We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×

Helicase–primase inhibitors for herpes simplex virus: looking to the future of non-nucleoside inhibitors for treating herpes virus infections

    Subhajit Biswas

    Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 0ES, UK

    School of Life Sciences, University of Lincoln, LN6 7TS, UK

    ,
    Soumi Sukla

    Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 0ES, UK

    School of Life Sciences, University of Lincoln, LN6 7TS, UK

    &
    Hugh J Field

    * Author for correspondence

    Queens’ College, University of Cambridge, CB3 9ET, UK.

    Published Online:https://doi.org/10.4155/fmc.13.192

    Helicase–primase inhibitors (HPIs) are the first new family of potent herpes virus (herpes simplex and varicella-zoster virus) inhibitors to go beyond the preliminary stages of investigation since the emergence of the original nucleoside analog inhibitors. To consider the clinical future of HPIs, this review puts the exciting new findings with two HPIs, amenamevir and pritelivir, into the historical context of antiviral development for the prevention and treatment of herpes simplex virus over the last century and, on this basis, the authors speculate on the potential evolution of these and other non-nucleoside inhibitors in the future.

    Papers of special note have been highlighted as: ▪ of interest ▪▪ of considerable interest

    References

    • Goodpasture EW, Teague O. Experimental production of herpetic lesions in organs and tissues of the rabbit. J. Med. Res.44,121–138 (1923).▪ Describes one of the first laboratory animal models for herpes simplex virus (HSV); the prototype for hundreds of studies that follow and the foundations for antiviral testing in animals.
    • Cutting WC, Dreisbach RH, Halpern RM et al. Chemotherapy of virus infections. J. Immunol.57,379–390 (1947).
    • Prusoff WH. Synthesis and biological activities of iododeoxyuridine, an analog of thymidine. Biochim. Biophys. Acta32,295–296 (1959).
    • Field HJ, De Clercq E. Antiviral drugs: a short history of their discovery and development. Microbiol. Today31,58–61 (2004).▪ Charts the historical sequence of the major discoveries in antiviral research.
    • Kaufman HE, Maloney ED. IDU and cytosine arabinoside in experimental herpetic keratitis. Arch. Ophthalmol.69,626–629 (1963).
    • Kaufman HE, Heidelberger C. Therapeutic antiviral action of 5-trifluormethyl-2´-deoxyuridine in herpes simplex keratitis. Science145,585–586 (1964).
    • De Clercq E, Field HJ. Antiviral prodrugs – the development of successful prodrug strategies for antiviral chemotherapy. Brit. J. Pharmacol.147,1–11 (2006).
    • Field HJ, Vere Hodge RA. Recent developments in anti-herpesvirus drugs. Brit. Med. Bul.106,213–249 (2013).
    • Oberg B. Inhibitors of Virus-Specific Enzymes in Problems of Antiviral Therapy. Stuart-Harris CH, Oxford J (Eds). Academic Press, London, UK, 35–69 (1983).
    • 10  Brockman RW, Sidwell RW, Arnett G, Shaddix S. Heterocyclic thyosemicarbazones: correlation between structure, inhibition of ribonucleotide reductase, and inhibition of DNA viruses. Proc. Soc. Exp. Biol. Med.133,609–614 (1970).
    • 11  Cheng YC, Nakayama K, Derse D et al.Herpes Virus Specific Enzymes: Properties, Physiological Roles and Pharmacological Implications in Herpesviruses, Clinical, Pharmacological and Basic Aspects. Shiota H, Cheng Y-C, Prusoff WH (Eds). Excerpta Medica International Congress Series, Amsterdam, The Netherlands, 47–56 (1982).
    • 12  Watkins AM, Dunford PJ, Moffatt AM et al. Inhibition of virus-encoded thymidine kinase suppresses herpes simplex virus replication in vitro and in vivo.Antivir. Chem. Chemother.9,9–18 (1998).
    • 13  Bucknall RA, Moores H, Simms R, Hesp B. Antiviral effects of aphidicolin, a new antibiotic produced by Cephalosporium aphidocola.Antimicrob. Agents Chemother.4,294–298 (1973).
