Evonik Evonik

X

Find Poviral manufacturers, exporters & distributors on PharmaCompass

PharmaCompass
API SUPPLIERS
API Suppliers

API Suppliers

0

US DMFs Filed

US DMFs Filed

0

CEP/COS Certifications

CEP/COS Certifications

0

JDMFs Filed

JDMFs Filed

0

Other Certificates

Other Certificates

0

Other Suppliers

Other Suppliers

0

API REF. PRICE (USD / KG)

0

INTERMEDIATES

0

DOSSIERS // FDF
USA (Orange Book)

USA (Orange Book)

0

Europe

Europe

0

Canada

Canada

0

Australia

Australia

0

South Africa

South Africa

0

Uploaded Dossiers

Uploaded Dossiers

0

GLOBAL SALES (USD Million)

U.S. Medicaid

0

Annual Reports

0

EXCIPIENTS

0

PATENTS & EXCLUSIVITIES

USFDA Orange Book Patents

0

USFDA Exclusivities

0

DIGITAL CONTENT

Blog #PharmaFlow

0

News

0

REF STANDARD

EDQM

0

USP

0

JP

0

Other Listed Suppliers

0

SERVICES

0

Poviral
Also known as: Aciclovir, Acycloguanosine, 59277-89-3, Zovirax, Vipral, Virorax
Molecular Formula
C8H11N5O3
Molecular Weight
225.20  g/mol
InChI Key
MKUXAQIIEYXACX-UHFFFAOYSA-N
FDA UNII
X4HES1O11F

A GUANOSINE analog that acts as an antimetabolite. Viruses are especially susceptible. Used especially against herpes.
Acyclovir is a Herpes Simplex Virus Nucleoside Analog DNA Polymerase Inhibitor, and Herpes Zoster Virus Nucleoside Analog DNA Polymerase Inhibitor, and Herpesvirus Nucleoside Analog DNA Polymerase Inhibitor. The mechanism of action of acyclovir is as a DNA Polymerase Inhibitor.
1 2D Structure

Poviral

2 Identification
2.1 Computed Descriptors
2.1.1 IUPAC Name
2-amino-9-(2-hydroxyethoxymethyl)-1H-purin-6-one
2.1.2 InChI
InChI=1S/C8H11N5O3/c9-8-11-6-5(7(15)12-8)10-3-13(6)4-16-2-1-14/h3,14H,1-2,4H2,(H3,9,11,12,15)
2.1.3 InChI Key
MKUXAQIIEYXACX-UHFFFAOYSA-N
2.1.4 Canonical SMILES
C1=NC2=C(N1COCCO)N=C(NC2=O)N
2.2 Other Identifiers
2.2.1 UNII
X4HES1O11F
2.3 Synonyms
2.3.1 MeSH Synonyms

