164
Views
1
CrossRef citations to date
0
Altmetric
Research Article

Design, Synthesis and Antiviral Evaluation of 2′-C-Methyl Branched Guanosine Pronucleotides: The Discovery of IDX184, a Potent Liver-Targeted HCV Polymerase Inhibitor

, , , , , , , , , , , , & show all
Pages 1675-1700 | Published online: 01 Oct 2015

References

  • Ashfaq UA , JavedT, RehmanS, NawazZ, RiazuddinS. An overview of HCV molecular biology, replication and immune response. Virol. J.8, 161 (2011).
  • Scheel TKH , RiceCM. Understanding the hepatitis C virus life cycle paves the way for highly effective therapies. Nat. Med.19 (7), 837–849 (2013).
  • Thomas DL . Global control of hepatitis C: where challenge meets opportunity. Nat. Med.19 (7), 850–858 (2013).
  • Lim TR , TanBH, MutimerDJ. Evolution and emergence of a new era of antiviral treatment for chronic hepatitis C infection. Int. J. Antimicrob. Agents43 (1), 17–25 (2014).
  • Au JS , PockrosPJ. Novel therapeutic approaches for hepatitis C. Clin. Pharmacol. Ther.95 (1), 78–88 (2014).
  • Conteduca V , SansonnoD, RussiSet al. Therapy of chronic hepatitis C virus infection in the era of direct-acting and host-targeting antiviral agents. J. Infect.68 (1), 1–20 (2014).
  • Hayes CN , ChayamaK. Emerging treatments for chronic hepatitis C. J. Formos. Med. Assoc.114 (3), 204–215 (2015).
  • Gerber L , WelzelTM, ZeuzemS. New therapeutic strategies in HCV: polymerase inhibitors. Liver Int.33 (Suppl 1), 85–92 (2013).
  • Soriano V , VispoE, de MendozaCet al. Hepatitis C therapy with HCV NS5B polymerase inhibitors. Expert Opin. Pharmacother.14 (9), 1161–1170 (2013).
  • Pockros PJ . Nucleoside/nucleotide analogue polymerase inhibitors in development. Clin. Liver Disease17 (1), 105–110 (2013).
  • Deval J , HuberM. Hepatitis C virus and its inhibitors: the polymerase as a target for nucleoside and nucleotide analogs. In : Cancer-Causing Viruses and their Inhibitors. GuptaSP ( Ed.). CRC Press, 41–84 (2014).
  • Deval J , SymonsJA, BeigelmanL. Inhibition of viral RNA polymerases by nucleoside and nucleotide analogs: therapeutic applications against positive-strand RNA viruses beyond hepatitis C virus. Curr. Opin. Virol.9, 1–7 (2014).
  • Coats SJ , Garnier-AmblardEC, AmblardFet al. Chutes and ladders in hepatitis C nucleoside drug development. Antivir. Res.102, 119–147 (2014).
  • De Clercq E . Sofosbuvir in the current context of hepatitis C Treatment. J. Symptoms Signs.3 (3), 126–131 (2014).
  • Kayali Z , SchmidtWN. Finally sofosbuvir: an oral anti-HCV drug with wide performance capability. Pharmacogenomics Person. Med.7, 387–398 (2014).
  • Sofia MJ . Beyond sofosbuvir: what opportunity exists for a better nucleoside/nucleotide to treat hepatitis C?Antivir. Res.107, 119–124 (July 2014).
  • Sommadossi JP , LacollaP : WO2001090121 (2001).
  • Sommadossi JP , LacollaP : WO2001092282 (2001).
  • Benzaria S , BardiotD, BouissetTet al. 2′-C-Methyl branched pyrimidine ribonucleoside analogues: potent inhibitors of RNA virus replication. Antivir. Chem. Chemother.18 (4), 225–242 (2007).
  • Pierra C , BenzariaS, AmadorAet al. NM283, an efficient prodrug of the potent anti-HCV agent 2′-C-methylcytidine. Nucleosides Nucleotides Nucleic Acids24 (5–7), 767–770 (2005).
