Rev Infect Dis 1988; 10 Suppl.3: S495–506

    Article  PubMed  Google Scholar 

  63. Mills J, Jacobson MA, O’Donnell JJ, et al. Treatment of cytomegalovirus retinitis in patients with AIDS. Rev Infect Dis 1988; 10 Suppl.3: S522–31

    Article  PubMed  Google Scholar 

  64. Dieterich DT, Chachona A, LaFleur F, et al. Ganciclovir treatment of gastrointestinal infection caused by cytomegalovirus in patients with AIDS. Rev Infect Dis 1988; 10 Suppl.3: S532–7

    Article  PubMed  Google Scholar 

  65. Hecht DW, Snydman DR, Crumpacker CS, et al. The Boston Renal Transplant CMV Study Group: ganciclovir for the treatment of renal transplant-associated primary cytomegalovirus pneumonia. J Infect Dis 1988; 157: 187–90

    Article  PubMed  CAS  Google Scholar 

  66. Crumpacker C, Marlowe S, Zhang JL, et al. Treatment of cyto-megalovirus pneumonia. Rev Infect Dis 1988; 10 Suppl.3: S538–46

    Article  PubMed  Google Scholar 

  67. Winkler I, Winkelman E, Scholl T, et al. Antiviral activity and pharmacokinetics of HOE 602, an acyclic nucleoside, in animal models. Antiviral Res 1990; 14: 61–74

    Article  PubMed  CAS  Google Scholar 

  68. Jacobson MA, de Miranda P, Cederberg DM, et al. Human pharmacokinetics and tolerance of oral ganciclovir. Antimicrob Agents Chemother 1987; 31: 1251–4

    Article  PubMed  CAS  Google Scholar 

  69. Drew WL, Ives D, Lalezari JP, et al. Oral ganciclovir as maintenance treatment for cytomegalovirus retinitis in patients with AIDS. Suntex Corporation Oral Ganciclovir Study Group. N Engl J Med 1995; 333: 615–20

    Article  PubMed  CAS  Google Scholar 

  70. Oral Ganciclovir European and Australian Cooperative Study Group. Intravenous versus oral ganciclovir: European/Australian comparative study of efficacy and safety in the prevention of cytomegalovirus retinitis recurrrence in patients with AIDS. AIDS 1995; 9: 471–7

    Google Scholar 

  71. Squires KE, Stempien MJ, Shadman A, et al. Oral ganciclovir (POG) vs. IV ganciclovir (IVG) maintenance therapy for CMV retinitis in patients with AIDS: preliminary results of a phase III study [abstract no. 540]. 1st National Conference on Human Retroviruses and Related Infections: 1993: Washington (DC)

    Google Scholar 

  72. Oral ganciclovir approved. AIDS Treat News 1995 Jan 6; 214: 1

  73. Machida H. In vitro anti-herpesvirus action of a novel antiviral agent, brovavir (BV-araU). Chemotherapy 1990; 38: 256–61

    CAS  Google Scholar 

  74. Machida H, Sakata S, Kuninaka A, et al. Antiherpes viral and anti-cellular effects of 2-β -D-arabinofuranosyl-E-5-(2-halogenovinyl) uracils. Antimicrob Agents Chemother 1981; 20: 47–52

    Article  PubMed  CAS  Google Scholar 

  75. Machida H. Brovavir: its antiherpesviral activity and mode of action. In: Lopez C, editor. Immunobiology and prophylaxis of human herpesvivus infections. New York: Plenum Press, 1990: 255–65

    Chapter  Google Scholar 

  76. Suzutani T, Machida H, Sakuma T, et al. Effects of various nucleosides on antiviral activity and metabolism of 1-β -D-arabinofuranosyl-1-5(2-bromovinyl) uracil against herpes simplex virus types 1 and 2. Antimicrob Agents Chemother 1988; 32: 1547–51

    Article  PubMed  CAS  Google Scholar 

  77. Ayisi NK, Wall RA, Wanklin RJ, et al. Comparative metabolism of £’-5-(2-bromovinyl)uracil in herpes simplex virus-infected cells. Mol Pharmacol 1987; 31: 422–9