1. 9-((2-hydroxyethoxy)methyl)guanine

2. Aci Sanorania

3. Aci-sanorania

4. Acic

5. Aciclobeta

6. Aciclostad

7. Aciclovir

8. Aciclovir Alonga

9. Aciclovir Sanorania

10. Aciclovir Von Ct

11. Aciclovir-sanorania

12. Acifur

13. Acipen Solutab

14. Acivir

15. Activir

16. Acyclo V

17. Acyclo-v

18. Acycloguanosine

19. Acyclovir Sodium

20. Alonga, Aciclovir

21. Antiherpes Creme

22. Avirax

23. Cicloferon

24. Clonorax

25. Cusiviral

26. Genvir

27. Herpetad

28. Herpofug

29. Herpotern

30. Herpoviric

31. Isavir

32. Laciken

33. Mapox

34. Maynar

35. Milavir

36. Opthavir

37. Sodium, Acyclovir

38. Solutab, Acipen

39. Supraviran

40. Viclovir

41. Vipral

42. Virax Puren

43. Virax-puren

44. Viraxpuren

45. Virherpes

46. Virmen

47. Virolex

48. Virupos

49. Virzin

50. Wellcome 248u

51. Wellcome-248u

52. Wellcome248u

53. Zoliparin

54. Zovirax

55. Zyclir

2.3.2 Depositor-Supplied Synonyms

1. Aciclovir

2. Acycloguanosine

3. 59277-89-3

4. Zovirax

5. Vipral

6. Virorax

7. Wellcome-248u

8. Aciclovirum

9. 9-[(2-hydroxyethoxy)methyl]guanine

10. 2-amino-9-((2-hydroxyethoxy)methyl)-1h-purin-6(9h)-one

11. Sitavig

12. Zovir

13. Acyclovir-side Chain-2-3h

14. Gerpevir

15. 9-(2-hydroxyethoxy)methylguanine

16. 9-hyroxyethoxymethylguanine

17. 9-((2-hydroxyethoxy)methyl)guanine

18. W-248-u

19. Novirus

20. 141294-79-3

21. 2-amino-9-[(2-hydroxyethoxy)methyl]-1,9-dihydro-6h-purin-6-one

22. Aciclovir [inn]

23. Nsc 645011

24. Chebi:2453

25. 2-amino-9-(2-hydroxyethoxymethyl)-1h-purin-6-one

26. 2-amino-9-(2-hydroxyethoxymethyl)-3h-purin-6-one

27. Acyclovir (aciclovir)

28. 2-amino-1,9-dihydro-9-((2-hydroxyethoxy)methyl)-6h-purin-6-one

29. 2-amino-1,9-dihydro-9-[(2-hydroxyethoxy)methyl]-6h-purin-6-one

30. 6h-purin-6-one, 2-amino-1,9-dihydro-9-[(2-hydroxyethoxy)methyl]-

31. Acyclovir-sidechain-2-3h

32. Zovirax (tn)

33. Mfcd00057880

34. 9-[(2-hydroxyethoxy)-methyl]guanine

35. Nsc-645011

36. Nsc-758477

37. X4hes1o11f

38. Mls000069633

39. Activir

40. 9-(2-hydroxyethoxymethyl)guanine

41. 6h-purin-6-one, 2-amino-1,9-dihydro-9-((2-hydroxyethoxy)methyl)-

42. Nsc645011

43. Ac2

44. Acyclovir [usan]

45. 2-amino-9-((2-hydroxyethoxy)methyl)-3h-purin-6(9h)-one

46. Ncgc00015061-02

47. Aciclovirum [latin]

48. Aciclovier

49. Hascovir

50. Smr000058225

51. Genvir

52. Maynar

53. Zyclir

54. Cas-59277-89-3

55. 6h-purin-6-one, 1,9-dihydro-2-amino-9-((2-hydroxyethoxy)methyl)-

56. Dsstox_cid_2556

57. 2-amino-9-(2-hydroxyethoxymethyl)purin-6-ol

58. Dsstox_rid_76626

59. Aciclovirum [inn-latin]

60. Dsstox_gsid_22556

61. Acicloftal

62. Cargosil

63. Viropump

64. Acyclofoam

65. 2-amino-9-[(2-hydroxyethoxy)methyl]-3,9-dihydro-6h-purin-6-one

66. 2-amino-9-[(2-hydroxyethoxy)methyl]-6,9-dihydro-3h-purin-6-one

67. Acic

68. Bw-248u

69. 2-amino-9-{[(2-hydroxyethyl)oxy]methyl}-1,9-dihydro-6h-purin-6-one

70. Acyclo-v

71. Acyclovir Lauriad

72. Drg-0008

73. Acyclovir (usp)

74. Bw 248u

75. Ccris 1953

76. Hsdb 6511

77. Sr-01000075540

78. Acyclovir [usan:usp]

79. Einecs 261-685-1

80. Acv & Pluronic F-68

81. Unii-x4hes1o11f

82. Acyclovir & Pluronic F-68

83. Cyclovir

84. Poviral

85. Sitavir

86. 2-amino-9-((2-hydroxyethoxy)methyl)-3,9-dihydro-6h-purin-6-one

87. Prestwick_6

88. 1pwy

89. Bw-248-u

90. Sitavig (tn)

91. Avaclyr

