Skip to main content
Log in

Valaciclovir

A Review of its Antiviral Activity, Pharmacokinetic Properties and Therapeutic Efficacy in Herpesvirus Infections

Drugs Aims and scope Submit manuscript

Abstract

Synopsis

Valaciclovir, the L-valyl ester of aciclovir (acyclovir), is an oral prodrug that undergoes rapid and extensive first-pass metabolism to yield aciclovir and the essential amino acid L-valine. Aciclovir, the active antiviral component of valaciclovir, shows good in vitro activity against the herpesviruses herpes simplex virus (HSV)-1, HSV-2 and varicella zoster virus.

The bio availability of aciclovir from oral valaciclovir is considerably greater than that achieved after oral aciclovir administration. Thus, valaciclovir delivers therapeutic aciclovir concentrations when administered in a less frequent oral dosage regimen than is required for aciclovir.

Valaciclovir is an effective treatment for herpes zoster in immunocompetent adults. In a large comparative study that included patients ≥50 years of age, valaciclovir (1000mg 3 times daily for 7 or 14 days) and oral aciclovir (800mg 5 times daily) were equally effective in achieving resolution of cutaneous zoster lesions. Importantly, valaciclovir was significantly more effective than aciclovir in reducing the duration of zoster-associated pain.

Preliminary results of several studies indicate that valaciclovir (500 to 1000mg twice daily for 5 to 10 days) is as effective as aciclovir (200mg 5 times a day for 5 to 10 days) in the treatment of genital herpes. In patients with first or recurrent episodes of genital herpes, valaciclovir reduced the duration of viral shedding, hastened lesion healing and decreased lesion-associated pain. Valaciclovir was also effective in suppressing recurrent episodes of genital herpes and significantly prolonged the time to a recurrent episode of infection compared with placebo.

Valaciclovir is a well tolerated drug; in herpes zoster and HSV studies its tolerability profile was similar to that of aciclovir or placebo.

Valaciclovir represents an advance in antiherpes drug therapy and is a useful treatment option for patients with herpes zoster or genital herpes. It is at least as effective as aciclovir and is administered in a more convenient oral dosage regimen. Thus, valaciclovir may ultimately succeed aciclovir as a first-line treatment for genital herpes or herpes zoster.

Antiviral Activity

After oral administration and absorption, valaciclovir is converted to aciclovir (acyclovir), which is the active antiviral component of valaciclovir. Thus, the antiviral activity of valaciclovir is the same as that of aciclovir.

Aciclovir is preferentially converted to its active antiviral form, aciclovir tri-phosphate, by herpesvirus-encoded enzymes (to the monophosphate) and cellular enzymes (from the monophosphate to the triphosphate) in herpesvirus-infected cells. Aciclovir triphosphate subsequently inhibits herpesvirus DNA polymerase, terminates viral DNA chain elongation and therefore completely prevents replication of viral DNA.

Aciclovir has good inhibitory activity against HSV (herpes simplex virus) and VZV (varicella zoster virus) in vitro. The descending order of susceptibility to the drug is HSV-1, HSV-2 and VZV Aciclovir is also active against Epstein-Barr virus and has moderate activity against cytomegalovirus and human herpesvirus 6. The antiviral activity of aciclovir has also been demonstrated in animal models of ocular, cutaneous and genital HSV infection.

Resistance of HSV or VZV to aciclovir is rare and almost all resistant isolates occur as a result of a deficiency in thymidine kinase. Nearly all clinical HSV or VZV isolates with resistance to aciclovir have been obtained from immuno-compromised patients who have received prolonged aciclovir therapy.

Pharmacokinetic Properties

In healthy volunteers and patients, valaciclovir is readily absorbed after oral administration and undergoes rapid and extensive first-pass metabolism to yield aciclovir and the essential amino acid L-valine.

The bioavailability of aciclovir following valaciclovir administration is greater than after oral aciclovir administration (54 vs 12 to 20%). The pharmacokinetics of aciclovir after valaciclovir were similar after single- or multiple-dose administration of valaciclovir in healthy adult volunteers. After single 100 to 1000mg doses of valaciclovir, the area under the plasma concentration-time curve (AUC) for aciclovir ranged from 2.28 to 19.5 mg/L • h. Following oral valaciclovir 2000mg 4 times daily, the daily AUC for aciclovir was 109 mg/L • h, which was similar to the daily systemic aciclovir exposure achieved with intravenous aciclovir 10 mg/kg 3 times daily (107 mg/L • h).

