Quick viewing(Text Mode)

Review Selective Anti-Herpesvirus Agents

Review Selective Anti-Herpesvirus Agents

Antiviral Chemistry & 2013; 23:93–101 (doi: 10.3851/IMP2533)

Review Selective anti-herpesvirus agents

Erik De Clercq1*

1Rega Institute for Medical Research, KU Leuven, Leuven, Belgium

*Corresponding author e-mail: [email protected]

This review article focuses on the anti-herpesvirus agents ) or are under clinical development (CMX001 effective against virus, varicella-zos- [the hexadecyloxypropyl prodrug of ], the heli- ter virus and cytomegalovirus, which have either been case-primase inhibitor BAY 57-1293 [now referred to as licensed for clinical use (idoxuridine, , brivu- AIC316], FV-100 [the valine ester of Cf 1743] and the din, acyclovir, , , and terminase inhibitor [AIC246]).

Idoxuridine and trifluridine

The era started with the discovery of 5-fluorouracil [10,11]. Consequently, BVaraU was not idoxuridine (IDU) and trifluridine (TFT; Figure 1), further developed for the treatment of VZV infections, which were both launched, and are still used today, for whereas BVDU was marketed for this indication in sev- the topical treatment of herpetic eye infections (that is, eral European countries (including Germany, Italy and herpetic keratitis) due to herpes simplex virus (HSV). Belgium), with the warning, however, that, as a precau- IDU was first described by Prusoff [1] in 1959 as a tion, it should not be combined with 5-fluorouracil (or potential anti-tumour agent, whereas both IDU and TFT any of its prodrugs). were subsequently described as anti-HSV agents for the treatment of herpetic keratitis by Kaufman et al. [2–5]. Brivudin and BVaraU The first antiviral drug used for the systemic treatment of herpesvirus (HSV and VZV) infections was vidara- IDU and TFT could be considered as the starting point bine (Figure 4). Its clinical use was prompted thanks for the synthesis of various new 5-substituted 2′-deoxy- to the pioneering efforts of Whitley et al. [12–14]. Vid- derivatives, the most prominent of this series arabine was later abandoned for a number of reasons, being brivudin (BVDU; Figure 2), which proved to be including relative insolubility in aqueous medium, and a highly specific inhibitor of HSV-1 as well as varicella- rapid deamination, by the ubiquitous adenosine deami- zoster virus (VZV) [6,7]. The arabinosyl counterpart of nase, to its inactive counterpart, the principal BVDU (BVaraU; Figure 2) was subsequently found to reason being that it was surpassed in potency by acyclo- be equally potent as, if not more so than, BVDU against vir, which, incidentally, was discovered in the original VZV [8]. attempts to inhibit the rapid deamination of vidarabine. However, BVDU and BVaraU can be cleaved by hydrolases to release the free aglycon, 5-bromovinylu- Acyclovir racil (BVU), which acts as a powerful inhibitor of dihy- drothymine dehydrogenase, the enzyme that catalyzes The antiviral drug era was greatly boosted by the dis- the hydrogenation of and its 5-substituted deriv- covery of acyclovir (Figure 5) as a selective antiherpetic atives (such as 5-fluorouracil), the first step in the catab- agent [15]. In two papers that appeared in December olism of the uracil derivatives (Figure 3). The conver- 1977 [16] and April 1978 [17], acyclovir was described sion of BVDU to BVU primarily occurs in the liver (and as a selective antiherpetic agent, whose antiviral activity is reversible); that of BVaraU to BVU takes place in the depended on a specific phosphorylation by the virus- gastrointestinal lumen (by the gastrointestinal bacteria) induced kinase (apparently, the guanosine [9], is essentially irreversible, and has been shown to analogue acyclovir shared sufficient structural require- lead to some casualties, due to the enhanced toxicity of ments with thymidine to be recognized as a substrate by