  • Pierra C , AmadorA, BenzariaSet al. Synthesis and pharmacokinetics of valopicitabine (NM283), an efficient prodrug of the potent anti-HCV agent 2′-C-methylcytidine. J. Med. Chem.49 (22), 6614–6620 (2006).
  • Sommadossi JP , GosselinG, PierraCet al. O2008082602 (2008).
  • Sommadossi JP , GosselinG, PierraCet al. WO2008082601 (2008).
  • Cretton-Scott E , Hernandez-SantiagoB, LarssonMet al. WO2010014134 (2010).
  • Buckwold VE , BeerBE, DonisRO. Bovine viral diarrhea virus as a surrogate model of hepatitis C virus for the evaluation of antiviral agents. Antivir. Res.60 (1), 1–15 (2003).
  • Migliaccio G , TomassiniJE, CarrollSSet al. Characterization of resistance to non-obligate chain-terminating ribonucleoside analogs that inhibit hepatitis C virus replication in vitro. J. Biol. Chem.278 (49), 49164–49170 (2003).
  • Eldrup AB , AllersonCR, BennettCFet al. Structure–activity relationship of purine ribonucleosides for inhibition of hepatitis C virus RNA-dependent RNA polymerase. J. Med. Chem.47 (9), 2283–2295 (2004).
  • Eldrup AB , PrhavcM, BrooksJet al. Structure–activity relationship of heterobase-modified 2′-C-methyl ribonucleosides as inhibitors of hepatitis C virus RNA replication. J. Med. Chem.47 (21), 5284–5297 (2004).
  • Clark JL , MasonJC, HolleckerLet al. Synthesis and antiviral activity of 2′-deoxy-2′-fluoro-2′-C-methyl purine nucleosides as inhibitors of hepatitis C virus RNA replication. Bioorg. Med. Chem. Lett.16 (6), 1712–1715 (2006).
  • McGuigan C , PerroneP, MadelaKet al. The phosphoramidate ProTide approach greatly enhances the activity of β-2′-C-methylguanosine against hepatitis C virus. Bioorg. Med. Chem. Lett.19 (15), 4316–4320 (2009).
  • Amblard F , FromentinE, DetorioMet al. Synthesis, antiviral activity, and stability of nucleoside analogs containing tricyclic bases. Eur. J. Med. Chem.44 (10), 3845–3851 (2009).
  • Olsen DB , EldrupAB, BartholomewLet al. A 7-deaza-adenosine analog is a potent and selective inhibitor of hepatitis C virus replication with excellent pharmacokinetic properties. Antimicrob. Agents Chemother.48 (10), 3944–3953 (2004).
  • Ray AS , HitchcockMJM. Metabolism of antiviral nucleosides and nucleotides. In : Antiviral Research. Strategy in Antiviral Drug Discovery. LaFeminaRL ( Ed.). ASM Press, Washington, DC, USA, 301–315 (2009).
  • Pradere U , Garnier-AmblardEC, CoatsSJet al. Synthesis of nucleoside phosphate and phosphonate prodrugs. Chem. Rev.114 (18), 9154–9218 (2014).
  • Hurwiotz SJ , SchinaziRF. Prodrug strategies for improved efficacy of nucleoside antiviral inhibitors. Curr. Opin. HIV AIDS.8 (6), 555–564 (2013).
  • Sofia MJ . Nucleotide prodrugs for the treatment of HCV infection. In : Advances in Pharmacolology Antiviral Agents. De ClercqE ( Ed.). Academic Press, Burlington, MA, USA, 39–73 (2013).
  • Peyrottes S , PerigaudC. Mononucleotide prodrug synthetic strategies. In : Chemical Synthesis of Nucleoside Analogues. MerinoP ( Ed.). John Wiley & Sons, Hoboken, NJ, USA, 229–261 (2013).
  • Ray AS , HostetlerKY. Application of kinase bypass strategies to nucleoside antivirals. Antivir. Res.92 (2), 277–291 (2011).
  • Peterson LW , McKennaCE. Prodrug approaches to improving the oral absorption of antiviral nucleotide analogues. Expert Opin. Drug Deliv.6 (4), 405–420 (2009).