The mean plasma elimination half-life (t½) of aciclovir after administration of single oral valaciclovir doses (100 to 1000mg) or oral valaciclovir 1000 to 8000 mg/day for 8 to 11 days to healthy volunteers was 2.62 to 3.13 hours. In patients with end-stage renal disease the t½ of aciclovir after a single 1000mg dose of valaciclovir was prolonged to 14 hours. However, one third of aciclovir may be removed by dialysis in these patients.

Metabolites of aciclovir (from valaciclovir) are well characterised and pharmacologically inactive. The kidneys are the main route of elimination of valaciclovir, aciclovir and other metabolites of the drug. Valaciclovir and aciclovir are also excreted in the faeces. After single or multiple doses (100 to 2000mg) of valaciclovir, aciclovir accounted for 80 to 85% of the recovered drug, 7 to 12% was recovered as metabolites and <1% as valaciclovir.

No clinically significant interactions were observed between valaciclovir and a thiazide diuretic, an aluminium- and magnesium-containing antacid, digoxin, cimetidine, probenecid, or a combination of cimetidine and probenecid.

Therapeutic Efficacy

The therapeutic efficacy of valaciclovir has been investigated in immunocompetent patients with herpes zoster (shingles) or genital HSV. Studies in immuno-compromised patients are in progress. Treatment was initiated within 72 hours of initial signs or symptoms of infection in patients with herpes zoster or first-episode genital herpes. In recurrent genital herpes studies, treatment was patient-initiated and commenced within 24 hours of the appearance of initial symptoms of a recurrence.

Valaciclovir 1000mg 3 times daily, administered for either 7 or 14 days, was as effective as 7 days’ treatment with aciclovir (800mg 5 times a day) in achieving resolution of the zoster rash in a large study that included 1141 patients (aged ≥50 years) with herpes zoster (including patients with ophthalmic zoster). In addition, both regimens of valaciclovir significantly hastened the resolution of zoster-associated pain by, on average, 22 or 34%, compared with aciclovir. Valaciclovir 1000mg 3 times a day for 7 days was also an effective treatment for younger patients (aged 18 to 50 years) with herpes zoster and was significantly more effective than placebo in terminating the formation of new zoster lesions. However, the relative effects of valaciclovir and placebo on other efficacy parameters, including the duration of zoster-associated pain, have not yet been reported.

Accumulating data, some of which are at present available in abstract form only, indicate that valaciclovir is also an effective treatment for patients with first or recurrent episodes of genital HSV. In a single study that included patients with a first episode of genital herpes, there were no differences in efficacy between valaciclovir 1000mg twice daily and aciclovir 200mg 5 times a day as determined by times to lesion healing, resolution of viral shedding and pain resolution. Valaciclovir 500 or 1000mg twice a day and aciclovir 200mg 5 times a day also demonstrated similar efficacy in patients with recurrent episodes of genital herpes and both agents significantly hastened the resolution of episodes (median times to lesion healing 4.0 to 4.8 days) compared with placebo (5.9 days). Recipients of valaciclovir 500 or 1000mg twice daily experienced a higher incidence of aborted recurrent episodes of genital herpes than aciclovir recipients. Valaciclovir (500mg once daily) has also been shown to suppress recurrent episodes of genital herpes. In patients who had experienced ≥8 recurrences of infection during the previous year, valaciclovir was significantly more effective than placebo in prolonging the time between recurrent episodes.

Tolerability

Valaciclovir is, like oral aciclovir, a well tolerated drug. In patients with herpes zoster, the most common adverse events reported during treatment with valaciclovir 1000mg 3 times daily were nausea, vomiting, headache and diarrhoea; the incidence of these events was similar to that in patients with herpes zoster who received oral aciclovir 800mg 5 times a day. Valaciclovir has also shown good tolerability in patients with genital herpes, individuals with hepatic and renal impairment and elderly volunteers. In patients with recurrent genital herpes, the tolerability profile of valaciclovir (500 to 1000mg twice daily) was similar to that of placebo.

Dosage and Administration

For adults with herpes zoster, the recommended dosage of valaciclovir is 1000mg 3 times daily for 7 days. Treatment with the drug should be initiated at the first signs and symptoms of herpes zoster, preferably within 72 hours of the appearance of the zoster rash.