    PubMed  CAS  Google Scholar 

  78. Machida H. Comparison of susceptibilities of varicella-zoster virus and herpes simplex virus to nucleoside analogs. Antimicrob Agents Chemother 1986; 29: 524–6

    Article  PubMed  CAS  Google Scholar 

  79. Lin JC, Machida H. Comparison of two bromovinyl nucleoside analogs, 1 -β -D-arabinofuranosyl.E-5-(2-bromovinyl)-uracil and E-5-(2-bromovinyl)-2-deoxyuridine, with acyclovir in inhibition of Epstein-Barr virus replication. Antimicrob Agents Chemother 1988: 32: 1068–72

    Article  PubMed  CAS  Google Scholar 

  80. Machida H, Nishitans M. Drug susceptibilities of isolates of varicella zoster virus in clinical study of oral brovavir. Microbiol Immunol 1990; 34: 401–11

    Google Scholar 

  81. Machida H, Sakata S. In vitro and in vivo antiviral activity of L-β -D arabinofuranosyl-E-5-(2-bromovinyl) uracil (BV-araU) and related compounds. Antiviral Res 1984; 4: 135–41

    Article  PubMed  CAS  Google Scholar 

  82. Cheng YC, Dutschman G, Fox JJ, et al. Differential activity of potential antiviral nucleoside analogs on herpes simplex virus-induced and human cellular thyimdine kinases. Anti-microb Agents Chemother 1981; 20: 420–3

    Article  CAS  Google Scholar 

  83. Maudgal PC, de Clerq E. Evaluation of bromovinyldeoxyurid-ine-related compounds in the treatment of experimental herpes simplex keratis. Arch Ophthalmol 1985; 103: 1393–7

    Article  PubMed  CAS  Google Scholar 

  84. Topke H, Graf M, Wutzier P, et al. Evaluation of (E)-5-(2-bromovinyl)-and 5-vinyl-1 -β -D-arabinofuranosyluracil (BrVaraU, VaraU) in the treatment of experimental herpes simplex virus type 1 keratitis in rabbits: comparison with (E)-5-(2-bromovinyl) 2’-deoxyuridine (BrVUdR). Antiviral Res 1988; 9: 273–80

    Article  PubMed  CAS  Google Scholar 

  85. Soike KF, Baskin G, Cantrell C et al. Investigation of antiviral activity of 1 -β -D-arabinofuranosylthymine (ara-T) and 1-β -D-arabinofuranosyl-E-5-(2-bromovyl) uracil (sorivudine) in monkeys infected with simian varicella virus. Antiviral Res 1984; 4: 245–57

    Article  PubMed  CAS  Google Scholar 

  86. Sherman J, DeVautt A, Natarajan M, et al. SQ32756 (BV-araU): characteristics and pharmacokinetics in healthy young and elderly male volunteers [abstract no. 1103]. 30th ICAAC; 1990: Washington (DC). Washington (DC): American Society for Microbiology, 1990

    Google Scholar 

  87. Ogiwara T, Mikami H, Nakamaru M, et al. Phase I clinical study of YN-72 (BV-araU, Brovavir). Jpn Pharmacol Ther 1990; 18: 507–23

    CAS  Google Scholar 

  88. Niimura M. A double-blind clinical study in patients with herpes zoster to establish YN-72 (brovavir) dose. Adv Exp Med Biol 1990; 278: 267–75

    Article  PubMed  CAS  Google Scholar 

  89. Niimura M, Takahashi M, Nishikawa T, et al. Multicentre double-blind study of YN-72 (BV-araU, brovavir) in patients with herpes zoster. Jpn J Clin Dermatol 1990; 44: 447–58

    Google Scholar 

  90. Hiraoka A, Masaoka T, Nagai K, et al. Clinical effect of BV-araU on varicella-zoster virus infection in immunocompromised patients with haematological malignancies. J Antimicrob Chemother 1991; 27: 361–7

    Article  PubMed  CAS  Google Scholar 

  91. Wallace MR, Bowler WA, Oldfield EC. Treatment of varicella in the immunocompetent adult. J Med Virol 1993; Suppl. 1: 90–2