  • Hecker SJ , ErionMD. Prodrugs of phosphates and phosphonates. J. Med. Chem.51 (8), 2328–2345 (2008).
  • Poijarvi-Virta P , LonnbergH. Prodrug approaches of nucleotides and oligonucleotides. Curr. Med. Chem.13 (28), 3441–3465 (2006).
  • Khan SR , NowakB, PlunkettWet al. Bis(pivaloyloxymethyl) thymidine 5′-phosphate is a cell membrane-permeable precursor of thymidine 5′-phosphate in thymidine kinase deficient CCRF CEM cells. Biochem. Pharmacol.69 (9), 1307–1313 (2005).
  • Mackman RL , ZhangL, PrasadVet al. Synthesis, anti-HIV activity, and resistance profile of thymidine phosphonomethoxy nucleosides and their bis-isopropyloxymethylcarbonyl (bisPOC) prodrugs. Bioorg. Med. Chem.15 (16), 5519–5528 (2007).
  • Gosselin G , GirardetJL, PerigaudCet al. New insights regarding the potential of the pronucleotide approach in antiviral chemotherapy. Acta. Biochim. Pol.43 (1), 195–208 (1996).
  • Perigaud C , GirardetJL, GosselinGet al. Comments on nucleotide delivery forms. Adv. Antivir. Drug Des.2, 147–172 (1996).
  • Perigaud C , GosselinG, LefebvreIet al. Rational design for cytosolic delivery of nucleoside monophosphates: “SATE” and “DTE” as enzyme-labile transient phosphate protecting groups. Bioorg. Med. Chem. Lett.3 (12), 2521–2526 (1993).
  • Puech F , GosselinG, LefebvreIet al. Intracellular delivery of nucleoside monophosphates through a reductase-mediated activation process. Antivir. Res.22 (2–3), 155–174 (1993).
  • Peyrottes S , EgronD, LefebvreIet al. SATE pronucleotide approaches: an overview. Mini Rev. Med. Chem.4 (4), 395–408 (2004).
  • Morales EHR , BalzariniJ, MeierC. Diastereoselective synthesis of cycloSaligenyl-nucleosyl-phosphotriesters. Chem. A Eur. J.17 (5), 1649–1659 (2011).
  • Meier C , BalzariniJ. Application of the cycloSal-prodrug approach for improving the biological potential of phosphorylated biomolecules. Antivir. Res.71 (2–3), 282–292 (2006).
  • Meier C , MeerbachA, BalzariniJ. Cyclosal-pronucleotides - Development of first and second generation chemical Trojan horses for antiviral chemotherapy. Front. Biosci.9 (1), 873–890 (2004).
  • Meier C . CycloSal-pronucleotides-design of the concept, chemistry, and antiviral activity. Adv. Antivir. Drug Des.4, 147–213 (2004).
  • Bookser BC , RaffaeleNB, ReddyKRet al. Synthesis of 3′-amino-3′-deoxyguanosine and 3′-amino-3′-deoxyxyloguanosine monophosphate hepdirect prodrugs from guanosine. Nucleosides Nucleotides Nucleic Acids28 (10), 969–986 (2009).
  • Erion MD , BulloughDA, LinCCet al. HepDirect prodrugs for targeting nucleotide-based antiviral drugs to the liver. Curr. Opin. Investig. Drugs.7 (2), 109–117 (2006).
  • Vernachio JH , BleimanB, BryantKDet al. INX-08189, a phosphoramidate prodrug of 6-O-methyl-2′-C-methyl guanosine, is a potent inhibitor of hepatitis C virus replication with excellent pharmacokinetic and pharmacodynamic properties. Antimicrob. Agents. Chemother.55 (5), 1843–1851 (2011).
  • Mehellou Y , BalzariniJ, McGuiganC. Aryloxy phosphoramidate triesters: a technology for delivering monophosphorylated nucleosides and sugars into cells. Chem. Med. Chem.4 (11), 1779–1791 (2009).