The recommended dosage of valaciclovir for patients with either a first episode or recurrent episodes of genital herpes is 500mg twice daily for 5 days, which may be extended to 10 days if necessary in first episodes of genital herpes, as these may be more severe.

Dosage modification of valaciclovir is not usually required in elderly patients, but the dosage of the drug should be reduced in patients with moderate to severe renal impairment.

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.

Institutional subscriptions

Similar content being viewed by others

References

  1. Crooks RJ, Murray A. Valaciclovir — a review of a promising new antiherpes agent. Antiviral Chem Chemother 19945 Suppl. 1: 31–7

    Google Scholar 

  2. Wagstaff AJ, Faulds D, Goa KL. Aciclovir: a reappraisal of its antiviral activity, pharmacokinetic properties and therapeutic efficacy. Drugs 1994; 47(1): 153–205

    Article  PubMed  CAS  Google Scholar 

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

    PubMed  Google Scholar 

  4. Youle MS, Gazzard BG, Johnson MA. Effects of high-dose acyclovir on herpesvirus disease and survival in patients with advanced HIV disease: a double-blind, placebo-controlled study. AIDS 1994 May; 8: 641–9

    Article  PubMed  CAS  Google Scholar 

  5. Prentice HG, Gluckman E, Powles RL, et al. Impact of long-term acyclovir on cytomegalovirus infection and survival after allogeneic bone marrow transplantation. Lancet 1994; 343: 749–53

    Article  PubMed  CAS  Google Scholar 

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

    Article  PubMed  Google Scholar 

  7. Balfour Jr HH, Kelly JM, Suarez CS, et al. Acyclovir treatment of varicella in otherwise healthy children. J Pediatr 1990; 116: 633–9

    Article  PubMed  Google Scholar 

  8. Balfour Jr HH, Rotbart HA, Feldman S, et al. Acyclovir treatment of varicella in otherwise healthy adolescents. J Pediatr 1992; 120: 627–33

    Article  PubMed  Google Scholar 

  9. Dunkle LM, Arvin AM, Whitley RJ, et al. A controlled trial of acyclovir for chickenpox in normal children. N Engl J Med 1991; 325: 1539–44

    Article  PubMed  CAS  Google Scholar 

  10. Balfour Jr HH. Clinical aspects of chickenpox and herpes zoster. J Int Med Res 1994; 22 Suppl. 1: 3A–13A

    PubMed  Google Scholar 

  11. O’Brien JJ, Campoli-Richards DM. Acyclovir: an updated review of its antiviral activity, pharmacokinetic properties and therapeutic efficacy. Drugs 1989; 37: 233–309

    Article  PubMed  Google Scholar 

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

    Google Scholar 

  13. Bacon TH, Schinazi RF. An overview of the further evaluation of penciclovir against herpes simplex virus and varicella zoster virus in cell culture highlighting contrasts with acyclovir. Antiviral Chem Chemother 1993; 4 Suppl. 1: 25–36

    CAS  Google Scholar 

  14. Glaxo Wellcome. Valaciclovir Prescribing Information. US, 1995

  15. Littler E. Safety and efficacy issues of herpesvirus drugs. Antiviral Chem Chemother 1994; 5 Suppl. 1: 11–6

    CAS  Google Scholar 

  16. Perry CM, Wagstaff AJ. Famciclovir: a review of its pharmacological properties and therapeutic efficacy in herpesvirus infections. Drugs 1995; 50(2): 396–415

    Article  PubMed  CAS  Google Scholar 

  17. Field HJ, Tewari D, Sutton D, et al. Comparison of efficacies of famciclovir and valaciclovir against herpes simplex virus type 1 in a murine immunosuppression model. Antimicrob Agents Chemother 1995 May; 39: 1114–9

    Article  PubMed  CAS  Google Scholar 

  18. Larder BA, Darby G. Virus drug-resistance: Mechanisms and consequences. Antiviral Res 1984; 4: 1–42

    Article  PubMed  CAS  Google Scholar 

  19. Darby G, Field HJ, Salisbury SA. Altered substrate specificity of herpes simplex virus thymidine kinase confers acyclovirresistance. Nature 1981; 289: 81–3

    Article  PubMed  CAS  Google Scholar 

  20. Snoeck R, Andrei G, De Clercq E. Chemotherapy of varicella zoster virus infections. Int J Antimicrob Agents 1994 Aug; 4: 211–26