92. Spectrum_001739

93. Aciclovir [jan]

94. Acyclovir [mi]

95. Aciclovir [iarc]

96. Acyclovir [hsdb]

97. Opera_id_1674

98. Prestwick0_000086

99. Prestwick1_000086

100. Prestwick2_000086

101. Prestwick3_000086

102. Spectrum2_001563

103. Spectrum3_001874

104. Spectrum4_000225

105. Spectrum5_001093

106. Acyclovir [vandf]

107. Lopac-a-4669

108. Aciclovir (jp17/inn)

109. Aciclovir [mart.]

110. Chembl184

111. A 4669

112. Aciclovir [who-dd]

113. Aciclovir [who-ip]

114. Acyclovir [usp-rs]

115. Schembl3175

116. Lopac0_000037

117. Aciclovirum [who-ip]

118. Bspbio_000012

119. Bspbio_003348

120. Kbiogr_000889

121. Kbioss_002219

122. Bidd:gt0646

123. Divk1c_000185

124. Spectrum1503603

125. Spbio_001466

126. Spbio_001951

127. 2-amino-9-[(2-hydroxyethoxy)methyl]hydropurin-6-one

128. Bpbio1_000014

129. Gtpl4829

130. Schembl9828560

131. 9(2-hydroxyethoxymethyl)guanine

132. Acyclovir [orange Book]

133. Aciclovir [ep Monograph]

134. Dtxsid1022556

135. Hms500j07

136. Kbio1_000185

137. Kbio2_002219

138. Kbio2_004787

139. Kbio2_007355

140. Kbio3_002850

141. Acyclovir [usp Monograph]

142. Ninds_000185

143. Hms1568a14

144. Hms1922e08

145. Hms2090g09

146. Hms2095a14

147. Hms2234k21

148. Hms3259n10

149. Hms3260g15

150. Hms3269m15

151. Hms3372k02

152. Hms3413d21

153. Hms3655c14

154. Hms3677d21

155. Hms3712a14

156. Pharmakon1600-01503603

157. Bcp11036

158. Zinc1530555

159. 9-(2-hydroxyethoxy Methyl) Guanine

160. Tox21_110075

161. Tox21_500037

162. Bbl009642

163. Bdbm50021776

164. Bdbm50103518

165. Ccg-39909

166. Nsc758477

167. Nsc780378

168. S1807

169. Stk796771

170. Stl257059

171. Stl301862

172. Akos000656213

173. Akos015995680

174. Akos022135433

175. Tox21_110075_1

176. Ac-8068

177. Cs-1353

178. Db00787

179. Ks-1027

180. Lp00037

181. Nc00717

182. Nsc-780378

183. Sdccgsbi-0050026.p003

184. Idi1_000185

185. Smp1_000007

186. Ncgc00015061-01

187. Ncgc00015061-03

188. Ncgc00015061-04

189. Ncgc00015061-05

190. Ncgc00015061-06

191. Ncgc00015061-07

192. Ncgc00015061-08

193. Ncgc00015061-09

194. Ncgc00015061-10

195. Ncgc00015061-12

196. Ncgc00015061-13

197. Ncgc00015061-28

198. Ncgc00015061-29

199. Ncgc00022426-03

200. Ncgc00093555-01

201. Ncgc00093555-02

202. Ncgc00093555-03

203. Ncgc00093555-04

204. Ncgc00167756-01

205. Ncgc00167756-02

206. Ncgc00260722-01

207. Ncgc00381719-03

208. Hy-17422

209. Sy051130

210. Acycloguanosine, >=99% (hplc), Powder

211. Aciclovir 1.0 Mg/ml In Dimethyl Sulfoxide

212. Am20100442

213. Eu-0100037

214. Ft-0621607

215. Ft-0657847

216. Sw196324-3

217. C06810

218. D00222

219. 277a893

220. A832236

221. A935190

222. Q147101

223. Q-200591

224. Sr-01000075540-1

225. Sr-01000075540-3

226. Sr-01000075540-5

227. 2-amino-9-[(2-hydroxyethoxy)methyl]-9h-purin-6-ol

228. 2-azanyl-9-(2-hydroxyethyloxymethyl)-3h-purin-6-one

229. Brd-k32318651-001-17-9

230. Aciclovir, British Pharmacopoeia (bp) Reference Standard

231. F2173-0946

232. Aciclovir, European Pharmacopoeia (ep) Reference Standard

233. Valaciclovir Hydrochloride Impurity B [ep Impurity]

234. 2-amino-9-(2-hydroxy-ethoxymethyl)-5,9-dihydro-purin-6-one

235. Acyclovir, United States Pharmacopeia (usp) Reference Standard

236. 2-amino-9-(2-hydroxy-ethoxymethyl)-5,9-dihydro-purin-6-one (acv)