©2013 International Medical Press 1359-6535 (print) 2040-2066 (online) 93 E De Clercq

the HSV-induced thymidine kinase, which also acts as (valaciclovir) also showed increased oral bioavailability a deoxycytidine kinase) [18,19]. Both BVDU and acy- and this finding [25] prompted the design of prodrugs of clovir were then shown to prevent the establishment of other antivirals as well. latent HSV infection in mice [20], and the molecular basis for the resistance of HSV against acyclovir was Famciclovir, valganciclovir, CMX001 and further characterized by Field and Darby et al. [21–23]. MIV-606 Valaciclovir Following the finding of increased oral bioavailability of valaciclovir, synthesized prodrugs included: famciclovir, In attempts to increase the aqueous solubility of acy- the double diacetyl precursor of 6-deoxy (Fig- clovir (so as to make it applicable as eye drops [instead ure 7); valganciclovir, the valine ester prodrug of ganciclo- of ointment], or amenable to intramuscular [instead of vir (Figure 8); CMX001, the hexadecyloxypropyl prodrug intravenous] injection), amino acid esters of acyclovir of cidofovir (Figure 9), and MIV-606, the valomaciclovir were synthesized (that is, glycyl- and alanyl-acyclovir; stearate prodrug of H2G (Figure 10). All these antiviral Figure 6) [24]. As an additional bonus, the valine ester prodrugs fulfil an essential requirement for antiviral effi- cacy, that is they possess an increased oral bioavailabil- ity [26]. Single-day famciclovir (1,000 mg twice daily) Figure 1. The first antivirals: 5-substituted 2′- has been recommended for recurrent genital prophylaxis [27], and valganciclovir and valaciclovir have been recom- mended for preemptive prophylaxis of cytomegalovirus O O (CMV) disease (after renal transplantation) [28]. I CF3 HN HN Foscarnet O N O N The smallest of all antivirals, which, for parenteral use, HO HO O O must be injected intravenously, is foscarnet (phospho- noformic acid [PFA]). PFA has been approved for the treatment of CMV infections, but can also be used, off HO HO label, for the treatment of HSV and VZV infections that are resistant to acyclovir. Characteristic for PFA is the IDU TFT Idoxuridine Triuridine presence of a phosphonate (P–C) bridge (Figure 11), a 5-lodo-2′- 5-Triuoromethyl-2′-deoxyuridine hallmark that guarantees stability against (spontaneous Herpid®, Stoxil® Viroptic® and enzymatic) degradation and which is also found in the acyclic nucleoside phosphonates (that is, cidofovir, and tenofovir).

Figure 2. The (E)-5-(2-bromovinyl) component marking highly potent activity against varicella-zoster virus

O H Br O H Br

HN HN H H

O N O N HO HO O O HO

HO HO

BVDU BVaraU Brivudin (E)-5-(2-Bromovinyl)-2′-deoxyuridine Zostex®, Brivirac®, Zerpex®

94 ©2013 International Medical Press Selective anti-herpesvirus agents

Current state of the art: approved Figure 3. (E)-5-(2-bromovinyl)uracil inhibits degradation of antiherpesvirus drugs 5-fluorouracil

The antiviral agents have been the subject of a fact file BVDU BVaraU O first published by De Clercq et al. [29] in 2006, followed H Br by a second edition in 2008 [30–32]. For the treatment HN of herpesvirus infections (HSV, VZV, CMV and Epstein- H Barr virus), the systemic antiviral agents, as currently O N BVU approved by the US Food and Drug Administration, are H listed in Table 1. This list includes acyclovir, valaciclo- vir and famciclovir for the treatment of HSV and VZV O O infections, and , valganciclovir, cidofovir and F F foscarnet for the treatment of CMV infections (the only HN HN antisense oligonucleotide that ever received marketing Dihydrothymine O N O N approval as an antiviral drug was [30]: it H dehydrogenase H was approved for the topical treatment, upon intravit- 5-Fluorouracil 5-Fluoro-5,6- real inoculation, of CMV retinitis). Off label, cidofovir dihydrouracil and foscarnet are occasionally used for the treatment of severe HSV or VZV infections that have become resistant BVDU, brivudin; BVU, (E)-5-(2-bromovinyl)uracil. to acyclovir, valaciclovir or famciclovir (Table 1).