    Article  PubMed  CAS  Google Scholar 

  21. Collins P, Nixon Ellis M. Sensitivity monitoring of clinical isolates of herpes simplex virus to acyclovir. J Med Virol 1993 Suppl. 1: 58–66

    Google Scholar 

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

    CAS  Google Scholar 

  23. Burnette TC, Harrington JA, Reardon JE, et al. Purification and characterization of a rat liver enzyme that hydrolyzes valaciclovir, the L-valyl ester prodrug of acyclovir. J Biol Chem 1995; 270(26): 15827–31

    Article  PubMed  CAS  Google Scholar 

  24. Soul-Lawton J, Seaber E, On N, et al. Absolute bioavailability and metabolic disposition of valaciclovir, the L-valyl ester of acyclovir, following oral administration to humans. Antimicrob Agents Chemother 1995 Dec; 39: 2759–64

    Article  PubMed  CAS  Google Scholar 

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

    Article  Google Scholar 

  26. Wang LH, Schultz M, Weiler S. Pharmacokinetics and safety of multiple-dose valaciclovir in geriatric volunteers with and without concomitant diuretic therapy. Antimicrob Agents Chemother 1996 Jan; 40: 80–5

    PubMed  CAS  Google Scholar 

  27. First MR, Blum MR, Brennan P, et al. Pharmacokinetics (PK) of 256U87, an acyclovir (ACV) prodrug in renal transplant (RTX) patients [abstract]. Clin Pharmacol Ther 1993 Feb; 53: 238

    Google Scholar 

  28. On NT, Ehninger G, Gluckman E, et al. Acyclovir pharmacokinetics following oral multiple-dose of valaciclovir (Valtrex™) in allogeneic BMT patients [abstract]. Bone Marrow Transplant 1995 Mar; 15 Suppl. 2: S122

    Google Scholar 

  29. Jacobson MA, Gallant J, Wang LH, et al. Phase I trial of valaciclovir, the L-valyl ester of acyclovir, in patients with advanced human immunodeficiency virus disease. Antimicrob Agents Chemother 1994 Jul; 38: 1534–40

    Article  PubMed  CAS  Google Scholar 

  30. Coakley D, Weiler S, Blum MR, et al. Valtrex(Rm) (valaciclovir) administration achieves increased acyclovir concentrations in multiple populations [abstract]. Pharmacotherapy 1994 May–Jun; 14: 373

    Google Scholar 

  31. Posner J, Soul-Lawton J, Weiler S, et al. Valtrex (Rm) (valaciclovir) — bioavailability in différent patient populations [abstract]. Can J Infect Dis 1995 Jul; 6 Suppl. C: 287C

    Google Scholar 

  32. 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 

  33. Obenshain MK, Warwick JC, Weiler S, et al. The absence of a food effect on acyclovir (ACV) bioavailability from a single Valtrex™ (VACV) dose. Pharm Res 1994; 11 Suppl. 10: S435

    Google Scholar 

  34. Blum MR, Liao SHT, de Miranda P. Overview of aciclovir disposition in adults and children. Am J Med 1982; 73 Suppl.: 186–92

    Article  PubMed  CAS  Google Scholar 

  35. Carrington D. Prospects for improved efficacy with antiviral prodrugs: will valaciclovir and famciclovir meet the clinical challenge? Int Antiviral News 1994 Apr; 2: 50–3

    Google Scholar 

  36. Rolan P. Pharmacokinetics of new antiherpetic agents. Clin Pharmacokinet 1995 Nov; 29: 333–40

    Article  PubMed  CAS  Google Scholar 

  37. de Miranda P, Burnette TC, Smith C, et al. Mechanisms of the enhanced oral bioavailability of acyclovir with the prodrug valacyclovir HC1 (Valtrex™) [abstract]. 34th International Conference on Antimicrobial Agents and Chemotherapy. 1994 Oct 4–7; Orlando, 83

    Google Scholar 

  38. de Bony F, Bidault R, Peck R, et al. Lack of interaction between valaciclovir, the L-valyl ester of acyclovir, and Maalox antacid. J Antimicrob Chemother 1996 Feb; 37: 383–7

    Article  PubMed  Google Scholar 

  39. Soul-Lawton J, Chan PQ, Posner J, et al. Lack of interaction between valaciclovir, the L-valyl ester of aciclovir, and digoxin. 7th International Congress for Infectious Diseases. 1996 June 10–13; Hong Kong, 40