237. 2-amino-9-[(2-hydroxyethoxy)methyl]-1,9-dihydro-6h-purin-6-one #

238. Valaciclovir Hydrochloride Hydrate Impurity B [ep Impurity]

239. Acv; Acycloguanosine; Acyclovir; Nsc 645011; Nsc-645011; Nsc645011

240. Acyclovir, Pharmaceutical Secondary Standard; Certified Reference Material

241. Aciclovir For Peak Identification 1, European Pharmacopoeia (ep) Reference Standard

242. Aciclovir For Peak Identification 2, European Pharmacopoeia (ep) Reference Standard

243. Aciclovir For System Suitability, European Pharmacopoeia (ep) Reference Standard

2.4 Create Date
2019-01-10
3 Chemical and Physical Properties
Molecular Weight 225.20 g/mol
Molecular Formula C8H11N5O3
XLogP3-1.9
Hydrogen Bond Donor Count3
Hydrogen Bond Acceptor Count5
Rotatable Bond Count4
Exact Mass225.08618923 g/mol
Monoisotopic Mass225.08618923 g/mol
Topological Polar Surface Area115 Ų
Heavy Atom Count16
Formal Charge0
Complexity308
Isotope Atom Count0
Defined Atom Stereocenter Count0
Undefined Atom Stereocenter Count0
Defined Bond Stereocenter Count0
Undefined Bond Stereocenter Count0
Covalently Bonded Unit Count1
4 Drug and Medication Information
4.1 Therapeutic Uses

Antiviral Agents

National Library of Medicine's Medical Subject Headings. Acyclovir. Online file (MeSH, 2014). Available from, as of November 19, 2013: https://www.nlm.nih.gov/mesh/2014/mesh_browser/MBrowser.html


IV acyclovir sodium is used for the treatment of initial and recurrent mucocutaneous herpes simplex virus (HSV-1 and HSV-2) infections and the treatment of varicella-zoster infections in immunocompromised adults and children; for the treatment of severe first episodes of genital herpes infections in immunocompetent individuals; and for the treatment of HSV encephalitis and neonatal HSV infections.

American Society of Health-System Pharmacists 2013; Drug Information 2013. Bethesda, MD. 2013, p. 766


Acyclovir is used orally for the treatment of initial and recurrent episodes of genital herpes; for the acute treatment of herpes zoster (shingles, zoster) in immunocompetent individuals; and for the treatment of varicella (chickenpox) in immunocompetent individuals.

American Society of Health-System Pharmacists 2013; Drug Information 2013. Bethesda, MD. 2013, p. 766


Oral acyclovir is indicated in the treatment of initial episodes of genital herpes infection in immunocompetent and immunocompromised patients. Parenteral acyclovir is indicated in the treatment of severe initial episodes of genital herpes infection in immunocompetent patients and in patients who are unable to take (or absorb) oral acyclovir. /Included in US product labeling/

Thomson.Micromedex. Drug Information for the Health Care Professional. 24th ed. Volume 1. Plus Updates. Content Reviewed by the United States Pharmacopeial Convention, Inc. Greenwood Village, CO. 2004., p. 26


For more Therapeutic Uses (Complete) data for ACYCLOVIR (15 total), please visit the HSDB record page.


4.2 Drug Warning

Parenteral acyclovir therapy can cause signs and symptoms of encephalopathy. ... Acyclovir should be used with caution in patients with underlying neurologic abnormalities and in patients with serious renal, hepatic, or electrolyte abnormalities or substantial hypoxia. The drug also should be used with caution in patients who have manifested prior neurologic reactions to cytotoxic drugs or those receiving intrathecal methotrexate or interferon.

