106
Views
3
CrossRef citations to date
0
Altmetric
Original Research

Metalloprotoporphyrin Inhibition of HCV NS3-4A Protease: Structure–Activity Relationships

ORCID Icon, , , ORCID Icon, , & show all
Pages 757-771 | Published online: 24 Feb 2020

References

  • TholeyDM, AhnJ. Impact of hepatitis C virus infection on hepatocellular carcinoma. Gastroenterol Clin North Am. 2015;44(4):761–773. doi:10.1016/j.gtc.2015.07.00526600218
  • KayaliZ, SchmidtWN. Finally sofosbuvir: an oral anti-HCV drug with wide performance capability. Pharmgenomics Pers Med. 2014;7:387–398. doi:10.2147/PGPM.S5262925540594
  • LuoD, VasudevanSG, LescarJ. The flavivirus NS2B-NS3 protease-helicase as a target for antiviral drug development. Antiviral Res. 2015;118:148–158. doi:10.1016/j.antiviral.2015.03.01425842996
  • LindenbachBD, RiceCM. Unravelling hepatitis C virus replication from genome to function. Nature. 2005;436(7053):933–938. doi:10.1038/nature0407716107832
  • JacobsonIM, McHutchisonJG, DusheikoG, et al. Telaprevir for previously untreated chronic hepatitis C virus infection. N Engl J Med. 2011;364(25):2405–2416. doi:10.1056/NEJMoa101291221696307
  • PoordadF, McConeJ Jr., BaconBR, et al. Boceprevir for untreated chronic HCV genotype 1 infection. N Engl J Med. 2011;364(13):1195–1206. doi:10.1056/NEJMoa101049421449783
  • ShermanKE, FlammSL, AfdhalNH, et al. Response-guided telaprevir combination treatment for hepatitis C virus infection. N Engl J Med. 2011;365(11):1014–1024. doi:10.1056/NEJMoa101446321916639
  • ChayamaK, TakahashiS, ToyotaJ, et al. Dual therapy with the nonstructural protein 5A inhibitor, daclatasvir, and the nonstructural protein 3 protease inhibitor, asunaprevir, in hepatitis C virus genotype 1b-infected null responders. Hepatology. 2012;55(3):742–748. doi:10.1002/hep.2472421987462
  • LokAS, GardinerDF, LawitzE, et al. Preliminary study of two antiviral agents for hepatitis C genotype 1. N Engl J Med. 2012;366(3):216–224. doi:10.1056/NEJMoa110443022256805
  • SchmidtWN, NelsonDR, PawlotskyJM, ShermanKE, ThomasDL, ChungRT. Direct-acting antiviral agents and the path to interferon independence. Clin Gastroenterol Hepatol. 2014;12(5):728–737. doi:10.1016/j.cgh.2013.06.02423872239
  • BourliereM, GordonSC, FlammSL, et al. Sofosbuvir, velpatasvir, and voxilaprevir for previously treated HCV infection. N Engl J Med. 2017;376(22):2134–2146. doi:10.1056/NEJMoa161351228564569
  • de LeuwP, StephanC. Protease inhibitor therapy for hepatitis C virus-infection. Expert Opin Pharmacother. 2018;19(6):577–587. doi:10.1080/14656566.2018.145442829595065
  • GaneE, PoordadF, WangS, et al. High efficacy of ABT-493 and ABT-530 treatment in patients with HCV genotype 1 or 3 infection and compensated cirrhosis. Gastroenterology. 2016;151(4):651–659.e651. doi:10.1053/j.gastro.2016.07.02027456384
  • JacobsonIM, LawitzE, GaneEJ, et al. Efficacy of 8 weeks of Sofosbuvir, Velpatasvir, and Voxilaprevir in patients with chronic HCV infection: 2 phase 3 randomized trials. Gastroenterology. 2017;153(1):113–122. doi:10.1053/j.gastro.2017.03.04728390869
  • LawitzE, GaneE, PearlmanB, et al. Efficacy and safety of 12 weeks versus 18 weeks of treatment with grazoprevir (MK-5172) and elbasvir (MK-8742) with or without ribavirin for hepatitis C virus genotype 1 infection in previously untreated patients with cirrhosis and patients with previous null response with or without cirrhosis (C-WORTHY): a randomised, open-label Phase 2 trial. Lancet (London, England). 2015;385(9973):1075–1086. doi:10.1016/S0140-6736(14)61795-5
