989
Views
15
CrossRef citations to date
0
Altmetric
Reviews

Imino sugar glucosidase inhibitors as broadly active anti-filovirus agents

, , &
Pages 1-7 | Received 29 Jul 2013, Accepted 22 Sep 2013, Published online: 25 Jan 2019

Abstract

Ebola virus and Marburg virus are members of the family of Filoviridae and are etiological agents of a deadly hemorrhagic fever disease. The clinical symptoms of Ebola and Marburg hemorrhagic fevers are difficult to distinguish and there are currently no specific antiviral therapies against either of the viruses. Therefore, a drug that is safe and effective against both would be an enormous breakthrough. We and others have shown that the folding of the glycoproteins of many enveloped viruses, including the filoviruses, is far more dependent upon the calnexin pathway of protein folding than are most host glycoproteins. Drugs that inhibit this pathway would be expected to be selectively antiviral. Indeed, as we summarize in this review, imino sugars that are competitive inhibitors of the host endoplasmic reticular α-glucosidases I and II, which are enzymes that process N-glycan on nascent glycoproteins and thereby inhibit calnexin binding to the nascent glycoproteins, have been shown to have antiviral activity against a number of enveloped viruses including filoviruses. In this review, we describe the state of development of imino sugars for use against the filoviruses, and provide an explanation for the basis of their antiviral activity as well as limitations.

Introduction

Filoviral hemorrhagic fevers are rare but highly lethal diseases associated with outbreaks in developing countries.Citation1,Citation2 The causative agents, Ebola virus and Marburg virus, are considered to be high level biothreat agents by the United States Centers for Disease Control and Prevention and the National Institutes of Health.Citation3 There are no effective vaccines to be used as prophylactics and no effective antiviral interventions to manage the diseases. Because the clinical symptoms of Marburg and Ebola hemorrhagic fevers are difficult to distinguish, a drug that is effective against either would be a first, and one that is effective against both the viruses would be an enormous breakthrough.

Filoviruses are non-segmented negative-strand RNA viruses that produce filamentous enveloped virions.Citation4 There are currently four known clinically relevant species of Ebola virusCitation4 and a single species of Marburg virus.Citation5 Initial virus replication occurs in mononuclear cells and viremia is usually apparent within 2 days after infection.Citation6,Citation7,Citation8,Citation9 Death can occur in up to 90% of the infections after 7–10 days of symptoms, usually due to hemorrhagic fevers.Citation5

Although the cell receptors for either Ebola virus or Marburg virus have not been fully characterized, the broad cell tropism of the viruses suggests a wide distribution of their receptors. In any regard, the trimeric envelope glycoprotein (GP) spikes of the filoviruses are believed to mediate their entry into host cells via endocytic pathways. Within endo/lysosomal compartments, host endosomal cysteine proteases (cathepsins) cleave the filoviral GP1 protein to generate an entry intermediate comprising an N-terminal GP1 fragment and GP2. Recent work indicates that a cleaved form of Ebola virus GP subsequently interacts with Niemann-Pick C1, an endo/lysosomal cholesterol transporter, to trigger membrane fusion.Citation9

As illustrated in , the pipeline of candidate anti-filovirus therapeutics is limited. However, promising work on inhibition of the virus entry into their host cells with small molecules,Citation21 recombinant C type lectinsCitation13,Citation22 or immuno-adhesion technology,Citation23 disruption of viral RNA cappingCitation24 and directly targeting viral RNA with antisense oligos or siRNAsCitation10,Citation11,Citation25,Citation26 have recently been reported.

As also indicates, the oligonucleotide-based therapeutics appear to be the farthest along in development, reaching Phase I clinical trials, but challenges remain and the extent to which these approaches will progress is uncertain. Some of these challenges are inherent to the oligonucleotide approach and the lack of efficient cell-specific delivery technologies.Citation27 Although a morpholino modification or a lipid nanoparticle formulation has been used for these approaches in their anti-filovirus applications, specific targeted delivery of oligonucleotide into disease-relevant tissues and cells remains a major issue to address.Citation28

Only a few small molecule antivirals have been shown to have in vivo efficacy against filoviruses. FGI-103, 104, 106 and NSC62914 were discovered using cell-based high-throughput screening, and their antiviral mechanisms are largely unknown. The other two families of small molecules target host enzymes, the S-adenosyl-L-homocysteine hydrolase and endoplasmic reticulum (ER) α-glucosidases I and II. The ER α-glucosidases are especially interesting because the broad-spectrum antiviral activities of their inhibitors have been extensively demonstrated with a variety of enveloped viruses from many families (). In addition, more than 1000 US FDA-approved drugs were tested for new antiviral indications.Citation18 This effort resulted in the discovery that chloroquine could disrupt entry and replication of two or more viruses in vitro and protect mice against Ebola virus challenge in vivo.