Figure 4. Vidarabine, the first antiviral drug for systemic use New anti-varicella-zoster virus drugs on the horizon: FV-100 NH In 2000, McGuigan et al. [33] described an exquisitely 2 N potent anti-VZV agent (Cf 1743), which was exclusively N active against VZV without any antiviral activity against any other virus including HSV; the prototype of this class N N of bicyclic furano pyrimidine nucleosides proved active HO against a large array of VZV strains at picomolar con- O HO centrations [34]. As previously carried out for acyclovir and ganciclovir, Cf 1743 (Figure 12) was converted to its valine ester, and this prodrug, dubbed FV-100, can OH be considered as the most potent and selective anti-VZV Arabinosyladenine agent reported to date [35]. Phase II clinical trials with Ara-A FV-100 are in progress. It should be administered once- Vidarabine ® daily (for 7 days) at a dose of 100, 200, 400 or 800 mg Vira-A (exact dosage still to be determined) [36], which com- pares favourably with valaciclovir that has to be given orally 3× daily at 1,000 mg. The helicase-primase inhibitor ASP2151 (Figure 13), Figure 5. Acyclovir, the ‘gold’-standard for treatment of assumed to be effective against VZV as well as HSV has herpes simplex virus infections been abandoned for further development [37].

New anti-herpes-simplex virus drugs on the O horizon: BAY 57-1293, AIC316, N HN The helicase-primase inhibitor BAY 57-1293 (Fig- H N N N ure 14) was first described by Kleymann et al. [38,39] 2 HO in 2002. This compound, together with BILS 179 BS O [40], focused on a new target for new antiviral agents active against HSV. BAY 57-1293 was transferred Acyclovir 9-(2-Hydroxyethoxymethyl) from Bayer to AiCuris for further development. It Zovirax® has been extensively studied for its potential to gen- erate drug-resistant mutants by Biswas and Field et

Antiviral Chemistry & Chemotherapy 23.3 95 E De Clercq

al. [41–52]. The clinical potential of BAY 57-1293 is CMX001, FV-100, BAY 57-1293 (AIC316) and restricted to HSV. It remains to be determined to what AIC246 (letermovir; Table 2). For an update on new extent BAY 57-1293 (AIC316) compares to the utility antivirals under development for the treatment of dou- of acyclovir, still the ‘gold’-standard for the treatment ble-stranded DNA virus (including HSV) infection, see of HSV infections. Dropulic and Cohen [53]. The development of mariba- vir (Figure 15) for the treatment of CMV infections has New anti-cytomegalovirus drugs on the been discontinued [54]. horizon: AIC246, letermovir Instead, AIC246 (letermovir; Figure 16), which inhib- its HCMV replication via a mechanism that involves At present, there are four new candidate compounds the viral terminase but is distinct from that of mariba- for the treatment of HSV, VZV and CMV infections: vir [55], is now being pursued for its clinical potential

Figure 6. Amino acyl prodrugs of acyclovir

O

N HN

N H2N N HO O

Acyclovir

O O O N N N HN HN HN N N H N N N H2N N H2N N 2 H N CH CO O H2N CH CO O H2N CH2 CO O 2 O O O CH CH3 H3CCH3 Glycyl-acyclovir Alanyl-acyclovir Valaciclovir Valyl-acyclovir Valtrex®, Zelitrex®

Figure 7. Famciclovir, the orally bioavailable prodrug of Figure 8. Valganciclovir, the orally bioavailable prodrug of penciclovir ganciclovir