American Society of Health-System Pharmacists 2013; Drug Information 2013. Bethesda, MD. 2013, p. 767


Acyclovir should be used with caution in patients receiving other nephrotoxic drugs concurrently since the risk of acyclovir-induced renal impairment and/or reversible CNS symptoms is increased in these patients. Adequate hydration should be maintained in patients receiving IV acyclovir; however, in patients with encephalitis, the recommended hydration should be balanced by the risk of cerebral edema. Because the risk of acyclovir-induced renal impairment is increased during rapid IV administration of the drug, acyclovir should be given only by slow IV infusion (over 1 hour).

American Society of Health-System Pharmacists 2013; Drug Information 2013. Bethesda, MD. 2013, p. 767


There are no adequate and controlled studies to date using acyclovir in pregnant women, and the drug should be used during pregnancy only when the potential benefits justify the possible risks to the fetus.

American Society of Health-System Pharmacists 2013; Drug Information 2013. Bethesda, MD. 2013, p. 767


Maternal Medication usually Compatible with Breast-Feeding: Acyclovir: Reported Sign or Symptom in Infant or Effect on Lactation: None. /from Table 6/

Report of the American Academy of Pediatrics Committee on Drugs in Pediatrics 93 (1): 140 (1994)


For more Drug Warnings (Complete) data for ACYCLOVIR (20 total), please visit the HSDB record page.


4.3 Drug Indication

An acyclovir topical cream is indicated to treat recurrent herpes labialis in immunocompetent patients 12 years and older. Acyclovir oral tablets, capsules, and suspensions are indicated to treat herpes zoster, genital herpes, and chickenpox. An acyclovir topical ointment is indicated to treat initial genital herpes and limited non-life-threatening mucocutaneous herpes simplex in immunocompromised patients. An acyclovir cream with hydrocortisone is indicated to treat recurrent herpes labialis, and shortening lesion healing time in patients 6 years and older. An acyclovir buccal tablet is indicated for the treatment of recurrent herpes labialis. An acyclovir ophthalmic ointment is indicated to treat acute herpetic keratitis.


Treatment of herpes simplex labialis


Prevention of recurrences of herpes simplex labialis, Treatment of recurrent herpes simplex labialis


5 Pharmacology and Biochemistry
5.1 Pharmacology

Acyclovir is a nucleoside analog that inhibits the action of viral DNA polymerase and DNA replication of different herpesvirus. Acyclovir has a wide therapeutic window as overdose is rare in otherwise healthy patients.


5.2 MeSH Pharmacological Classification

Antiviral Agents

Agents used in the prophylaxis or therapy of VIRUS DISEASES. Some of the ways they may act include preventing viral replication by inhibiting viral DNA polymerase; binding to specific cell-surface receptors and inhibiting viral penetration or uncoating; inhibiting viral protein synthesis; or blocking late stages of virus assembly. (See all compounds classified as Antiviral Agents.)


5.3 FDA Pharmacological Classification
5.3.1 Active Moiety
ACYCLOVIR
5.3.2 FDA UNII
X4HES1O11F
5.3.3 Pharmacological Classes
Herpes Simplex Virus Nucleoside Analog DNA Polymerase Inhibitor [EPC]; Herpes Zoster Virus Nucleoside Analog DNA Polymerase Inhibitor [EPC]; Herpesvirus Nucleoside Analog DNA Polymerase Inhibitor [EPC]; Nucleoside Analog [EXT]; DNA Polymerase Inhibitors [MoA]
5.4 ATC Code

J05AB01

S76 | LUXPHARMA | Pharmaceuticals Marketed in Luxembourg | Pharmaceuticals marketed in Luxembourg, as published by d'Gesondheetskeess (CNS, la caisse nationale de sante, www.cns.lu), mapped by name to structures using CompTox by R. Singh et al. (in prep.). List downloaded from https://cns.public.lu/en/legislations/textes-coordonnes/liste-med-comm.html. Dataset DOI:10.5281/zenodo.4587355