    Article  PubMed  Google Scholar 

  92. Dehertogh D, Boag F, Bodsworth N, et al. The safety and efficacy of sorivudine (BV-araU) for the treatment of zoster in HIV-infected adults [abstract no. A/7]. Abstracts of the 34th ICAAC; 1994 Oct 4–7: Orlando (FL). Washington (DC): American Society for Microbiology, 1994

    Google Scholar 

  93. Desgranges Razaka G, De Clerq E, et al. Effect of (E)-5-(2-bromo-vinyl) uracil on the catabolism and antitumor activity of 5-fluorouvacil in rats and leukemic mice. Cancer Res 1986; 46: 1094–101

    Google Scholar 

  94. de Clerq E. Antiviral activity spectrum and target of action of different classes of nucleoside analogues. Nucleosides Nucleotides 1994; 13: 1271–95

    Article  Google Scholar 

  95. de Clerq E. Antivirais for the treatment of herpesvirus infections. J Antimicrob Chemother 1993; 32 Suppl.A: 121–32

    Article  Google Scholar 

  96. Suzutani T, Machida H. Analysis of toxic and mutagenic activities of antiherpesvirus nucleosides against HeLa cells and herpes simplex virus type 1. Mutation Res 1992; 267: 125–31

    Article  PubMed  CAS  Google Scholar 

  97. Wutzler P, de Clercq E, Wutke K, et al. Oral brivudin versus intravenous acyclovir in the treatment of herpes zoster in immunocompromised patients: a randomized double-blind trial [abstract no. 144]. Antiviral Res 1995; 26: A302

    Article  Google Scholar 

  98. Braitman A, Swerdel MR, Olsen SJ, et al. Evaluation of SQ 34,514: pharmacokinetics and efficacy in experimental herpesvirus infections in mice. Antimicrob Agents Chemother 1991; 35: 1464–8

    Article  PubMed  CAS  Google Scholar 

  99. Terry BJ, Cianci CW, Hagen ME. Inhibition of herpes simplex virus type 1 DNA polymerase by [1R(1α,2β, 3α)]-9-[2,3-bis(hydroxymethyl)cyclobutyl]guanine. Mol Pharmacol 1991; 40: 591–6

    PubMed  CAS  Google Scholar 

  100. Bisacchi GS, Braitman A, Cianci CW, et al. Synthesis and antiviral activity of enantiomeric forms of cyclobutyl nucleoside analogues. J Med Chem 1991; 34: 1415–21

    Article  PubMed  CAS  Google Scholar 

  101. Petty BG, Wachsman M, Jordan MC, et al. Sequential ascending multiple-dose safety and pharmacokinetic study of oral lobucavir (BMS-180194) in asymptomatic volunteers seropositive for HIV and CMV [abstract no. 133]. Antiviral Res 1995; 26: A296

    Google Scholar 

  102. DeClerq E, Sakuma T, Baha M, et al. Antiviral activity of phosphonyl metholyalkyl derivatives of purine and pyrimidines. Antiviral Res 1987; 8: 261–72

    Article  Google Scholar 

  103. DeClerq E, Holy A, Rosenberg I, et al. A novel selective broad-spectrum anti-DNA virus agent. Nature 1986; 323: 464–7

    Article  Google Scholar 

  104. Snoeck R, Sakuma T, De Clerq E, et al. (S)-1-(3-hydroxy-2 phosphonyl methoxypropyl) cytosine, a potent and selective inhibitor of human cytomegalovirus replication. Antimicrob Agents Chemother 1988; 32: 1839–44

    Article  PubMed  CAS  Google Scholar 

  105. Snoeck R, DeClerq E. (S)-1-(3-hydroxy-2-phosphopyl methoxypropyl) cytosine (HPMPC), a potent and selective inhibitor of cytomegalovirus infection in vitro and in vivo. In: Touraine JL, Traeger J, Betueb H, et al., editors. Transplantation and clinical immunology XXIII. Amsterdam: Elsevier, 1991: 169–76