N O N O O N H N N N HN 2 N N H3C CO O HN HN H N N N 2 N N HO H2N N H2N N HO H2N CH CO O O O O CH CO HC3 CH3 OH CH3 OH OH Penciclovir Famciclovir Ganciclovir Valganciclovir Denavir®, Vectavir® Diacetyl 6-deoxy-9-(4-hydroxy-3- Cymevene®, Cytovene® Valcyte® hydroxymethyl-but-1-yl)guanine Famvir®

96 ©2013 International Medical Press Selective anti-herpesvirus agents

Figure 9. CMX001, the orally bioavailable prodrug of cidofovir

NH2 NH2

N N

O N O N O O

O P OH O P O (CH2)3 O (CH2)15CH3 OH OH HO HO Cidofovir Hexadecyloxypropyl-cidofovir Vistide® CMX001

Figure 10. The orally bioavailable H2G prodrug Figure 11. Foscarnet, the smallest of all antiviral drugs

O

N HN O H3C CH3 OH N CH H2N N HO P C O CH OH O C NH2 O O PFA C Foscarnet O (CH2)16CH3 Phosphonoformic acid Foscavir® Valomaciclovir stearate H2G prodrug MIV-606

Table 1. State of the art: currently approved systemic antiviral agents against herpesvirus infections

Drug HSV VZV CMV EBV

Acyclovir + + - - Valaciclovir + + - - Famciclovir + + - - Ganciclovir - - + - Valganciclovir - - + - Cidofovir OL OL + - Foscarnet OL OL + -

CMV, cytomegalovirus; EBV, Epstein–Barr virus; HSV, herpes simplex virus; OL, off label; VZV, varicella-zoster virus; +, approved; -, unapproved.

Antiviral Chemistry & Chemotherapy 23.3 97 E De Clercq

Figure 12. The bicyclic furanopyrimidine analogue Cf 1743 and its valine ester: exquisitely specific against varicella-zoster virus

O O

N N

O N O N HO H N CH CO O O 2 O CH HC3 CH3

OH OH

Cf 1743 FV-100 (valine ester of Cf 1743)

Figure 13. Helicase-primase inhibitor, effective against both Figure 14. Helicase-primase inhibitor, under development for herpes simplex virus and varicella-zoster virus herpes simplex virus

O H CH N 3 N O O N O N S NH N O S 2 S O N O N O CH3 BAY 57-1293 ASP2151 AIC316 Pritelivir

Development discontinued.

to treat cytomegalovirus infections. It exhibits excellent Tenofovir, a topical microbicide effective against in vitro inhibitory activity against HCMV laboratory HIV, as demonstrated by the CAPRISA study [62], was strains and clinical isolates [56], but no other betaher- recently reported to inhibit HSV-2 replication [63]. How- pesviruses (that is, rat cytomegalovirus, or human her- ever, tenofovir did so within the concentration range of pesvirus type 6 [HHV-6]) or a- or g-herpesviruses [57]. 20–200 mg/ml, whereas adefovir inhibited HSV-2 rep- Initial clinical results point to the successful treatment of lication within a concentration range of 0.5–50 mg/ml, multidrug-resistant human cytomegalovirus (HCMV) and cidofovir did so within the concentration range of disease with AIC246 [58]. 0.2–50 mg/ml [63].

Epilogue Acknowledgements

Anti-HSV agents may indirectly affect HIV-1 virus This review was originally based on the lecture presented titres: through its inhibitory effect on HSV replication, at the Symposium on Viruses and Antiviral Therapy (in valaciclovir may help reducing plasma HIV-1 levels in honour of Hugh J Field), Queens’ College, Cambridge, HIV-1/HSV-2-coinfected individuals [59–61]. UK, 23 March 2012.