  • Cahard D , McGuiganC, BalzariniJ. Aryloxy phosphoramidate triesters as protides. Mini Rev. Med. Chem.4 (4), 371–381 (2004).
  • Cheng J , ZhouX, ChouTFet al. Identification of the amino acid-AZT-phosphoramidase by affinity T7 phage display selection. Bioorg. Med. Chem. Lett.19 (22), 6379–6381 (2009).
  • Chou TF , BaraniakJ, KaczmarekRet al. Phosphoramidate pronucleotides: A comparison of the phosphoramidase substrate specificity of human and Escherichia coli histidine triad nucleotide binding proteins. Mol. Pharm.4 (2), 208–217 (2007).
  • Drontle DP , WagnerCR. Designing a pronucleotide stratagem: lessons from amino acid phosphoramidates of anticancer and antiviral pyrimidines. Mini Rev. Med. Chem.4 (4), 409–419 (2004).
  • Egron D , ImbachJL, GosselinGet al. S-Acyl-2-thioethyl phosphoramidate diester derivatives as mono-nucleotide prodrugs. J. Med. Chem.46 (21), 4564–4571 (2003).
  • Egron D , PerigaudC, GosselinGet al. Synthesis and study of a new series of phosphoramidate derivatives as mononucleotide prodrugs. Nucleosides Nucleotides Nucleic Acids20 (4–7), 751–754 (2001).
  • Beltran T , EgronD, PomponAet al. Rational design of a new series of pronucleotide. Bioorg. Med. Chem. Lett.11 (13), 1775–1777 (2001).
  • Satoh T . Role of carboxylesterases in xenobiotic metabolism. In : Reviews in Biochemical Toxicology. HodgsonE, BendJR, PhilpotRM ( Eds.). Elsevier, North Holland, 155–81 (1987).
  • Jochum A , SchliengerN, EgronDet al. Biolabile constructs for pronucleotide design. J. Organomet. Chem.690 (10), 2614–2625 (2005).
  • Shafiee M , DefermeS, VillardA-Let al. New bis(SATE) prodrug of AZT 5′-monophosphate: in vitro anti-HIV activity, stability, and potential oral absorption. J. Pharm. Sci.90 (4), 448–463 (2001).
  • Atherton FR , OpenshawHT, ToddAR. Studies on phosphorylation. Part II. The reaction of dialkyl phosphites with polyhalogen compounds in presence of bases. A new method for the phosphorylation of amines. J. Chem. Soc.660–663 (1945).
  • Atherton FR , ToddAR. Studies on phosphorylation. Part III. Further observations on the reaction of phosphites with polyhalogen compounds in the presence of bases and its application to the phosphorylation of alcohols. J. Chem. Soc.674–678 (1947).
  • Perigaud C , PeyrottesS, DumontetC : WO2009053654 (2009).
  • Mayes BA , ArumugasamyJ, BalogluEet al. Synthesis of a nucleoside phosphoramidate prodrug inhibitor of HCV NS5B polymerase: phenylboronate as a transient protecting group. Org. Process Res. Dev.18 (6), 717–724 (2014).
  • McGuigan C , PerroneP : WO2008062206 (2008).
  • Maccoss M , OlsenDB : WO2006012078 (2006).
  • McGuigan C , PathiranaRN, MahmoodNet al. Aryl phosphates derivatives of AZT retain activity against HIV1 in cell lines which are resistant to the action of AZT. Antivir. Res.17 (4), 311–321 (1992).
  • Iyer VV , GriesgraberGW, RadmerMRet al. Synthesis, in vitro anti-breast cancer activity, and intracellular decomposition of amino acid methyl ester and alkyl amide phosphoramidate monoesters of 3′-azido-3′-deoxythymidine (AZT). J. Med. Chem.43 (11), 2266–2274 (2000).
  • Valette G , PomponA, GirardetJLet al. Decomposition pathways and in vitro HIV inhibitory effects of isoddA pronucleotides: towards a rational approach for intracellular delivery of nucleoside 5′-monophosphates. J. Med. Chem.39 (10), 1981–1990 (1996).