    Google Scholar 

  106. Cin JC, DeClerq E, Pagano JS. Inhibitory effects of acyclic nucleoside phosphonate analogs, including (S)-1-(3-hydroxy-2-phosphonyl methoky propyl) cytosine, on Epstein-Barr virus replacation. Antimicrob Agents Chemother 1991; 35: 2440–3

    Article  Google Scholar 

  107. Bronson JJ, Ghazzouli I, Hitchcock MJM, et al. Synthesis and antiviral activity of the nucleotide analogue (S)-1-[3-hydroxy-2-(phosphonylmethoxy) propyl)-cytosine. J Med Chem 1989; 32: 1457–63

    Article  PubMed  CAS  Google Scholar 

  108. Biron KK, Stanet SC, Sorrell JBC, et al. Metabolic activation of the nucleoside analog 9-[2-hydroxy-1-(hydroxymethyl) ethoxy] methylguanine in human diploid fibroblasts infected with human cytomegalovirus. Proc Nat Acad Sci USA 1985; 82: 2473–7

    Article  PubMed  CAS  Google Scholar 

  109. Ho HT, Woods KL, Bronson JJ, et al. Intracellular metabolism of the antiherpes agent (S)-1-[3-hydroxy-2-(phosphonyl-methoxy) propyl] cytosine. Mol Pharm 1991; 41: 197–202

    Google Scholar 

  110. Talarico C, Stanet S, Lambe C, et al. Mode of action studies on the anti-cytomegalovirus nucleoside analog [1-(2-hydroxy-1 -hydroxymethyl) ethoxy-methyl) cytosine] [abstract no. 92]. Antiviral Res 1990; S1: 87

    Google Scholar 

  111. de Clerq E, Holy A. Efficacy of (S)-2-(3-hydroxy-2-phos-phonyl methoxy propyl) cytosine in various models of herpes simplex virus infection in mice. Antimicrob Agents Chemother 1991; 35: 701–6

    Article  Google Scholar 

  112. Maudgal PC, de Clerq E. Effects of phosphonylmethoxy alkylpurine and pyrmidin and derivatives on TK and TK-HSV-1 keratitis in rabbits. Antiviral Res 1991; 16: 93–100

    Article  PubMed  CAS  Google Scholar 

  113. Neyts J, Balzarini J, de Clerq E. Efficacy of (S)-1-(3-hydroxy-2-phosphonyl-methoxypropyl) cytosine [HPMPC] against intraperitoneal and intracerebral murine cytomegalovirus infections [abstract no. 167]. Antiviral Res 1991; Suppl. 1: 133

    Article  Google Scholar 

  114. Neyts J, Balzarin J, Naesens L, et al. Efficacy of (S)-1-(3-hydroxy-2-phosphonyl-methoxypropyl) cyfosine and 9-(1,3-dihydroxy-2-propoxy methyl) guanin for the treatment of murine cytomegalovirus infection in severe combined immunodeficiency mice. J Med Virol 1992; 37: 67–71

    Article  PubMed  CAS  Google Scholar 

  115. Soike KF, Huang JL, Zhang JY, et al. Evaluation of infrequent dosing regimens with (S)-1-[3-hydroxy-2-(phosphonyl-me-thoxy)-propyl] cytosine (S-HPMPC) on simian varicella infection in monkeys. Antiviral Res 1991; 16: 17–28

    Article  PubMed  CAS  Google Scholar 

  116. Stals FS, de Clerq E, Bruggeman CA. Comparative activity of (S)-1-(3-hydroxy-2-phosphonyl methoxy propyl) cytosine and 9-(1,3-dihydroxy-2-propoxymethyl) guanine against rat cytomegalovirus infection in vitro and in vivo. Antimicrob Agents Chemother 1991; 35: 2262–6

    Article  PubMed  CAS  Google Scholar 

  117. Lalezari JP, Drew WL, Glutzer E, et al. (S)-1-[3-hydroxy-2- (phosphonylmethoxy)propyl]cytosine (cidofovir): results of a phase I/II study of a novel antiviral nucleotide analogue. J Infect Dis 1995; 171: 788–96