98 ©2013 International Medical Press Selective anti-herpesvirus agents

Table 2. New antiviral agents at the horizon: in vitro antiviral activity

Agent Herpes simplex virus Varicella-zoster virus Cytomegalovirus

CMX001 + + + FV-100 - + - BAY 57-1293 (AIC316) + - - Maribavira - - + AIC246 (letermovir) - - + aDiscontinued. +, antiviral activity; -, antiviral inactivity.

Figure 15. Specific inhibitor of human cytomegalovirus Figure 16. Specific inhibitor of human cytomegalovirus

Cl N CH3 O N H N O Cl CH3 HO OH O N CF3

N N HO OH F N O 1263W94 1-(β-L-Ribofuranosyl)-2-isopropylamino-5,6- dichlorobenzimidazole AIC246 Letermovir

Development discontinued.

Disclosure statement 9. Machida H, Watanabe Y, Kano F, Sakata S, Kumagai M, Yamaguchi T. Deglycosylation of antiherpesviral 5-substituted arabinosyluracil derivatives by rat liver The author declares no competing interests. extract and enterobacteria cells. Biochem Pharmacol 1995; 49:763–766. 10. Nakayama H, Kinouchi T, Kataoka K, Akimoto S, References Matsuda Y, Ohnishi Y. Intestinal anaerobic bacteria hydrolyse sorivudine, producing the high blood concentration 1. Prusoff WH. Synthesis and biological activities of of 5-(E)-(2-bromovinyl)uracil that increases the level and iododeoxyuridine, an analog of thymidine. Biochim Biophys toxicity of 5-fluorouracil. Pharmacogenetics 1997; 7:35–43. Acta 1959; 32:295–296. 11. Diasio RB. Sorivudine and 5-fluorouracil; a clinically 2. Kaufman HE. Clinical cure of by significant drug-drug interaction due to inhibition of 5-iodo-2′-deoxyuridine. Proc Soc Exp Biol Med 1962; dihydropyrimidine dehydrogenase. Br J Clin Pharmacol 109:251–252. 1998; 46:1–4. 3. Kaufman HE, Nesburn AB, Maloney ED. IDU therapy of 12. Whitley RJ, Ch’ien LT, Dolin R, Galasso GJ, Alford CA, herpes simplex. Arch Ophthalmol 1962; 67:583–591. Jr. arabinoside therapy of herpes zoster in the immunosuppressed. NIAID collaborative antiviral study. 4. Kaufman HE, Martola E-L, Dohlman C. Use of 5-iodo-2′- N Engl J Med 1976; 294:1193–1199. deoxyuridine (IDU) in treatment of herpes simplex keratitis. Arch Ophthalmol 1962; 68:235–239. 13. Whitley RJ, Soong SJ, Dolin R, Galasso GJ, Ch’ien LT, Alford CA. Adenine arabinoside therapy of biopsy-proved 5. Kaufman HE, Heidelberger C. Therapeutic antiviral action herpes simplex encephalitis – National Institute of Allergy of 5-trifluoromethyl-2′-deoxyuridine in herpes simplex and Infectious Diseases collaborative antiviral study. N Engl keratitis. Science 1964; 145:585–586. J Med 1977; 297:289–294. 6. De Clercq E, Descamps J, De Somer P, Barr PJ, Jones AS, 14. Whitley RJ, Arvin A, Prober C, et al. A controlled trial Walker RT. (E)-5-(2-Bromovinyl)-2′-deoxyuridine: a potent comparing vidarabine with acyclovir in neonatal herpes and selective anti-herpes agent. Proc Natl Acad Sci U S A simplex virus infection. N Engl J Med 1991; 324:444–449. 1979; 76:2947–2951. 15. Field HJ, De Clercq E. Antiviral drugs – a short history of 7. De Clercq E, Degreef H, Wildiers J, et al. Oral (E)-5-(2- their discovery and development. Microbiol Today 2004; bromovinyl)-2′-deoxyuridine in severe herpes zoster. BMJ 31:58–61. 1980; 281:1178. 16. Elion GB, Furman PA, Fyfe JA, de Miranda P, Beauchamp 8. Shigeta S, Yokota T, Iwabuchi T, et al. Comparative efficacy L, Schaeffer HJ. Selectivity of action of an antiherpetic of antiherpes drugs against various strains of varicella- agent, 9-(2-hydroxyethoxymethyl) guanine. Proc Natl Acad zoster virus. J Infect Dis 1983; 147:576–584. Sci U S A 1977; 74:5716–5720.