  • Roman CA , WasserthalP, BalzariniJet al. Diastereoselective synthesis of (aryloxy)phosphoramidate prodrugs. Eur. J. Org. Chem. (25), 4899–4909 (2011).
  • Murakami E , TolstykhT, BaoHet al. Mechanism of activation of PSI-7851 and its diastereoisomer PSI-7977. J. Biol. Chem.285 (45), 34337–34347 (2010).
  • Troev K , TashevE, BorisovG. On the interaction between dialkyl phosphites and amine hydrochlorides. Phosphorus Sulfur. Silicon. Relat. Elem.11 (3), 363–367 (1981).
  • Surleraux D , DoussonCB, GosselinGet al. US20130064794 (2013).
  • Dhareshwar SS , StellaVJ. Your prodrug releases formaldehyde: should you be concerned? No!J. Pharm. Sci.97 (10), 4184–4193 (2008).
  • Jones RJ , BischofbergerN. Minireview: nucleotide prodrugs. Antivir. Res.27 (1–2), 1–17 (1995).
  • Cretton-Scott E , PerigaudC, PeyrottesSet al. In vitro antiviral activity and pharmacology of IDX184, a novel and potent inhibitor of HCV replication. J. Hepatol.48 (Suppl. 2), S220 (2008).
  • Surleraux D , DoussonCB, ParsyCC : WO2013056046 (2013).
  • Standring DN , LanfordR, Cretton-ScottEet al. Potent antiviral activity of second generation nucleoside inhibitors, IDX102 and IDX184, in HCV-infected chimpanzees. J. Hepatol.48 (Suppl. 2), S30 (2008).
  • Standring DN , LanfordR, LiBet al. Antiviral activity of the liver-targeted nucleotide HCV polymerase inhibitor IDX184 correlates with trough serum levels of the nucleoside metabolite in HCV-infected chimpanzees. J. Hepatol.50 (Suppl. 1), S37 (2009).
  • Standring DN , PerigaudC, PeyrottesSet al. Pharmacology of IDX184, a liver-targeted nucleotide prodrug for the treatment of HCV. Glob. Antivir. J.5 (Suppl. 1), 22 (2009).
  • Lallos L , La CollaM, SerraIet al. Combination of IDX184, a nucleotide prodrug polymerase inhibitor, with other classes of HCV inhibitors is additive to synergistic in the HCV replicon in vitro. Hepatol.50 (Suppl. 4), 267A (2009).
  • La Colla M , LallosLB, SerraIet al. A triple combination of direct-acting antiviral agents demonstrates robust anti-HCV activity in vitro. J. Hepatol.52 (Suppl. 1), S299 (2010).
  • Lam AM , EspirituC, BansalSet al. Hepatitis C virus nucleotide inhibitors PSI-352938 and PSI-353661 exhibit a novel mechanism of resistance requiring multiple mutations within replicon RNA. J. Virol.85 (23), 12334–12342 (2011).
  • Bilello JP , LallosLB, McCarvilleJFet al. In vitro activity and resistance profile of samatasvir, a novel NS5A replication inhibitor of hepatitis C virus. Antimicrob. Agents. Chemother.58 (8), 4431–4442 (2014).
  • Mayes BA , WangJ, ArumugasamyJet al. Scalable synthesis of a nucleoside phosphoramidate prodrug inhibitor of HCV NS5B RdRp: challenges in the production of a diastereomeric mixture. Org. Process Res. Dev.19 (4), 520–530 (2015).
  • Mayes BA , MoussaAM, Gasparac-KnezicRet al. Pharmaceutical compositions of 2′-C-methyl-guanosine,5′-[2[(3-hydroxy-2,2-dimethyl-1oxopropyl)thio]ethyl, N-(phenylmethyl)phosphoramidate]: US20130217644 (2013).
  • Zhou XJ , PietropaoloK, ChenJet al. Safety and pharmacokinetics of IDX184, a liver-targeted nucleotide polymerase inhibitor of hepatitis C virus, in healthy subjects. Antimicrob. Agents. Chemother.55 (1), 76–81 (2011).