    Article  PubMed  CAS  Google Scholar 

  118. Lalezari J, Stagg R, Kupperman B, et al. A phase II/III randomized study of immediate versus deferred intravenous (IV) cidofovir (CDV, HPMPC) for AIDS patients with peripheral CMV retinitis (CMV-R). International Conference on Ocular Infections; 1995: Jerusalem

    Google Scholar 

  119. Palmer J, Vogt PE, Kern ER. Prevention and treatment of experimental genital herpes simplex virus type 2 (HSV-2) infections with topical HPMPC [abstract no. 205]. Antiviral Res 1995; 26: A334

    Google Scholar 

  120. Marcelletti JF, Pope LE, Khalil MH, et al. Lidakol™. Drugs Future 1992; 10: 879–2

    Google Scholar 

  121. Katz DH, Marcelletti JF, Khalil MH, et al. Antiviral activity of 1-docosanol, an inhibitor of lipid-enveloped viruses including herpes simplex. Proc Natl Acad Sci USA 1991; 88: 10825–9

    Article  PubMed  CAS  Google Scholar 

  122. Katz DH, Marcelletti JF, Pope LE, et al. n-Docosanol: broad spectrum antiviral activity against lipid-enveloped viruses. In: Slow infections of the central nervous system. Ann NY Acad Sci 1994; 724: 472–88

    Article  PubMed  CAS  Google Scholar 

  123. Marcelletti JF, McFadden RR, Khalil MH, et al. Comparison of n-Docosanol cream (Lidakol™) with acyclovir ointment (Zovirax™) in the inhibition of cutaneous herpes simplex virus (HSV) infections in two guinea pig model systems [abstract]. Antiviral Res 1995; 26: A333

    Google Scholar 

  124. McKeough MB, Spruance SL. Lidakol™ cream vs Zovirax™ ointment for the treatment of experimental dorsal cutaneous herpes simplex virus type 1 (HSV-1) infection in the guinea pig. Antiviral Res 1995; 26: 332

    Google Scholar 

  125. Pope LE, Khalil MH, Marcelletti JF, et al. Skin penetration and systemic absorption of the antiviral agent n-docosanol from topically applied cream formulations [abstract]. J Invest Med 1995; 43: 222A

    Google Scholar 

  126. Katz DH, Habbema L, de Boulle, et al. n-Docosanol cream is an effective topical treatment of recurrent herpes labialis [abstract]. Antiviral Res 1994; 23 Suppl.1: 75

    Google Scholar 

  127. Miller CJ, Gersten MJ, Davis RC, et al. n-Docosanol prevents vaginal transmission of SIVmac251 in rhesus macaques [abstract]. Antiviral Res 1995; 26: A277

    Article  Google Scholar 

  128. Azad RF, Driver VB, Tonaka K, et al. Antiviral activity of a phosphorothioate oligonucleotide complementary to RNA of the human cytomegalovirus major immediate-early region. Antimicrob Agents Chemother 1993; 37: 1945–54

    Article  PubMed  CAS  Google Scholar 

  129. Anderson KP, Azad RF, Driver VB, et al. Preclinical evaluation of a phosphorothioate oligonucleotide with potent antiviral activity against human cytomegalovirus [abstract no. 10]. Antiviral Res 1994; 23: A40

    Article  Google Scholar 

  130. Anderson KP, Azad RF, Brown-Driver V, et al. Preclinical and clinical antiviral activity of ISIS 2922, an antisense oligonucleotide complementary to cytomegalovirus immediate early RNA [abstract]. First IAC of Japan; 1994 Dec: Kyoto.

    Google Scholar 

  131. Palestine AG, Cantrill H, Ai E, et al. Treatment of cytomegalo-virus (CMV) retinitis with ISIS 2922 [abstract]. Fort Lauderdale (FL): Association for Research in Vision and Ophthalmology, 1995 May

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Rights and permissions

Reprints and permissions

About this article

Cite this article

Alrabiah, F.A., Sacks, S.L. New Antiherpesvirus Agents. Drugs 52, 17–32 (1996). https://doi.org/10.2165/00003495-199652010-00002

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/00003495-199652010-00002

Keywords

Navigation