    Google Scholar 

  40. Rolan PE, Malliot F, On NT, et al. The effects of cimetidine and probenecid on the conversion of a valine ester of acyclovir, 256U, to acyclovir and acyclovir renal clearance in healthy volunteers [abstract]. Br J Clin Pharmacol 1993 May; 35: 533P

    Article  Google Scholar 

  41. Herne K, Cirelli R, Lee P, et al. Antiviral therapy of acute herpes zoster in older patients. Drugs Aging 1996 Feb; 8: 97–112

    Article  PubMed  CAS  Google Scholar 

  42. Beutner K. Herpes zoster in the elderly. J Geriatr Drug Ther 1995; 10(2): 25–51

    Article  Google Scholar 

  43. Beutner KR, Friedman DJ, Forszpaniak C, et al. Valaciclovir compared with aciclovir for improved therapy for herpes zoster in immunocompetent adults. Antimicrob Agents Chemother 1995 Jul; 39: 1546–53

    Article  PubMed  CAS  Google Scholar 

  44. Jeffries D, Barton S, Johnson R, et al. Issues in herpes management. Antiviral Chem Chemother 1994; 5 Suppl. 1: 23–9

    Google Scholar 

  45. Smiley ML, The International Valaciclovir Zoster Study Group. The efficacy and safety of valaciclovir for the treatment of herpes zoster. 33rd International Conference on Antimicrobial Agents and Chemotherapy. 1993 Oct 17–20; New Orleans, 340

    Google Scholar 

  46. Johnson RW, Crooks RJ, The International Valaciclovir Zoster Study Group. The clinical efficacy of valaciclovir in the treatment of herpes zoster [abstract]. J Europ Acad Dermatol and Venereol 1994; 5 Suppl. 1: S75.

    Google Scholar 

  47. Murray AB. Valaciclovir — an improvement over aciclovir for the treatment of zoster. Antiviral Chem Chemother 1995; 6 Suppl. 1: 34–8

    CAS  Google Scholar 

  48. Fife KH, International Valaciclovir HSV Study Group. Valaciclovir or acyclovir for the treatment of first episode genital herpes [abstract]. 35th Interscience Conference on Antimicrobial Agents and Chemotherapy. 1995 Sep 17–20; San Francisco, 243

    Google Scholar 

  49. Patel R, Delehanty J. Valaciclovir for the treatment of first episode genital herpes. 1st European Congress of Antimicrobial Chemotherapy. 1996 May 14–17, Glasgow

    Google Scholar 

  50. Spruance SL, Tyring SK, DeGregorio B, et al. A large-scale placebo-controlled, dose-ranging trial of peroral valaciclovir for episodic treatment of recurrent herpes genitalis. Arch Intern Med 1996 Aug 12/26; 156: 1729–35

    Article  PubMed  CAS  Google Scholar 

  51. International VHSVSG, Smiley ML. Valaciclovir and acyclovir for the treatment of recurrent genital herpes simplex virus infections [abstract]. 33rd Interscience Conference on Antimicrobial Agents and Chemotherapy. 1993 Oct 17–20; New Orleans, 341

    Google Scholar 

  52. GlaxoWellcome plc. Regulatory Submission report BQRT/ 95/0011

  53. Kinghorn GI, Fiddian AP, The International Valaciclovir HSV Study Group. Prevention of lesion development in recurrent genital herpes with oral valaciclovir [abstract]. J Europ Acad Dermat Venereol 1995; 5 Suppl. 1: no. P187, S162.

    Google Scholar 

  54. Woolley P, Crooks RJ, The International Valaciclovir HSV Group. Prevention of vesicular lesions in recurrent genital herpes with oral valaciclovir. 1st European Congress of Chemotherapy. 1996 May 14–17; Glasgow, W117

    Google Scholar 

  55. Aoki F. Valtrex (valaciclovir) for the treatment of recurrent genital herpes. Can J Infect Dis. 1995 Jul; 6: 287C.

    Google Scholar 

  56. Safrin S, International VHSVSG, et al. Valtrex (valaciclovir, VACV) for the treatment of recurrent genital herpes [abstract]. Antiviral Res 1995 Mar; 26: A234

    Article  Google Scholar 

  57. de Ruiter A, Thin RN. Genital herpes: a guide to pharmacological therapy. Drugs 1994; 47(2): 297–304

    Article  PubMed  Google Scholar 

  58. Ruhnek-Forsbek M, Sandstrom E, et al. Treatment of recurrent genital herpes simplex infection with oral acyclovir. J Antimicrob Chemother 1985; 16: 621–8