As for many other enveloped viruses,Citation29,Citation30,Citation31,Citation32,Citation33,Citation34,Citation35,Citation36 the morphogenesis of filoviruses appears to depend upon ER α-glucosidases mediated processing of envelope glycoproteins. Therefore, the amplification and propagation of filoviruses are sensitive to imino sugar derivatives that inhibit the ER α-glucosidases I and II.Citation37 Due to their unusual antiviral mechanism, imino sugars should be complementary to the other antiviral approaches and have the advantage of being broadly active.

Table 1 Antiviral therapeutics in clinical and preclinical development for the management of human pathogenic filoviruses

TARGETING HOST FUNCTIONS TO SUPPRESS VIRAL INFECTION

Medical management of virus infection can, in theory and has in practice, involve targeting either virus or host functions. Targeting virus specified functions such as viral enzymes (DNA or RNA polymerases, proteases, helicases, neuraminidases) has been enormously effective and offered opportunities for great selectivity.Citation38,Citation39,Citation40 On the other hand, drugs targeting host functions, upon which the viruses rely, have been approached with more skepticism because of concerns regarding selectivity and toxicity. shows a few examples that have been attempted for filoviruses, and discovery of the virus receptor may lead to a few more. The success of the interferon therapy makes the point that it is possible to identify host functions that can serve as targets of broad spectrum antivirals.Citation41,Citation42 However, there are, to date, very few examples of small molecule antivirals that target host functions and retain broad activity. It is a short list, including inhibitors of host cellular cyclophilins,Citation43,Citation44 inosine 5-monophosphate dehydrogenase,Citation45 3-hydroxy-3-methylglutaryl-coenzyme A reductase,Citation46 S-adenosyl-L-homocysteine hydrolaseCitation14,Citation47 and ER α-glucosidases. The ER α-glucosidase inhibitors are thus in a rare group, having been shown to have selective antiviral activity for multiple enveloped viruses in tissue cultures and, in several cases, in animal models (). Why viral functions are more sensitive to glucosidase inhibition than are host functions is becoming appreciated and touched on briefly below.

Table 2 Broad-spectrum antiviral activity of imino sugar derivatives in vitro and in vivo

FUNCTION OF ER α-GLUCOSIDASES AND CONSEQUENCES OF INHIBITION

Briefly, as illustrated in , the N-linked glycans of mammalian glycoproteins are ‘processed’ with the sequential removal of their terminal glucose residues by the ER-resident glucosidases I and II, shortly after becoming glycosylated at specific asparagine residues. Remarkably, cells in culture can tolerate complete shutdown of these ER α-glucosidases.Citation74,Citation75,Citation76,Citation77,Citation78 Gucosidase I and II knockout mice have limited life spans.Citation79,Citation80,Citation81 People can tolerate long-term (months and years) treatment with glucosidase inhibitors under conditions where there is substantial repression of the ER enzymes. However, there are troubling adverse effects (i.e., gastric distress) which must be taken into consideration.Citation36,Citation82

Figure 1 Glucosidase mediated steps in the N-linked glycosylation pathway. The pathway of N-linked glycosylation at asparagine of nascent polypeptides is shown. Polypeptides synthesized by translation in the ER are shown as the ribbon line (black) as unfolded, with the unprocessed 3-glucose terminal containing ‘lollipop’ oligosaccharide structure, attached at an asparagine. The terminal glucose and second to terminal glucoses (Glu) of this oligosaccharide is removed processively by ER resident, membrane bound, glucosidases I and II. Following removal of these glucose molecules, the protein chaperon, Calnexin, mediates folding of the polypeptide, which is then transported to the Golgi and further processed for secretion. Inhibition of ER glucosidases prevents polypeptide interaction with Calnexin and results in polypeptide misfolding, retention and/or degradation.