    • 14  Coen DM, Furman PA, Aschman DP, Schaffer PA. Mutations in the herpes simplex virus DNA polymerase gene conferring hypersensitivity to aphidicolin. Nucleic Acids Res.11,5287–5297 (1983).
    • 15  Helgstrand E, Eriksson B, Johansson NG et al. Trisodium phosphonoformate, a new antiviral compound. Science201,819–821 (1978).
    • 16  Wagstaff AJ, Bryson HM. Foscarnet. A reappraisal of its antiviral activity, pharmacokinetic properties and therapeutic use in immunocompromised patients with viral infections. Drugs48,199–226 (1994).
    • 17  Lischka P, Hewlett G, Wunberg T et al.In vitro and in vivo activities of the novel anticytomegalovirus compound AiC 246. Antimicrob. Agents Chemother.54,1290–1297 (2010).▪ First definitive account of letermovir, describing an exciting advance in cytomegalovirus chemotherapy.
    • 18  Vere Hodge A. Scientific report: highlights of the 25th ICAR 16–19 April, 2012, Sapporo, Japan. Antivir. Chem. Chemother.23,19–33, (2012).
    • 19  Spector FC, Liang L, Giordano H, Sivaraja M, Peterson MG. Inhibition of herpes simplex virus replication by a 2-amino thiazole via interactions with the helicase component of the UL5-UL8-UL52 complex. J. Virol.72,6979–6987 (1998).
    • 20  Field HJ, Mickleburgh I. The helicase-primase complex as a target for efective herpesvirus antivirals in DNA helicases and DNA motor proteins. In: DNA Helicases and DNA Motor Proteins. Spies M (Ed.). Springer, Berlin, Germany, 145–159 (2013).
    • 21  Birkmann A, Hewlett G, Rubsamen-Schaeff H, Zimmermann H. Helicase primase inhibitors as the potential next gerneration of highly active drugs against herpes simplex viruses. Future Virol.6,1199–1209 (2011).
    • 22  Biswas S, Field HJ. Helicase–Primase Inhibitors: A New Approach to Combat Herpes Simplex Virus and Varicella Zoster Virus in Antiviral Drug Strategies. De Clercq E (Ed.). Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim, Germany 129–146 (2011).▪ An exhaustive chapter on helicase–primase inhibitors (HPI) and contains a detailed table of HPI-resistant mutations, a must-read in conjunction with this review.
    • 23  Chono K, Katsumata K, Kontani T et al. ASP2151, a novel helicase–primase inhibitor, possesses antiviral activity against varicella-zoster virus and herpes simplex virus types 1 and 2. J. Antimicrob. Chemother.65,1733–1741 (2010).▪▪ First report on a multispectrum helicase–primase inhibitors with potent antiviral activities against HSV and varicella-zoster virus.
    • 24  Kleymann G, Fischer R, Betz UA et al. New helicase–primase inhibitors as drug candidates for the treatment of herpes simplex disease. Nat. Med.8,392–398 (2002).▪▪ First report on N-methyl-N-(4-methyl-5-sulfamoyl-1,3-thiazol-2-yl)-2-(4-pyridin-2-yl-phenyl)acetamide (AIC316 or pritelivir) as a potent HPI against HSV.
    • 25  Crute JJ, Grygon CA, Hargrave KD et al. Herpes simplex virus helicase-primase inhibitors are active in animal models of human disease. Nat. Med.8,386–391 (2002).▪▪ A must-read paper on the BILS series of HPIs with potent in vitro and in vivo anti-HSV activities.
    • 26  Duan J, Liuzzi M, Paris W et al. Oral bioavailability and in vivo efficacy of the helicase-primase inhibitor BILS 45 BS against acyclovir-resistant herpes simplex virus type 1. Antimicrob. Agents Chemother.47,1798–1804 (2003).
    • 27  Liuzzi M, Kibler P, Bousquet et al. Isolation and characterization of herpes simplex virus type 1 resistant to inothiazolylphenyl-based inhibitors of the viral helicase-primase. Antiviral Res.64,161–170 (2004).
    • 28  Biswas S, Jennens L, Field HJ. The helicase primase inhibitor, BAY57–1293 shows potent therapeutic antiviral activity superior to famciclovir in BALB/c mice infected with herpes simplex virus type 1. Antiviral Res.75,30–35 (2007).