D - Dermatologicals

D06 - Antibiotics and chemotherapeutics for dermatological use

D06B - Chemotherapeutics for topical use

D06BB - Antivirals

D06BB03 - Aciclovir


J - Antiinfectives for systemic use

J05 - Antivirals for systemic use

J05A - Direct acting antivirals

J05AB - Nucleosides and nucleotides excl. reverse transcriptase inhibitors

J05AB01 - Aciclovir


S - Sensory organs

S01 - Ophthalmologicals

S01A - Antiinfectives

S01AD - Antivirals

S01AD03 - Aciclovir


5.5 Absorption, Distribution and Excretion

Absorption

The oral bioavailability of acyclovir is 10-20% but decreases with increasing doses. Acyclovir ointment is <0.02-9.4% absorbed. Acyclovir buccal tablets and ophthalmic ointment are minimally absorbed. The bioavailability of acyclovir is not affected by food. Acyclovir has a mean Tmax of 1.10.4 hours, mean Cmax of 593.7-656.5ng/mL, and mean AUC of 2956.6-3102.5h/*ng/mL.


Route of Elimination

The majority of acyclovir is excreted in the urine as unchanged drug. 90-92% of the drug can be excreted unchanged through glomerular filtration and tubular secretion. <2% of the drug is recovered in feces and <0.1% is expired as CO2.


Volume of Distribution

The volume of distribution of acyclovir is 0.6L/kg.


Clearance

The renal clearance of acyclovir is 248mL/min/1.73m2. The total clearance in neonates if 105-122mL/min/1.73m2.


Absorption of acyclovir from the GI tract is variable and incomplete. 15-30% of an oral dose of the drug is absorbed. Some data suggest that GI absorption of acyclovir may be saturable; in a crossover study in which acyclovir was administered orally to healthy adults as 200 mg capsules, 400 mg tablets, or 800 mg tablets 6 times daily, the extent of absorption decreased with increasing dose, resulting in bioavailabilities of 20, 15, or 10%, respectively. ... This decrease in bioavailability appears to be a function of increasing dose, not differences in dosage forms. In addition, steady-state peak and trough plasma acyclovir concentrations were not dose proportional over the oral dosing range of 200-800 mg 6 times daily, averaging 0.83 and 0.46, 1.21 and 0.63, or 1.61 and 0.83 ug/ml for the 200, 400, or 800 mg dosing regimens, respectively. Peak plasma concentrations usually occur within 1.5-2.5 hours after oral administration.

American Society of Health-System Pharmacists 2013; Drug Information 2013. Bethesda, MD. 2013, p. 769


In a multiple dose study in neonates up to 3 months of age, IV infusion over 1 hour of 5, 10, or 15 mg/kg of acyclovir every 8 hours resulted in mean steady state peak serum concentrations of 6.8, 13.9, and 19.6 ug/ml, respectively, and mean steady state trough serum concentration of 1.2, 2.3, and 3.1 ug/ml, respectively. In another multiple dose study in pediatric patients, IV infusion over 1 hour of 250 or 500 mg/sq m of acyclovir every 8 hours resulted in mean steady state peak serum concentrations of 10.3 and 20.7 ug/ml, respectively.

American Society of Health-System Pharmacists 2013; Drug Information 2013. Bethesda, MD. 2013, p. 769


Acyclovir is widely distributed into body tissues and fluids including the brain, kidney, saliva, lung, liver, muscle, spleen, uterus, vaginal mucosa and secretions, cerebrospinal fluid, and herpetic vesicular fluid. The drug also is distributed into semen, achieving concentrations about 1.4 and 4 times those in plasma during chronic oral therapy at dosages of 400 mg and 1 g daily, respectively. The apparent volume of distribution of acyclovir is reported to be 32.4-61.8 liter/1.73 sq m in adults and 28.8, 31.6, 42, or 51.2-53.6 liter/1.73 sq m in neonates up to 3 months of age, children 1-2 years; 2-7 years; or 7-12 years of age, respectively.