  • SulkowskiM, HezodeC, GerstoftJ, et al. Efficacy and safety of 8 weeks versus 12 weeks of treatment with grazoprevir (MK-5172) and elbasvir (MK-8742) with or without ribavirin in patients with hepatitis C virus genotype 1 mono-infection and HIV/hepatitis C virus co-infection (C-WORTHY): a randomised, open-label phase 2 trial. Lancet (London, England). 2015;385(9973):1087–1097. doi:10.1016/S0140-6736(14)61793-1
  • ZeuzemS, FosterGR, WangS, et al. Glecaprevir-pibrentasvir for 8 or 12 weeks in HCV genotype 1 or 3 infection. N Engl J Med. 2018;378(4):354–369. doi:10.1056/NEJMoa170241729365309
  • SchmidtWN, MathahsMM, ZhuZ. Heme and HO-1 Inhibition of HCV, HBV, and HIV. Front Pharmacol. 2012;3:129. doi:10.3389/fphar.2012.0012923060790
  • TsengC-K, LinC-K, WuY-H, et al. Human heme oxygenase 1 is a potential host cell factor against dengue virus replication. Sci Rep. 2016;6:32176. doi:10.1038/srep3217627553177
  • GozzelinoR, JeneyV, SoaresMP. Mechanisms of cell protection by heme oxygenase-1. Annu Rev Pharmacol. 2010;50:323–354. doi:10.1146/annurev.pharmtox.010909.105600
  • LiuY-S, LiH-S, QiD-F, et al. Zinc protoporphyrin IX enhances chemotherapeutic response of hepatoma cells to cisplatin. World J Gastroenterol. 2014;20(26):8572–8582. doi:10.3748/wjg.v20.i26.857225024611
  • LohmannV, KornerF, KochJO, HerianU, TheilmannL, BartenschlagerR. Replication of subgenomic hepatitis C virus RNAs in a hepatoma cell line. Science. 1999;285(5424):110–113. doi:10.1126/science.285.5424.11010390360
  • ZhuZ, WilsonAT, MathahsMM, et al. Heme oxygenase-1 suppresses hepatitis C virus replication and increases resistance of hepatocytes to oxidant injury. Hepatology. 2008;48(5):1430–1439. doi:10.1002/hep.2249118972446
  • LindenbachBD, EvansMJ, SyderAJ, et al. Complete replication of hepatitis C virus in cell culture. Science. 2005;309(5734):623–626. doi:10.1126/science.111401615947137
  • ZhuZ, WilsonAT, LuxonBA, et al. Biliverdin inhibits hepatitis C virus nonstructural 3/4A protease activity: mechanism for the antiviral effects of heme oxygenase? Hepatology. 2010;52(6):1897–1905. doi:10.1002/hep.2392121105106
  • LaP, FernandoAP, WangZ, et al. Zinc protoporphyrin regulates cyclin D1 expression independent of heme oxygenase inhibition. J Biol Chem. 2009;284(52):36302–36311. doi:10.1074/jbc.M109.03164119850937
  • MosmannT. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods. 1983;65(1–2):55–63. doi:10.1016/0022-1759(83)90303-46606682
  • ZhuZ, MathahsMM, SchmidtWN. Restoration of type I interferon expression by heme and related tetrapyrroles through inhibition of NS3/4A protease. J Infect Dis. 2013;208(10):1653–1663. doi:10.1093/infdis/jit33823901085
  • AbdallaMY, BritiganBE, WenF, et al. Down-regulation of heme oxygenase-1 by hepatitis C virus infection in vivo and by the in vitro expression of hepatitis C core protein. J Infect Dis. 2004;190(6):1109–1118. doi:10.1086/jid.2004.190.issue-615319861
  • JiangY, AndrewsSW, CondroskiKR, et al. Discovery of danoprevir (ITMN-191/R7227), a highly selective and potent inhibitor of hepatitis C virus (HCV) NS3/4A protease. J Med Chem. 2014;57(5):1753–1769. doi:10.1021/jm400164c23672640
  • PietschmannT, LohmannV, RutterG, KurpanekK, BartenschlagerR. Characterization of cell lines carrying self-replicating hepatitis C virus RNAs. J Virol. 2001;75(3):1252–1264. doi:10.1128/JVI.75.3.1252-1264.200111152498