Glucosidase inhibitors have been approved for the management of type II diabetes, Gaucher's disease. Glucosidase inhibitors had also advanced to phase II human trials for management of hepatitis C virus and human immunodeficiency virus infection.Citation82,Citation83 Thus, although mammalian cells and animals tolerate significant, and even total, repression of ER glucosidases, many viruses cannot. Indeed, completion of the life cycle of many enveloped viruses requires functional glucosidases.Citation29 For example, viruses such as the hepadnaviruses, flaviviruses, filoviruses and influenza virus are significantly repressed by imino sugars under conditions where there is no detectable affect upon the host cells, or in the cases of animal experiments, only limited affect upon the host animalsCitation36,Citation74,Citation84 (). The sensitivity of these viruses to glucosidase inhibitors is presumably because they possess envelope proteins that must oligomerize, and they have an apparent requirement for glucosidase processing of their glycoproteins presumably to enable time-sensitive proper calnexin mediated folding for their oligomerization.Citation35,Citation49,Citation52,Citation58,Citation85

Taken together, the trend is clear and has not been broken for any of the viruses that have been tested so far: enveloped viruses that bud from intracellular membranes and/or use calnexin dependent pathways are selectively sensitive to glucosidase inhibitors. This has been expanded to viruses from four families causing hemorrhagic fevers (Filoviridae, Bunyaviridae, Arenaviridae and Flaviviridae) ().

CURRENT USES AND LIMITATIONS OF GLUCOSIDASE INHIBITORS

A wide variety of structurally diverse compounds, from both nature and synthesis, have been identified as glucosidase inhibitors.Citation86,Citation87 Some of them have been successfully used to treat human diseases.Citation82,Citation83 For example, inhibition of intestinal glucosidases digesting carbohydrates is therapeutic for the management of type II diabetes.Citation88 The imino sugar N-butyl-deoxynorjirimycin (NBDNJ) is currently approved for the treatment of Gaucher's disease, with patients taking near gram amounts a day for many years.Citation89,Citation90 However, the molecular target of the imino sugar in Gaucher's disease is a glucocerebroside transferase, which is highly sensitive to NBDNJ, and not ER α-glucosidases. Nevertheless, these successes demonstrate the principle of tolerability of this category of drugs, as well as their practical therapeutic benefits.

The benefits of imino sugar glucosidase inhibitors in the treatment of viral infections have been less clear. The currently available and human-tested glucosidase inhibitors are all limited by side effects and poor pharmacokinetic properties that make them less appealing for controlling chronic diseases. For example, although glucosidase inhibitor cellgosivir met the viral reduction milestone for the treatment of chronic hepatitis C in human studies, the drug was not advanced presumably because of gastritis and, frankly, the emergence of more potent direct acting antivirals. Thus, a consistent problem has been maintaining serum concentrations of compounds that are antiviral without causing the intestinal upsets, due to the inhibition of intestinal glucosidases.Citation30,Citation91

ANTIVIRAL ACTIVITY OF IMINO SUGARS AGAINST HEMORRHAGIC FEVER VIRUSES

There are now several reports showing that orally administrated imino sugar derivatives inhibited dengue virusCitation59,Citation60,Citation61,Citation62 and Japanese encephalitis virus Citation67 in mice, as well as woodchuck hepatitis virus in woodchucks.Citation30 Although the reduction of viremia level was limited, the results were encouraging. We note, however, that reduction of viremia by even 1–2 logs has been anticipated to significantly improve clinical outcomes by reducing the severity of disease and increasing survival rates.Citation39,Citation92 Along these lines, we recently reported imino sugar compounds that significantly reduced mortality in mouse model of lethal derivatives inhibited dengue virus infection.Citation63 Taken together, these results make it reasonable to consider finding broadly active glucosidase inhibitors reaching therapeutic levels in people for multiple viral indications.

However, as indicated, one major limitation for developing imino sugar antivirals has been the lack of potency and/or poor pharmacokinetic properties, which lead to difficulty in maintaining therapeutic drug concentrations in vivo. We have improved the antiviral potency of imino sugars from the platform (NBDNJ) by more than 500-fold (). For example, based upon the encouraging in vivo efficacy results achieved with Dengue mouse models, we have synthesized imino sugar glucosidase inhibitors, represented by tert butyl urea (u) DNJ (19029), with submicromolar antiviral activity against four families of hemorrhagic fever viruses in cultured cells.Citation37 In addition, significant in vivo efficacy in Ebola and Marburg virus infected animals has also been achieved (70%–80% protection).

Figure 2 Modifications of the imino sugar NBDNJ that greatly improve antiviral activity but not enzyme inhibitory activity. The imino sugar NBDNJ is a butylated DNJ with millimolar antiviral activity in vitro. Alterations of its side chain as represented in compounds CM-10-18 and IHVR-19029, improve antiviral activity by up to 1000-fold, but only modestly improve enzyme inhibition. This is presumably because the improvements are largely related to cell access. See text. ND, not done.