    • 29  Betz UAK, Fischer R, Kleymann G, Hendrix M, Rübsamen-Waigmann H. Potent in vivo antiviral activity of the herpes simplex virus primase-helicase inhibitor BAY57–1293. Antimicrob. Agents Chemother.4,1766–1772 (2002).
    • 30  Baumeister J, Fischer R, Eckenberg P, Henninger K, Ruebsamen-Waigmann H, Kleymann G. Superior efficacy of helicase-primase inhibitor BAY 57–1293 for herpes infection and latency in the guinea pig model of human genital herpes disease. Antivir. Chem. Chemother.18,35–48 (2007).▪ Only paper, to date, that presents preliminary data on the potential of pritelivir in reducing establishment of HSV latency and preventing virus reactivation in guinea pig model.
    • 31  Sasaki S, Miyazaki D, Haruki T et al. Efficacy of herpes virus helicase-primase inhibitor, ASP2151, for treating herpes simplex keratitis in mouse model. Br. J. Ophthalmol.97,498–503 (2013).
    • 32  Chono K, Katsumata K, Kontani T, Shiraki K, Suzuki H. Characterization of virus strains resistant to the herpes virus helicase-primase inhibitor ASP2151 (Amenamevir). Biochem. Pharmacol.84,459–467 (2012).
    • 33  Chen Y, Bai P, Mackay S et al. Herpes simplex virus type 1 helicase-primase: DNA binding and consequent protein oligomerization and primase activation. J. Virol.85,968–978 (2011).
    • 34  Biswas S, Kleymann G, Swift M et al. A single drug resistance mutation in HSV-1 UL52 primase points to a difference between two helicase-primase inhibitors in their mode of interaction with the antiviral target. J. Antimicrob. Chemother.61,1044–1047 (2008).▪ Describes how information on drug-resistance mutations provides vital clues to understanding differences among HPIs and general mechanism of drug action.
    • 35  Field HJ, Mickelburgh I, Huang M-L et al. Sensitivity of clinical isolates to helicase primase inhibitors and no detection of resistance mutations above background frequency. Presented at: 25th International Conference on Antiviral Research. Sapporo, Japan, 16–19 April 2012.
    • 36  Biswas S, Jennens L, Field HJ. Single amino acid substitutions in the HSV-1 helicase protein that confer resistance to the helicase-primase inhibitor BAY 57–1293 are associated with increased or decreased virus growth characteristics in tissue culture. Arch. Virol.152,1489–1500 (2007).
    • 37  Biswas S, Field HJ. Herpes simplex virus helicase–primase inhibitors: recent findings from the study of drug resistance mutations. Antivir. Chem. Chemother.19,1–6 (2008).
    • 38  Field HJ, Biswas S. Antiviral drug resistance and helicase-primase inhibitors of herpes simplex virus. Drug Resist. Update14,45–51 (2011).
    • 39  Zhu L, Weller SK. The six conserved helicase motifs of the UL5 gene product, a component of the herpes simplex virus type 1 helicase-primase, are essential for its function. J. Virol.66,469–479 (1992).
    • 40  Biswas S, Tiley LS, Zimmermann H, Birkmann A, Field HJ. Mutations close to functional motif IV in HSV-1 UL5 helicase that confer resistance to HSV helicase–primase inhibitors, variously affect virus growth rate and pathogenicity. Antiviral Res.80,81–85 (2008).
    • 41  Biswas S, Miguel RN, Sukla S, Field HJ. A mutation in helicase motif IV of herpes simplex virus type 1 UL5 that results in reduced growth in vitro and lower virulence in a murine infection model is related to the predicted helicase structure. J. Gen. Virol.90,1937–1942 (2009).
    • 42  Biswas S, Swift M, Field HJ. High frequency of spontaneous helicase–primase inhibitor (BAY 57–1293) drug-resistant variants in certain laboratory isolates of HSV-1. Antivir. Chem. Chemother.18,13–23 (2007).
    • 43  Biswas S, Smith C, Field HJ. Detection of HSV-1 variants highly resistant to the helicase-primase inhibitor BAY 57–1293 at high frequency in two of ten recent clinical isolates of HSV-1. J. Antimicrob. Chemother.60,274–279 (2007).