  • StuyverLJ, McBrayerTR, TharnishPM, et al. Dynamics of subgenomic hepatitis C virus replicon RNA levels in Huh-7 cells after exposure to nucleoside antimetabolites. J Virol. 2003;77(19):10689–10694. doi:10.1128/JVI.77.19.10689-10694.200312970456
  • ShangL, LinK, YinZ. Resistance mutations against HCV protease inhibitors and antiviral drug design. Curr Pharm Des. 2014;20(5):694–703. doi:10.2174/1381612811319999000823688081
  • PawlotskyJ-M. New hepatitis C therapies: the toolbox, strategies, and challenges. Gastroenterology. 2014;146(5):1176–1192. doi:10.1053/j.gastro.2014.03.00324631495
  • SerreSBN, JensenSB, GhanemL, et al. Hepatitis C virus genotype 1 to 6 protease inhibitor escape variants: in vitro selection, fitness, and resistance patterns in the context of the infectious viral life cycle. Antimicrob Agents Chemother. 2016;60(6):3563–3578. doi:10.1128/AAC.02929-1527021330
  • SorboMC, CentoV, Di MaioVC, et al. Hepatitis C virus drug resistance associated substitutions and their clinical relevance: update 2018. Drug Resist Updat. 2018;37:17–39. doi:10.1016/j.drup.2018.01.00429525636
  • LohmannV. Hepatitis C virus cell culture models: an encomium on basic research paving the road to therapy development. Med Microbiol Immunol. 2019;208(1):3–24. doi:10.1007/s00430-018-0566-x30298360
  • LamarreD, AndersonPC, BaileyM, et al. An NS3 protease inhibitor with antiviral effects in humans infected with hepatitis C virus. Nature. 2003;426(6963):186–189. doi:10.1038/nature0209914578911
  • NgTI, TripathiR, ReischT, et al. In vitro antiviral activity and resistance profile of the next-generation hepatitis C Virus NS3/4A protease inhibitor glecaprevir. Antimicrob Agents Chemother. 2018;62(1):e01620–e016217.29084747
  • TaylorJG, ZipfelS, RameyK, et al. Discovery of the pan-genotypic hepatitis C virus NS3/4A protease inhibitor voxilaprevir (GS-9857): a component of Vosevi®. Bioorg Med Chem Lett. 2019;29:2428–2436. doi:10.1016/j.bmcl.2019.03.03731133531
  • SummaV, LudmererSW, McCauleyJA, et al. MK-5172, a selective inhibitor of hepatitis C virus NS3/4a protease with broad activity across genotypes and resistant variants. Antimicrob Agents Chemother. 2012;56(8):4161–4167. doi:10.1128/AAC.00324-1222615282
  • StaudingerR, AbrahamNG, LevereRD, KappasA. Inhibition of human immunodeficiency virus-1 reverse transcriptase by heme and synthetic heme analogs. P Assoc Am Physician. 1996;108(1):47–54.
  • LinL, HuJ. Inhibition of hepadnavirus reverse transcriptase-epsilon RNA interaction by porphyrin compounds. J Virol. 2008;82(5):2305–2312. doi:10.1128/JVI.02147-0718094191
  • McPheeF, CalderaPS, BemisGW, McDonaghAF, KuntzID, CraikCS. Bile pigments as HIV-1 protease inhibitors and their effects on HIV-1 viral maturation and infectivity in vitro. Biochem J. 1996;320:681–686. doi:10.1042/bj32006818973584
  • LowJGH, OoiEE, VasudevanSG. Current status of dengue therapeutics research and development. J Infect Dis. 2017;215(suppl_2):S96–S102. doi:10.1093/infdis/jiw42328403438
  • SarrazinC, HezodeC, ZeuzemS, PawlotskyJM. Antiviral strategies in hepatitis C virus infection. J Hepatol. 2012;56(Suppl 1):S88–S100. doi:10.1016/S0168-8278(12)60010-522300469
  • LoveRA, PargeHE, WickershamJA, et al. The crystal structure of hepatitis C virus NS3 proteinase reveals a trypsin-like fold and a structural zinc binding site. Cell. 1996;87(2):331–342. doi:10.1016/S0092-8674(00)81350-18861916
  • NogalesD, LightnerDA. On the structure of bilirubin in solution - C-13(H-1) heteronuclear overhauser effect Nmr analyses in aqueous buffer and organic-solvents. J Biol Chem. 1995;270(1):73–77. doi:10.1074/jbc.270.1.737814422