MOLECULAR BASIS OF THE ANTIVIRAL SELECTIVITY OF GLUCOSIDASE INHIBITORS

Imino sugars, such as those with DNJ head groups, are glucose mimetics competitive inhibitors of the N-glycan processing enzymes glucosidase I and II (). As stated and shown in , all N-linked glycans, following transfer to acceptor asparagine amino acids on nascent glycoproteins, are ‘trimmed’ in the ER by a series of sequentially active glycoprocessing enzymes. ER α-glucosidases I and II are the first enzymes to function in this pathway.Citation77,Citation78 Specifically, all nascent N-linked glycoproteins contain three terminal glucose residues at the distal termini of their N-glycans following transfer of the oligosaccharide to the protein by way of the enzyme oligosaccharyltransferase. Immediately following this transfer, the terminal glucoses are removed, sequentially, by the action of ER resident enzymes glucosidases I and then II. The ER chaperons, calnexin and calreticulin, recognize monoglucosylated N-glycans and then ‘fold’ the nascent glycoprotein after which it is further processed by mannosidases and transferred to the Golgi where it is further modified into its characteristic complex carbohydrate structures. Underglucosylated polypeptides that have not been folded and have not transferred to the Golgi may get second chances following reglucosylation and the possibility of calnexin and calreticulin interactions by the enzyme UDP-glucose glycoprotein: glucosyltransferase. However, ultimately, misfolded or unfolded polypeptides are sent to the proteasomes where they are degraded. Thus, glucosidase inhibitors prevent the interaction of nascent N-glycoprotiens with calnexin and calreticulin and cause their misfolding and degradation. Misfolded or unfolded polypeptides may be sent to the proteasomes for degradation. It has been known for more than 20 years that many viruses are sensitive to glucosidase inhibition, and the sensitivity of Sindbis virus, influenza virus and fowl plague virus envelope proteins was reported in the 1980s.Citation33

Viruses that depend upon calnexin and calreticulin would thus be expected to be the most sensitive to glucosidase inhibition. And we developed a ‘biogenesis’ theory that viruses that bud from the endoplasmic reticulum would be sensitive to glucosidase inhibitors. This was not to say that viruses that did not bud from the ER would not be sensitive to glucosidase inhibitors, and indeed the infectivity of retroviruses which bud from the plasma membrane is apparently greatly affected by glucosidase inhibitors.Citation93 We would now offer a refinement of the ‘biogenesis’ theory, in light of what is now known, to claim that viruses that depend upon the calnexin/calreticulin type pathway for morphogenesis will be sensitive to glucosidase inhibition.

These predictions, for the most part, seem to be supported by experimental results. Most strikingly is the example of hepatitis B virus and bovine viral diarrhea virus which are both sensitive to glucosidase inhibition, and are two completely different viruses.Citation8,Citation94,Citation95,Citation96 Hepatitis B virus has a DNA genome and is a human pararetrovirus that primarily infects hepatocytes. Bovine viral diarrhea virus is a flavivirus with an RNA genome and grows primarily in non-liver cells. Both, however, acquire their envelopes by budding into the endoplasmic reticulum.Citation8,Citation94,Citation95,Citation96 Both, we now know, also depend upon calnexin for the maturation of specific viral glycoproteins.Citation97,Citation98,Citation99,Citation100 As shows, both are also greatly inhibited by glucosidase inhibitors at concentrations that do not apparently affect cell viability.

ADDITIONAL BIOLOGICAL ACTIVITY OF IMINO SUGARS

Although imino sugars have been developed as host glucosidase inhibitors to disrupt viral glycoprotein folding and consequentially, virion morphogenesis, the compounds have additional biological activities due to either on-target suppression of ER α-glucosidases or off-target interference of other cellular components.Citation101 For example, on-target inhibition of ER glucosidase results in viral glycoprotein degradation has been shown to enhance major histocompatibility complex I presentation of epitopes derived from viral envelope glycoproteins. Moreover, changes in the glycan structures associated with viral glycoproteins may also alter their interaction with C-type lectins and consequentially, inhibit pro-inflammatory cytokine production.Citation62 In addition, some imino sugars may even have off target activities, repressing the virus by mechanisms that do not involve inhibition of ER glucosidases.Citation59 Off-target activities are not unusual in compound development, and in the case of extra biological effects upon antigen presentation and cytokine function, could even be beneficial. They may also provide interesting research leads. However, it is essential that such activities be recognized and be part of the decision to either progress or disqualify a compound, and if compounds with off-target activity are advanced, these activities must be monitored.