American Society of Health-System Pharmacists 2013; Drug Information 2013. Bethesda, MD. 2013, p. 769


Acyclovir crosses the placenta. Limited data indicate that the drug is distributed into milk, generally in concentrations greater than concurrent maternal plasma concentrations, possibly via an active transport mechanism.

American Society of Health-System Pharmacists 2013; Drug Information 2013. Bethesda, MD. 2013, p. 769


For more Absorption, Distribution and Excretion (Complete) data for ACYCLOVIR (13 total), please visit the HSDB record page.


5.6 Metabolism/Metabolites

Acyclovir is <15% oxidized to 9-carboxymethoxymethylguanine by alcohol dehydrogenase and aldehyde dehydrogenase and 1% 8-hydroxylated to 8-hydroxy-acyclovir by aldehyde oxidase. Acyclovir is becomes acyclovir monophosphate due to the action of viral thymidine kinase. Acyclovir monophosphate is converted to the diphosphate form by guanylate kinase. Acyclovir diphosphate is converted to acyclovir triphosphate by nucleoside diphosphate kinase, pyruvate kinase, creatine kinase, phosphoglycerate kinase, succinyl-CoA synthetase, phosphoenolpyruvate carboxykinase and adenylosuccinate synthetase.


Acyclovir is metabolized partially to 9-carboxymethoxymethylguanine and minimally to 8-hydroxy-9-(2-hydroxyethoxymethyl)guanine. In vitro, acyclovir also is metabolized to acyclovir monophosphate, diphosphate, and triphosphate in cells infected with herpes viruses, principally by intracellular phosphorylation of the drug by virus coded thymidine kinase and several cellular enzymes.

American Society of Health-System Pharmacists 2013; Drug Information 2013. Bethesda, MD. 2013, p. 769


5.7 Biological Half-Life

The clearance of acyclovir varies from 2.5-3 hours depending on the creatinine clearance of the patient. The plasma half life of acyclovir during hemodialysis is approximately 5 hours. The mean half life in patients from 7 months to 7 years old is 2.6 hours.


Plasma concentrations of acyclovir appear to decline in a biphasic manner. In adults with normal renal function, the half-life of acyclovir in the initial phase averages 0.34 hours and the half-life in the terminal phase averages 2.1-3.5 hours. In adults with renal impairment, both half-life in the initial phase and half-life in the terminal phase may be prolonged, depending on the degree of renal impairment. In a study in adults with anuria, the half-life in the initial phase of acyclovir averaged 0.71 hours. In several studies, the half-life in the terminal phase of acyclovir averaged 3,3.5, or 19.5 hours in adults with creatinine clearances of 50-80 or 15-50 ml/minute per 1.73 sq m or with anuria, respectively. In patients undergoing hemodialysis, the half-life in the terminal phase of acyclovir during hemodialysis averaged 5.4-5.7 hours.

American Society of Health-System Pharmacists 2013; Drug Information 2013. Bethesda, MD. 2013, p. 769


In neonates, the half-life of acyclovir depends principally on the maturity of renal mechanisms for excretion as determined by gestational age, chronologic age, and weight. In children older than 1 year of age, the half-life of the drug appears to be similar to that of adults. The half-life in the terminal phase averages 3.8-4.1, 1.9, 2.2-2.9, or 3.6 hours in neonates up to 3 months of age, children 1-2 years, 2-12 years, or 12-17 years of age, respectively.

American Society of Health-System Pharmacists 2013; Drug Information 2013. Bethesda, MD. 2013, p. 769


5.8 Mechanism of Action

Acyclovir is becomes acyclovir monophosphate due to the action of viral thymidine kinase. Acyclovir monophosphate is converted to the diphosphate form by guanylate kinase. Acyclovir diphosphate is converted to acyclovir triphosphate by nucleoside diphosphate kinase, pyruvate kinase, creatine kinase, phosphoglycerate kinase, succinyl-CoA synthetase, phosphoenolpyruvate carboxykinase and adenylosuccinate synthetase. Acyclovir triphosphate has higher affinity for viral DNA polymerase than cellular DNA polymerase and incorporates into the DNA where the missing 2' and 3' carbons causes DNA chain termination. In other cases acyclovir triphosphate competes so strongly for viral DNA polymerase that other bases cannot associate with the enzyme, inactivating it.