    Article  Google Scholar 

  59. Patel R, Crooks RJ, Bell AR, et al. Once-daily valaciclovir for suppresion of recurrent genital herpes — the first placebo-controlled clinical trial. 1st European Congress of Chemotherapy. 1996 May 14–17, Glasgow

    Google Scholar 

  60. Drucker JL, Miller JM, International Valaciclovir HSV Study Group. Once-daily valaciclovir sustains the suppressive efficacy and safety record of aciclovir in recurrent genital herpes. European Congress of Chemotherapy. 1996 May 14–17; Glasgow, W118

    Google Scholar 

  61. Feinberg J, Cooper D, Hurwitz S. Phase III study of valaciclovir (VACV) for cytomegalovirus (CMV) prophylaxis in patients with advanced HIV disease [abstract]. 35th Interscience Conference on Antimicrobial Agents and Chemotherapy. 1995 Sep 17–20; San Francisco, 243

    Google Scholar 

  62. Feinberg J, Cooper D, Hurwitz S. Phase III International Study of Valaciclovir (VACV) for cytomegalovirus (CMV) prophylaxis in patients with advanced HIV disease. 11th International Conference on AIDS. 1996 Jul 7–12; Vancouver, 225.

    Google Scholar 

  63. Sacks SL, Alrabiah F. Novel herpes treatment: a review. Exp Opin Invest Drugs 1996; 5(2): 169–83

    Article  CAS  Google Scholar 

  64. Mortality Morbidity Weekly Report. 1993 Oct 22; 42 (41): 306

    Google Scholar 

  65. Glaxo Wellcome. Valaciclovir Prescribing Information. UK, Nov 1995

  66. Conlon CP. Herpes Zoster. Prescr J 1995; 35(2): 46–52

    Google Scholar 

  67. Hong W, Cousins M. Postherpetic neuralgia: Is it preventable and treatable? Mod Med Aust 1996; 39(3): 84–7

    Google Scholar 

  68. Goldmeier D, Johnson A, Byrne M, et al. Psychosocial implications of recurrent genital herpes simplex virus infections. Genitourin Med 1988; 64: 327–30

    PubMed  CAS  Google Scholar 

  69. Manne S, Sandler I. Coping and adjustment to genital herpes. J Behav Med 1984; 7: 391–410

    Article  PubMed  CAS  Google Scholar 

  70. Beauchamp LM, Krenitsky TA. Acyclovir prodrugs: the road to valaciclovir. Drugs Future 1993 Jul; 18: 619–28

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Additional information

Various sections of the manuscript reviewed by: F.Y. Aoki, Department of Medical Microbiology, The University of Manitoba, Winnipeg, Manitoba, Canada; K. Beutner, Solano Dermatology Associates, A Medical Corporation, Vallejo, California, USA; W.K. Cheong, Specialist Skin Clinic, Singapore; C.P. Conion, Nuffield Department of Medicine, John Radcliffe Hospital, Oxford, England; H.J. Field, Centre for Veterinary Science, Cambridge University, Cambridge, England; B.G. Gazzard, Chelsea and Westminster Hospital, London, England; G.R. Kinghorn, Department of Genitourinary Medicine, Royal Hallamshire Hospital, Sheffield, England; R. Patel, Department of Genito Urinary Medicine, Royal South Hants Hospital, Southampton, England; C. Pederson, Department of Infectious Diseases, Hvidovre Hospital, Hvidovre, Denmark; R. Snoeck, Rega Institute, Katholieke Universiteit, Leuven, Belgium; S.K. Tyring, The University of Texas Medical Branch at Galveston, Center for Clinical Studies, Nassau Bay, Texas, USA; R.J. Whitley, Department of Pediatrics, Division of Clinical Virology, The University of Alabama at Birmingham, 616 Children’s Hospital, Birmingham, Alabama, USA; E.H. Wiltink, Department of Clinical Pharmacy, St Antonius Hospital, Nieuwegein, The Netherlands; M.J. Wood, Department of Infection and Tropical Medicine, Birmingham Heartlands Hospital, Birmingham, England; P.D. Woolley, Withington Hospital, Manchester, England.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Perry, C.M., Faulds, D. Valaciclovir. Drugs 52, 754–772 (1996). https://doi.org/10.2165/00003495-199652050-00009

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/00003495-199652050-00009

Keywords

Navigation