CONCLUSIONS

The imino sugars have, in the past, been proposed primarily for use in chronic infections such as human immunodeficiency virus and chronic viral hepatitis.Citation30,Citation82,Citation83 However, they may be best suited to treat acute viral infections, such as filoviral hemorrhagic fevers which involve weeks of therapy, as opposed to chronic infections which may require years of use. This would certainly avoid the complications that sometimes appear with longer-term use. Moreover, the new generation of compounds being brought forward should be designed to avoid or reduce intestinal distress. Finally, used for life-threatening infections such as the hemorrhagic fevers or severe influenza, either alone or in combinations with other management regimes, the glucosidase inhibitors could provide a powerful option in the treatment tool kit.

Acknowledgments

The authors appreciate help from Ms Erica Litschi, Department of Microbiology and Immunology, Drexel University College of Medicine, for manuscript preparation and the United States National Institutes of Health (R01 AI104636) and the Hepatitis B Foundation and Commonwealth Wealth of Pennsylvania for support.

Notes

Abbreviations: EBOV, Ebola virus; mAb, monoclonal antibody; MARV, Marburg virus; PMO, phosphorodiamidate morpholino oligomer; SNALP-siRNA, stable nucleic acid lipid particle-small intefering RNA.

  • Hensley LE, Alves DA, Geisbert JBet al.Pathogenesis of Marburg hemorrhagic fever in cynomolgus macaques. J Infect Dis2011;204( Suppl 3):S1021–S1031.
  • Guo H, Pan X, Mao Ret al.Alkylated porphyrins have broad antiviral activity against hepadnaviruses, flaviviruses, filoviruses, and arenaviruses. Antimicrob Agents Chemother2011;55:478–486.
  • Geisbert TW, Jahrling PB.Exotic emerging viral diseases: progress and challenges. Nat Med.2004;10( 12 Suppl):S110–S121.
  • Towner JS, Sealy TK, Khristova MLet al.Newly discovered ebola virus associated with hemorrhagic fever outbreak in Uganda. PLoS Pathog2008;4:e1000212.
  • Kortepeter MG, Bausch DG, Bray M.Basic clinical and laboratory features of filoviral hemorrhagic fever. J Infect Dis2011;204( Suppl 3):S810–S816.
  • Bray M.Pathogenesis of viral hemorrhagic fever. Curr Opin Immunol2005;17:399–403.
  • Bausch DG, Sprecher AG, Jeffs B, Boumandouki P.Treatment of Marburg and Ebola hemorrhagic fevers: a strategy for testing new drugs and vaccines under outbreak conditions. Antiviral Res2008;78:150–161.
  • Miller S, Krijnse-Locker J.Modification of intracellular membrane structures for virus replication. Nat Rev Microbiol2008;6:363–374.
  • Krishnan A, Miller EH, Herbert ASet al.Niemann-Pick C1 (NPC1)/NPC1-like1 chimeras define sequences critical for NPC1's function as a flovirus entry receptor. Viruses2012;4:2471–2484.
  • Warren TK, Warfield KL, Wells Jet al.Advanced antisense therapies for postexposure protection against lethal filovirus infections. Nat Med2010;16:991–994.
  • Geisbert TW, Lee AC, Robbins Met al.Postexposure protection of non-human primates against a lethal Ebola virus challenge with RNA interference: a proof-of-concept study. Lancet2010;375:1896–1905.
  • Olinger GG Jr, Pettitt J, Kim Det al.Delayed treatment of Ebola virus infection with plant-derived monoclonal antibodies provides protection in rhesus macaques. Proc Natl Acad Sci USA2012;109:18030–18035.
  • Michelow IC, Lear C, Scully Cet al.High-dose mannose-binding lectin therapy for Ebola virus infection. J Infect Dis2011;203:175–179.
  • Bray M, Driscoll J, Huggins JW.Treatment of lethal Ebola virus infection in mice with a single dose of an S-adenosyl-l-homocysteine hydrolase inhibitor. Antiviral Res2000;45:135–147.
  • Warren TK, Warfield KL, Wells Jet al.Antiviral activity of a small-molecule inhibitor of filovirus infection. Antimicrob Agents Chemother2010;54:2152–2159.