Antiviral Chemistry & Chemotherapy 23.3 99 E De Clercq

17. Schaeffer HJ, Beauchamp L, de Miranda P, Elion GB, 37. Andrei G, Snoeck R. Emerging drugs for varicella-zoster virus Bauer DJ, Collins P. 9-(2-Hydroxyethoxymethyl) guanine infections. Expert Opin Emerg Drugs 2011; 16:507–535. activity against viruses of the herpes group. Nature 1978; 272:583–585. 38. Kleymann G, Fischer R, Betz UA, et al. New helicase- primase inhibitors as drug candidates for the treatment of 18. De Clercq E. Selective antiherpes agents. Trends Pharmacol herpes simplex disease. Nat Med 2002; 8:392–398. Sci 1982; 3:492–495. 39. Betz UA, Fischer R, Kleymann G, Hendrix M, Rübsamen- 19. De Clercq E. Biochemical aspects of the selective antiherpes Waigmann H. Potent in vivo antiviral activity of the herpes activity of nucleoside analogues. Biochem Pharmacol 1984; simplex virus primase-helicase inhibitor BAY 57-1293. 33:2159–2169. Antimicrob Agents Chemother 2002; 46:1766–1772. 20. Field HJ, De Clercq E. Effects of oral treatment with 40. Crute JJ, Grygon CA, Hargrave KD, et al. Herpes simplex acyclovir and bromovinyldeoxyuridine on the establishment virus helicase-primase inhibitors are active in animal models of maintenance of latent herpes simplex virus infection in of human disease. Nat Med 2002; 8:386–391. mice. J Gen Virol 1981; 56:259–265. 41. Field HJ, Biswas S, Mohammad IT. Herpesvirus latency and 21. Field HJ, Darby G. Pathogenicity in mice of strains of herpes therapy – from a veterinary perspective. Antiviral Res 2006; simplex virus which are resistant to acyclovir in vitro and in 71:127–133. vivo. Antimicrob Agents Chemother 1980; 17:209–216. 42. Biswas S, Jennens L, Field HJ. The helicase primase inhibitor, 22. Darby G, Field HJ, Salisbury SA. Altered substrate BAY 57-1293 shows potent therapeutic antiviral activity specificity of herpes simplex virus thymidine kinase confers superior to famciclovir in BALB/c mice infected with herpes acyclovir-resistance. Nature 1981; 289:81–83. simplex virus type 1. Antiviral Res 2007; 75:30–35. 23. Field HJ, Darby G, Wildy P. Isolation and characterization 43. Biswas S, Swift M, Field HJ. High frequency of spontaneous of acyclovir-resistant mutants of herpes simplex virus. J Gen helicase-primase inhibitor (BAY 57-1293) drug-resistant Virol 1980; 49:115–124. variants in certain laboratory isolates of HSV-1. Antivir 24. Colla L, De Clercq E, Busson R, Vanderhaeghe H. Synthesis Chem Chemother 2007; 18:13–23. and antiviral activity of water-soluble esters of acyclovir 44. Biswas S, Jennens L, Field HJ. Single amino acid [9-[(2-hydroxyethoxy)methyl]guanine]. J Med Chem 1983; substitutions in the HSV-1 helicase protein that confer 26:602–604. resistance to the helicase-primase inhibitor BAY 57-1293 25. Beauchamp LM, Dolmatch BL, Schaeffer HJ, et al. are associated with increased or decreased virus growth Modifications on the heterocyclic base of acyclovir: characteristics in tissue culture. Arch Virol 2007; syntheses and antiviral properties. J Med Chem 1985; 152:1489–1500. 28:982–987. 45. Biswas S, Smith C, Field HJ. Detection of HSV-1 variants 26. De Clercq E, Field HJ. Antiviral prodrugs the development highly resistant to the helicase-primase inhibitor BAY of successful prodrug strategies for antiviral chemotherapy. 57-1293 at high frequency in 2 of 10 recent clinical isolates Br J Pharmacol 2006; 147:1–11. of HSV-1. J Antimicrob Chemother 2007; 60:274–279. 27. Abudalu M, Tyring S, Koltun W, Bodsworth N, Hamed 46. Biswas S, Kleymann G, Swift M, et al. A single drug- K. Single-day, patient-initiated famciclovir therapy versus resistance mutation in HSV-1 UL52 primase points to a 3-day valacyclovir regimen for recurrent genital herpes: a difference between two helicase-primase inhibitors in their randomized, double-blind, comparative trial. Clin Infect Dis mode of interaction with the antiviral target. J Antimicrob 2008; 47:651–658. Chemother 2008; 61:1044–1047. 