    • 44  Sukla S, Biswas S, Birkmann A, Lischka P, Zimmermann H, Field H. Mismatch primer-based PCR reveals that helicase-primase inhibitor resistance mutations pre-exist in herpes simplex virus type 1 clinical isolates and are not induced during incubation with the inhibitor. J. Antimicrob. Chemother.65,1347–1352 (2010).▪ Evidence that certain HPI-resistance mutations pre-exist at high frequency in parental virus populations and were not induced by pritelivir.
    • 45  Biswas S, Sukla S, Paulsen D et al. Efficacious treatment of a helicase-primase inhibitor resistant herpes simplex virus-1 mutant with AIC316 in a murine infection model. Presented at: 36th Annual International Herpesvirus Workshop. Gdansk, Poland, 24–28 July 2011.
    • 46  Sukla S, Biswas S, Birkmann A et al. Effects of therapy using a helicase-primase inhibitor (HPI) in mice infected with deliberate mixtures of wild-type HSV-1 and an HPI-resistant UL5 mutant. Antiviral Res.87,67–73 (2010).
    • 47  James SH, Kimberlin DW, Whitley RJ. Antiviral therapy for herpesvirus central nervous system infections: neonatal herpes simplex virus infection, herpes simplex encephalitis, and congenital cytomegalovirus infection. Antiviral Res.83,207–213 (2009).
    • 48  Spruance SL, Bodsworth N, Resnick H et al. Single-dose, patient-initiated famciclovir: a randomized, double-blind, placebo-controlled trial for episodic treatment of herpes labialis. J. Am. Acad. Dermatol.55,47–53 (2006).
    • 49  Harmenberg J, Oberg B, Spruance S. Prevention of ulcerative lesions by episodic treatment or recurrent herpes labialis: a literature review. Acta Dermato-Venereologica90,122–130 (2010).
    • 50  Harmenberg J. Intracellular pools of thymidine reduce the antiviral activity of acyclovir. Intervirology20,48–51 (1983).
    • 51  Field HJ, Huang M-L, Lay EM, Mickleburgh I, Zimmermann H, Birkmann A. Baseline sensitivity of HSV-1 and HSV-2 clinical isolates and defined acyclovir-resistant strains to the helicase–primase inhibitor pritelivir. Antiviral Res.100,297–299 (2013).
    • 52  Dodson MS, Lehman IR, Association of DNA helicase and primase activities with a subassembly of the herpes simplex virus 1 helicase-primase composed of the UL5 and UL52 gene products. Proc. Natl Acad. Sci. USA88,1105–1109 (1991).
    • 53  Cavanaugh, NA, Ramirez-Aguilar KA, Urban M, Kuchta RD. Herpes simplex virus-1 helicase–primase: roles of each subunit in DNA binding and phosphodiester bond formation. Biochemistry48,10199–10207 (2009).
    • 54  Chen Y, Carrington-Lawrence SD, Bai P, Weller SK. Mutations in the putative zinc-binding motif of UL52 demonstrate a complex interdependence between the UL5 and UL52 subunits of the human herpes simplex virus type 1 helicase/primase complex. J. Virol.79,9088–9096 (2005).
    • 55  Chen Y, Livingston CM, Carrington-Lawrence SD, Bai P, Weller SK. A mutation in the human herpes simplex virus type I UL52 zinc finger motif results in defective primase activity but can recruit viral polymerase and support viral replication efficiently. J. Virol.81,8742–8751 (2007).
    • 101  AiCuris successfully completed Phase I for novel herpes compound. www.aicuris.com/index.php/fuseaction/download/lrn_file/20091104-press-release-hsv-phase-i-e-final.pdf (Accessed 26 June 2013)
    • 102  AiCuris: no resistant herpes simplex virus (HSV) observed in Phase II trial with herpes drug AIC316. www.drugs.com/clinical_trials/aicuris-no-resistant-herpes-simplex-virus-hsv-observed-phase-ii-trial-herpes-aic316-12297.html (Accessed 26 June 2013)
    • 103  AiCuris surpasses 50% enrollment milestone in Phase II. Superiority trial for herpes drug pritelivir. www.aicuris.com/index.php/fuseaction/download/lrn_file/2013-01-29_pr_50percent-recruitment-aic316-press-release.pdf (Accessed 26 June 2013)
    • 104  Clinical trials. http://clinicaltrials.gov/ct2/show/NCT01658826?term=aic316&rank=1 (Accessed 26 June 2013)