  • Kinch MS, Yunus AS, Lear Cet al.FGI-104: a broad-spectrum small molecule inhibitor of viral infection. Am J Transl Res2009;1:87–98.
  • Aman MJ, Kinch MS, Warfield Ket al.Development of a broad-spectrum antiviral with activity against Ebola virus. Antiviral Res2009;83:245–251.
  • Madrid PB, Chopra S, Manger IDet al.A systematic screen of FDA-approved drugs for inhibitors of biological threat agents. PLoS ONE2013;8:e60579.
  • Panchal RG, Reid SP, Tran JPet al.Identification of an antioxidant small-molecule with broad-spectrum antiviral activity. Antiviral Res2012;93:23–29.
  • Chang J, Warren TK, Zhao Xet al.Small molecule inhibitors of ER alpha-glucosidases are active against multiple hemorrhagic fever viruses. Antiviral Res2013;98:432–440.
  • Cote M, Misasi J, Ren Tet al.Small molecule inhibitors reveal Niemann-Pick C1 is essential for Ebola virus infection. Nature2011;477:344–348.
  • Hensley LE, Stevens EL, Yan SBet al.Recombinant human activated protein C for the postexposure treatment of Ebola hemorrhagic fever. J Infect Dis2007;196( Suppl 2):S390–S399.
  • Radoshitzky SR, Warfield KL, Chi Xet al.Ebolavirus delta-peptide immunoadhesins inhibit marburgvirus and ebolavirus cell entry. J Virol2011;85:8502–8513.
  • Huggins J, Zhang ZX, Bray M.Antiviral drug therapy of filovirus infections: S-adenosylhomocysteine hydrolase inhibitors inhibit Ebola virus in vitro and in a lethal mouse model. J Infect Dis1999;179( Suppl 1):S240–S247.
  • Warfield KL, Panchal RG, Aman MJ, Bavari S.Antisense treatments for biothreat agents. Curr Opin Mol Ther2006;8:93–103.
  • Warfield KL, Swenson DL, Olinger GGet al.Gene-specific countermeasures against Ebola virus based on antisense phosphorodiamidate morpholino oligomers. PLoS Pathog2006;2:e1.
  • Rettig GR, Behlke MA.Progress toward in vivo use of siRNAs-II. Mol Ther2012;20:483–512.
  • Subramanya S, Kim SS, Abraham Set al.Targeted delivery of small interfering RNA to human dendritic cells to suppress dengue virus infection and associated proinflammatory cytokine production. J Virol2010;84:2490–2501.
  • Block TM, Jordan R.Iminosugars as possible broad spectrum anti hepatitis virus agents: the glucovirs and alkovirs. Antivir Chem Chemother2001;12:317–325.
  • Block TM, Lu X, Mehta ASet al.Treatment of chronic hepadnavirus infection in a woodchuck animal model with an inhibitor of protein folding and trafficking. Nat Med1998;4:610–614.
  • Block TM, Lu X, Platt FMet al.Secretion of human hepatitis B virus is inhibited by the imino sugar N-butyldeoxynojirimycin. Proc Natl Acad Sci USA1994;91:2235–2239.
  • Bridges CG, Ahmed SP, Kang MS, Nash RJ, Porter EA, Tyms AS.The effect of oral treatment with 6-O-butanoyl castanospermine (MDL 28,574) in the murine zosteriform model of HSV-1 infection. Glycobiology1995;5:249–253.
  • Datema R, Romero PA, Rott R, Schwarz RT.On the role of oligosaccharide trimming in the maturation of Sindbis and influenza virus. Arch Virol1984;81:25–39.
  • Chang J, Wang L, Ma Det al.Novel imino sugar derivatives demonstrate potent antiviral activity against flaviviruses. Antimicrob Agents Chemother2009;53:1501–1508.
  • Qu X, Pan X, Weidner Jet al.Inhibitors of endoplasmic reticulum α-glucosidases potently suppress hepatitis C virus virion assembly and release. Antimicrob Agents Chemother2011;55:1036–1044.
  • Dwek RA, Butters TD, Platt FM, Zitzmann N.Targeting glycosylation as a therapeutic approach. Nat Rev Drug Discov2002;1:65–75.
  • Chang J, Warren TK, Zhao Xet al.Small molecule inhibitors of ER alpha-glucosidases are active against multiple hemorrhagic fever viruses. Antiviral Res2013;98:432–440.
  • Shurtleff AC, Nguyen TL, Kingery DA, Bavari S.Therapeutics for filovirus infection: traditional approaches and progress towards in silico drug design. Expert Opin Drug Discov2012;7:935–954.