28. Reischig T, Jindra P, Hes O, Svecová M, Klaboch J, Treska V. 47. Biswas S, Tiley LS, Zimmermann H, Birkmann A, Field HJ. Valacyclovir prophylaxis versus preemptive valganciclovir Mutations close to functional motif IV in HSV-1 UL5 helicase therapy to prevent cytomegalovirus disease after renal that confer resistance to HSV helicase-primase inhibitors, transplantation. Am J Transplant 2008; 8:69–77. variously affect virus growth rate and pathogenicity. Antiviral Res 2008; 80:81–85. 29. De Clercq E, Brancale A, Vere Hodge A, Field HJ. Antiviral Chemistry & Chemotherapy’s current antiviral agents 48. Biswas S, Field HJ. Herpes simplex virus helicase-primase FactFile 2006 (1st edition). Antivir Chem Chemother 2006; inhibitors: recent findings from the study of drug resistance 17:113–166. mutations. Antivir Chem Chemother 2008; 19:1–6. 30. Field HJ, De Clercq E. Antiviral Chemistry & 49. Biswas S, Miguel RN, Sukla S, Field HJ. A mutation in Chemotherapy’s current antiviral agents FactFile (2nd helicase motif IV of herpes simplex virus type 1 UL5 that edition): DNA viruses. Antivir Chem Chemother 2008; results in reduced growth in vitro and lower virulence in a 19:51–62. murine infection model is related to the predicted helicase structure. J Gen Virol 2009; 90:1937–1942. 31. De Clercq E, Field HJ. Antiviral Chemistry & Chemotherapy’s current antiviral agents FactFile (2nd edition): RNA viruses. 50. Sukla S, Biswas S, Birkmann A, et al. Effects of therapy Antivir Chem Chemother 2008; 19:63–74. using a helicase-primase inhibitor (HPI) in mice infected with deliberate mixtures of wild-type HSV-1 and an HPI- 32. De Clercq E, Field HJ. Antiviral Chemistry & resistant UL5 mutant. Antiviral Res 2010; 87:67–73. Chemotherapy’s current antiviral agents FactFile (2nd edition): retroviruses. Antivir Chem Chemother 2008; 51. Sukla S, Biswas S, Birkmann A, Lischka P, Zimmermann H, 19:75–105. Field HJ. Mismatch primer-based PCR reveals that helicase- primase inhibitor resistance mutations pre-exist in herpes 33. McGuigan C, Barucki H, Carangio A, et al. Highly potent simplex virus type 1 clinical isolates and are not induced and selective inhibition of varicella-zoster virus by bicyclic during incubation with the inhibitor. J Antimicrob Chemother furopyrimidine nucleosides bearing an aryl side chain. J 2010; 65:1347–1352. Med Chem 2000; 43:4993–4997. 52. Field HJ, Biswas S. Antiviral drug resistance and helicase- 34. Andrei G, Sienaert R, McGuigan C, De Clercq E, Balzarini J, primase inhibitors of herpes simplex virus. Drug Resist Snoeck R. Susceptibilities of several clinical varicella-zoster Updat 2011; 14:45–51. virus (VZV) isolates and drug-resistant VZV strains to bicyclic furano pyrimidine nucleosides. Antimicrob Agents 53. Dropulic LK, Cohen JI. Update on new antivirals under Chemother 2005; 49:1081–1086. development for the treatment of double-stranded DNA virus infections. Clin Pharmacol Ther 2010; 88:610–619. 35. Migliore M. Susceptibilities of several clinical varicella- zoster virus (VZV) isolates and drug-resistant VZV strains 54. Biron KK. Antiviral drugs for cytomegalovirus diseases. to bicyclic furano pyrimidine nucleosides. Antivir Chem Antiviral Res 2006; 71:154–163. Chemother 2010; 20:107–115. 55. Goldner T, Hewlett G, Ettischer N, Ruebsamen-Schaeff H, 36. Pentikis HS, Matson M, Atiee G, et al. Pharmacokinetics Zimmermann H, Lischka P. The novel anticytomegalovirus and safety of FV-100, a novel oral anti-herpes zoster compound AIC246 (letermovir) inhibits human , administered in single and multiple cytomegalovirus replication through a specific antiviral doses to healthy young adult and elderly adult volunteers. mechanism that involves the viral terminase. J Virol 2011; Antimicrob Agents Chemother 2011; 55:2847–2854. 85:10884–10893.