  • Zhou XJ , PietropaoloK, Sullivan-BolyaiJet al. IDX184, a liver-targeted nucleotide HCV polymerase inhibitor: results of a first-in-man safety and pharmacokinetic study. J. Hepatol.50 (Suppl. 1), S351 (2009).
  • ClinicalTrials Database: NCT01157104. https://clinicaltrials.gov/ct2/show/NCT01157104.
  • Idenix Press . Idenix Pharmaceuticals provides updates on three clinical development programs. Press release. http://ir.idenix.com/releasedetail.cfm?ReleaseID=611432.
  • Lalezari J , AsmuthD, CasiroAet al. Short-term monotherapy with IDX184, a liver-targeted nucleotide polymerase inhibitor, in patients with chronic hepatitis C virus infection. Antimicrob. Agents. Chemother.56 (12), 6372–6378 (2012).
  • Lalezari J , PoordadF, MehraPet al. Antiviral activity, pharmacokinetics and safety of IDX184 in combination with pegylated interferon (PEGIFN) and ribavirin (RBV) in treatment-naive HCV genotype 1-infected subjects. J. Hepatol.52 (Suppl. 1), S469 (2010).
  • Lalezari J , O'RiordanW, PoordadFet al. A Phase IIa study of IDX184 in combination with pegylated interferon (pegIFN) and ribavirin (RBV) in treatment-naive genotype 1 HCV-infected subjects. Hepatology52 (Suppl. 4), 337A (2010).
  • Lalezari J , BoxT, O'RiordanWet al. IDX184 in combination with pegylated interferon-α2a and ribavirin for 2 weeks in treatment-naive patients with chronic hepatitis C. Antivir. Ther.18 (6), 755–764 (2013).
  • McCarville JF , DubucG, DonovanEet al. No resistance to IDX184 was detected in 3-day and 14-day clinical studies of IDX184 in genotype 1-infected HCV subject. J. Hepatol.54 (Suppl. 1), S488–S489 (2011).
  • Sheridan C . Calamitous HCV trial casts shadow over nucleoside drugs. Nat. Biotechnol.30 (11), 1015–1016 (2012).
  • Arnold JJ , SharmaSD, FengJYet al. Sensitivity of mitochondrial transcription and resistance of RNA polymerase II dependent nuclear transcription to antiviral ribonucleosides. PLoS Pathog.8 (11), e1003030 (2012).
  • FirstWord Pharma . Idenix Pharmaceuticals provides updates on IDX184 and IDX19368 development programs. Press release : www.firstwordpharma.com/node/1055565?tsid=17#axzz3WANE2q5f.
  • Baudy RB , ButeraJA : WO2004092189 (2004).
  • Ghose AK , CrippenG. Atomic physicochemical parameters for three dimensional structure-directed quantitative structure–activity relationships. I. Partition coefficient as a measure of hydrophobicity. J. Comput. Chem.7 (4), 567–577 (1986).
  • Viswanadhan VN , GhoseAK, RevankarGRet al. Atomic physicochemical parameters for three dimensional structure directed quantitative structure–activity relationships. 4. Additional parameters for hydrophobic and dispersive interactions and their application for an automated superposition of certain naturally occurring nucleoside antibiotics. J. Chem. Inf. Comp. Sci.29 (3), 163–172 (1989).
  • Ghose AK , ViswanadhanVN, WendoloskiJJ. Prediction of hydrophobic (lipophilic) properties of small organic molecules using fragmental methods: an analysis of ALOGP and CLOGP methods. J. Phys. Chem. A.102 (21), 3762–3772 (1998).
  • Luo G , HamatakeRK, MathisDM, RacelaJ, RigatKL, ColonnoRJ. De novo initiation of RNA synthesis by the RNA-dependent RNA polymerase (NS5B) of hepatitis C virus. J. Virol.74, 851–863 (2000).
  • Zhu Q , GuoJT, SeegerC. Replication of hepatitis C virus subgenomes in nonhepatic epithelial and mouse hepatoma cells. J. Virol.77, 9204–9210 (2003).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.