  • Noble CG, Chen YL, Dong Het al.Strategies for development of dengue virus inhibitors. Antiviral Res2010;85:450–462.
  • Chatel-Chaix L, Germain MA, Gotte M, Lamarre D.Direct-acting and host-targeting HCV inhibitors: current and future directions. Curr Opin Virol2012;2:588–598.
  • de Clercq E.Antivirals and antiviral strategies. Nat Rev Microbiol2004;2:704–720.
  • Khattab MA.Targeting host factors: a novel rationale for the management of hepatitis C virus. World J Gastroenterol2009;15:3472–3479.
  • Inoue K, Umehara T, Ruegg UTet al.Evaluation of a cyclophilin inhibitor in hepatitis C virus-infected chimeric mice in vivo. Hepatology2007;45:921–928.
  • Gallay PA.Cyclophilin inhibitors: a novel class of promising host-targeting anti-HCV agents. Immunol Res2012;52:200–210.
  • Furuta Y, Takahashi K, Shiraki Ket al.T-705 (favipiravir) and related compounds: novel broad-spectrum inhibitors of RNA viral infections. Antiviral Res2009;82:95–102.
  • Ikeda M, Kato N.Life style-related diseases of the digestive system: cell culture system for the screening of anti-hepatitis C virus (HCV) reagents: suppression of HCV replication by statins and synergistic action with interferon. J Pharmacol Sci2007;105:145–150.
  • Bray M, Raymond JL, Geisbert T, Baker RO.3-deazaneplanocin A induces massively increased interferon-alpha production in Ebola virus-infected mice. Antiviral Res2002;55:151–159.
  • Ahmed SP, Nash RJ, Bridges CGet al.Antiviral activity and metabolism of the castanospermine derivative MDL 28,574, in cells infected with herpes simplex virus type 2. Biochem Biophys Res Commun1995;208:267–273.
  • Taylor DL, Fellows LE, Farrar GHet al.Loss of cytomegalovirus infectivity after treatment with castanospermine or related plant alkaloids correlates with aberrant glycoprotein synthesis. Antiviral Res1988;10:11–26.
  • Lu X, Lu Y, Geschwindt R, Dwek RA, Block TM.Hepatitis B virus MHBs antigen is selectively sensitive to glucosidase-mediated processing in the endoplasmic reticulum. DNA Cell Biol2001;20:647–656.
  • Mehta A, Carrouee S, Conyers Bet al.Inhibition of hepatitis B virus DNA replication by imino sugars without the inhibition of the DNA polymerase: therapeutic implications. Hepatology2001;33:1488–1495.
  • Fischer PB, Collin M, Karlsson GBet al.The alpha-glucosidase inhibitor N-butyldeoxynojirimycin inhibits human immunodeficiency virus entry at the level of post-CD4 binding. J Virol1995;69:5791–5797.
  • Fischer PB, Karlsson GB, Dwek RA, Platt FM.N-butyldeoxynojirimycin-mediated inhibition of human immunodeficiency virus entry correlates with impaired gp120 shedding and gp41 exposure. J Virol1996;70:7153–7160.
  • Taylor DL, Kang MS, Brennan TM, Bridges CG, Sunkara PS, Tyms AS.Inhibition of alpha-glucosidase I of the glycoprotein-processing enzymes by 6-O-butanoyl castanospermine (MDL 28,574) and its consequences in human immunodeficiency virus-infected T cells. Antimicrob Agents Chemother1994;38:1780–1787.
  • Taylor DL, Sunkara PS, Liu PS, Kang MS, Bowlin TL, Tyms AS.6-0-butanoylcastanospermine (MDL 28,574) inhibits glycoprotein processing and the growth of HIVs. AIDS1991;5:693–698.
  • Kaluza G, Repges S, McDowell W.The significance of carbohydrate trimming for the antigenicity of the Semliki Forest virus glycoprotein E2. Virology1990;176:369–378.
  • Steinmann E, Whitfield T, Kallis Set al.Antiviral effects of amantadine and iminosugar derivatives against hepatitis C virus. Hepatology2007;46:330–338.
  • Courageot MP, Frenkiel MP, dos Santos CD, Deubel V, Despres P.Alpha-glucosidase inhibitors reduce dengue virus production by affecting the initial steps of virion morphogenesis in the endoplasmic reticulum. J Virol2000;74:564–572.
  • Schul W, Liu W, Xu HY, Flamand M, Vasudevan SG.A dengue fever viremia model in mice shows reduction in viral replication and suppression of the inflammatory response after treatment with antiviral drugs. J Infect Dis2007;195:665–674.