Acyclovir is a synthetic purine nucleoside analogue with in vitro and in vivo inhibitory activity against herpes simplex virus types 1 (HSV-1), 2 (HSV-2), and varicella-zoster virus (VZV). The inhibitory activity of acyclovir is highly selective due to is affinity for the enzyme thymidine kinase (TK) encoded by HSV and VZV. This viral enzyme converts acyclovir into acyclovir monophosphate, a nucleotide analogue. The monophosphate is further converted into diphosphate by cellular guanylate kinase adn into triphosphate by a number of cellualr enzymes. In vitro, acyclovir triphosphate stops replication of herpes viral DNA.

Physicians Desk Reference 65th ed. PDR Network, LLC, Montvale, NJ. 2011, p. 1650


Acyclovir inhibits viral DNA synthesis ... . Its selectivity of action depends on interaction with two distinct viral proteins. Cellular uptake and initial phosphorylation are facilitated by HSV thymidine kinase. The affinity of acyclovir for HSV thymidine kinase is about 200-fold greater than for the mammalian enzyme. Cellular enzymes convert the monophosphate to acyclovir triphosphate, which is present in 40- to 100-fold higher concentrations in HSV-infected than in uninfected cells, and competes for endogenous deoxyguanosine triphosphate (dGTP). The immunosuppressive agent mycophenolate mofetil potentiates the antiherpes activity of acyclovir and related agents by depleting intracellular dGTP pools. Acyclovir triphosphate competitively inhibits viral DNA polymerases and, to a much smaller extent, cellular DNA polymerases. Acyclovir triphosphate also is incorporated into viral DNA, where it acts as a chain terminator because of the lack of 3'-hydroxyl group. By a mechanism termed suicide inactivation, the terminated DNA template containing acyclovir binds the enzyme and leads to irreversible inactivation of the DNA polymerase.

Brunton, L. Chabner, B, Knollman, B. Goodman and Gillman's The Pharmaceutical Basis of Therapeutics, Twelth Edition, McGraw Hill Medical, New York, NY. 2011, p. 1594


The concentration of the endogenous neurotoxin quinolinic acid (QA) is increased in the central nervous system of mice with herpes simplex encephalitis. /The authors/ have previously shown that the antiherpetic agent acyclovir (AC) has the ability to reduce QA-induced neuronal damage in rat brain, by attenuating lipid peroxidation. The mechanism by which QA induces lipid peroxidation includes the enhancement of the iron (Fe)-mediated Fenton reaction and the generation of free radicals, such as the superoxide anion (O(2)(-)). Thus, the present study determined whether AC has the ability to reduce Fe(2+)-induced lipid peroxidation, O(2)(-) generation and QA-induced superoxide anion generation, and to bind free Fe. O(2)(-) and Fe(2+) are also cofactors of the enzymes, indoleamine-2,3-dioxygenase (IDO) and 3-hydroxyanthranilate-3,4-dioxygenase (3-HAO) respectively. These enzymes catalyse steps in the biosynthesis of QA; thus, the effect of AC on their activity was also investigated. AC significantly attenuates Fe(2+)-induced lipid peroxidation and O(2)(-) generation. AC reduces O(2)(-) generation in the presence of QA and strongly binds Fe(2+) and Fe(3+). It also reduces the activity of both IDO and 3-HAO, which could be attributed to the superoxide anion scavenging and iron binding properties, respectively, of this drug.

PMID:17174341 Muller AC et al; Life Sci 80 (10): 918-25 (2007)


Ask Us for Pharmaceutical Supplier and Partner
Ask Us, Find A Supplier / Partner
No Commissions, No Strings Attached, Get Connected for FREE

What are you looking for?

How can we help you?

The request can't be empty

Please read our Privacy Policy carefully

You must agree to the privacy policy

The name can't be empty
The company can't be empty.
The email can't be empty Please enter a valid email.
The mobile can't be empty
Post Enquiry
POST ENQUIRY