100 ©2013 International Medical Press Selective anti-herpesvirus agents

56. Lischka P, Hewlett G, Wunberg T, et al. In vitro and in 60. Drake AL, Roxby AC, Ongecha-Owuor F, et al. Valacyclovir vivo activities of the novel anticytomegalovirus compound suppressive therapy reduces plasma and breast milk AIC246. Antimicrob Agents Chemother 2010; 54:1290–1297. HIV-1 RNA levels during pregnancy and postpartum: a 57. Marschall M, Stamminger T, Urban A, et al. In vitro randomized trial. J Infect Dis 2012; 205:366–375. evaluation of the activities of the novel anticytomegalovirus 61. Drake AL, Roxby AC, Kiarie J, et al. Infant safety during compound AIC246 (letermovir) against herpesviruses and after maternal valacyclovir therapy in conjunction with and other human pathogenic viruses. Antimicrob Agents antiretroviral HIV-1 prophylaxis in a randomized clinical Chemother 2012; 56:1135–1137. trial. PLoS ONE 2012; 7:e34635. 58. Kaul DR, Stoelben S, Cober E, et al. First report of 62. Abdool Karim Q, Abdool Karim SS, Frohlich JA, et al. successful treatment of multidrug-resistant cytomegalovirus Effectiveness and safety of tenofovir gel, an antiretroviral disease with the novel anti-CMV compound AIC246. Am J microbicide, for the prevention of HIV infection in women. Transplant 2011; 11:1079–1084. Science 2010; 329:1168–1174. 59. Mugwanya K, Baeten JM, Mugo NR, Irungu E, Ngure K, 63. Andrei G, Lisco A, Vanpouille C, et al. Topical tenofovir, a Celum C. High-dose valacyclovir HSV-2 suppression results microbicide effective against HIV, inhibits herpes simplex in greater reduction in plasma HIV-1 levels compared with virus-2 replication. Cell Host Microbe 2011; 10:379–389. standard dose acyclovir among HIV-1/HSV-2 coinfected persons: a randomized, crossover trial. J Infect Dis 2011; 204:1912–1917. Received 4 December 2012; accepted 24 December 2012; published online 23 January 2013

Antiviral Chemistry & Chemotherapy 23.3 101