  • WestA-R, OatesP-S. Mechanisms of heme iron absorption: current questions and controversies. World J Gastroenterol. 2008;14(26):4101–4110. doi:10.3748/wjg.14.410118636652
  • DangTN, BishopGM, DringenR, RobinsonSR. The putative heme transporter HCP1 is expressed in cultured astrocytes and contributes to the uptake of hemin. Glia. 2010;58(1):55–65. doi:10.1002/glia.v58:119533605
  • ShayeghiM, Latunde-DadaGO, OakhillJS, et al. Identification of an intestinal heme transporter. Cell. 2005;122(5):789–801. doi:10.1016/j.cell.2005.06.02516143108
  • HalfonP, LocarniniS. Hepatitis C virus resistance to protease inhibitors. J Hepatol. 2011;55(1):192–206. doi:10.1016/j.jhep.2011.01.01121284949
  • VermehrenJ, SarrazinC. The role of resistance in HCV treatment. Best Pract Res Clin Gastroenterol. 2012;26(4):487–503. doi:10.1016/j.bpg.2012.09.01123199507
  • EspinozaJA, GonzalezPA, KalergisAM. Modulation of antiviral immunity by heme oxygenase-1. Am J Pathol. 2017;187(3):487–493. doi:10.1016/j.ajpath.2016.11.01128082120
  • ProtzerU, SeyfriedS, QuasdorffM, et al. Antiviral activity and hepatoprotection by heme oxygenase-1 in hepatitis B virus infection. Gastroenterology. 2007;133(4):1156–1165. doi:10.1053/j.gastro.2007.07.02117919491
  • QiuL, FanH, JinW, et al. miR-122-induced down-regulation of HO-1 negatively affects miR-122-mediated suppression of HBV. Biochem Biophys Res Commun. 2010;398(4):771–777. doi:10.1016/j.bbrc.2010.07.02120633528
  • WangS, AveryJE, HannafonBN, LindSE, DingW-Q. Zinc protoporphyrin suppresses cancer cell viability through a heme oxygenase-1-independent mechanism: the involvement of the Wnt/beta-catenin signaling pathway. Biochem Pharmacol. 2013;85(11):1611–1618. doi:10.1016/j.bcp.2013.03.01123523860
  • WangS, HannafonBN, LindSE, DingW-Q. Zinc protoporphyrin suppresses beta-catenin protein expression in human cancer cells: the potential involvement of lysosome-mediated degradation. PLoS One. 2015;10(5):e0127413. doi:10.1371/journal.pone.012741326000787
  • ZhuZ, TranH, MathahsMM, SchmidtWN Specific ZnPP inhibition and degradation of telomerase in human hepatoma cells (abstract #1833). Poster presented at American Association for Study of Liver Diseases, Annual Meeting; Washington, DC: 10 20–24; 2017.
  • IyerJK, ShiL, ShankarAH, SullivanDJ Jr. Zinc protoporphyrin IX binds heme crystals to inhibit the process of crystallization in Plasmodium falciparum. Mol Med. 2003;9(5–8):175–182. doi:10.1007/BF0340218214571325
  • CohenRS, WongRJ, StevensonDK. Understanding neonatal jaundice: a perspective on causation. Pediatr Neonatol. 2010;51(3):143–148. doi:10.1016/S1875-9572(10)60027-720675237
  • FangJ, SawaT, AkaikeT, et al. In vivo antitumor activity of pegylated zinc protoporphyrin: targeted inhibition of heme oxygenase in solid tumor. Cancer Res. 2003;63(13):3567–3574.12839943
  • FangJ, SawaT, AkaikeT, GreishK, MaedaH. Enhancement of chemotherapeutic response of tumor cells by a heme oxygenase inhibitor, pegylated zinc protoporphyrin. Int J Cancer. 2004;109(1):1–8. doi:10.1002/(ISSN)1097-021514735461
  • FillebeenC, Rivas-EstillaAM, BisaillonM, et al. Iron inactivates the RNA polymerase NS5B and suppresses subgenomic replication of hepatitis C virus. J Biol Chem. 2005;280(10):9049–9057. doi:10.1074/jbc.M41268720015637067
  • YuasaK, NaganumaA, SatoK, et al. Zinc is a negative regulator of hepatitis C virus RNA replication. Liver Int. 2006;26(9):1111–1118. doi:10.1111/liv.2006.26.issue-917032412