  • Whitby K, Pierson TC, Geiss Bet al.Castanospermine, a potent inhibitor of dengue virus infection in vitro and in vivo. J Virol2005;79:8698–8706.
  • Chang J, Schul W, Butters TDet al.Combination of alpha-glucosidase inhibitor and ribavirin for the treatment of dengue virus infection in vitro and in vivo. Antiviral Res2011;89:26–34.
  • Perry ST, Buck MD, Plummer EMet al.An iminosugar with potent inhibition of dengue virus infection in vivo. Antiviral Res2013;98:35–43.
  • Chang J, Schul W, Yip A, Xu X, Guo JT, Block TM.Competitive inhibitor of cellular alpha-glucosidases protects mice from lethal dengue virus infection. Antiviral Res2011;92:369–371.
  • Gu B, Mason P, Wang Let al.Antiviral profiles of novel iminocyclitol compounds against bovine viral diarrhea virus, West Nile virus, dengue virus and hepatitis B virus. Antivir Chem Chemother2007;18:49–59.
  • Miller JL, Lachica R, Sayce ACet al.Liposome-mediated delivery of iminosugars enhances efficacy against dengue virus in vivo. Antimicrob Agents Chemother2012;56:6379–6386.
  • Rathore AP, Paradkar PN, Watanabe Set al.Celgosivir treatment misfolds dengue virus NS1 protein, induces cellular pro-survival genes and protects against lethal challenge mouse model. Antiviral Res2011;92:453–460.
  • Watanabe S, Rathore AP, Sung Cet al.Dose- and schedule-dependent protective efficacy of celgosivir in a lethal mouse model for dengue virus infection informs dosing regimen for a proof of concept clinical trial. Antiviral Res2012;96:32–35.
  • Wu SF, Lee CJ, Liao CL, Dwek RA, Zitzmann N, Lin YL.Antiviral effects of an iminosugar derivative on flavivirus infections. J Virol2002;76:3596–3604.
  • Fukushi M, Yoshinaka Y, Matsuoka Yet al.Monitoring of S protein maturation in the endoplasmic reticulum by calnexin is important for the infectivity of severe acute respiratory syndrome coronavirus. J Virol2012;86:11745–11753.
  • Bolt G, Pedersen IR, Blixenkrone-Moller M.Processing of N-linked oligosaccharides on the measles virus glycoproteins: importance for antigenicity and for production of infectious virus particles. Virus Res1999;61:43–51.
  • Schlesinger S, Malfer C, Schlesinger MJ.The formation of vesicular stomatitis virus (San Juan strain) becomes temperature-sensitive when glucose residues are retained on the oligosaccharides of the glycoprotein. J Biol Chem1984;259:7597–7601.
  • Silber AM, Candurra NA, Damonte EB.The effects of oligosaccharide trimming inhibitors on glycoprotein expression and infectivity of Junin virus. FEMS Microbiol Lett1993;109:39–43.
  • Saito T, Yamaguchi I.Effect of glycosylation and glucose trimming inhibitors on the influenza A virus glycoproteins. J Vet Med Sci2000;62:575–581.
  • Block TM, Deshmane S, Masonis J, Maggioncalda J, Valyi-Nagi T, Fraser NW.An HSV LAT null mutant reactivates slowly from latent infection and makes small plaques on CV-1 monolayers. Virology1993;192:618–630.
  • Stanley P.Glycosylation mutants of animal cells. Annu Rev Genet1984;18:525–552.
  • Stanley P, Caillibot V, Siminovitch L.Selection and characterization of eight phenotypically distinct lines of lectin-resistant Chinese hamster ovary cell. Cell.1975;6:121–128.
  • Hammond C, Braakman I, Helenius A.Role of N-linked oligosaccharide recognition, glucose trimming, and calnexin in glycoprotein folding and quality control. Proc Natl Acad Sci USA1994;91:913–917.
  • Hebert DN, Foellmer B, Helenius A.Glucose trimming and reglucosylation determine glycoprotein association with calnexin in the endoplasmic reticulum. Cell1995;81:425–433.
  • Bijvoet AG, van de Kamp EH, Kroos MAet al.Generalized glycogen storage and cardiomegaly in a knockout mouse model of Pompe disease. Hum Mol Genet1998;7:53–62.
  • Raben N, Lu N, Nagaraju Ket al.Conditional tissue-specific expression of the acid alpha-glucosidase (GAA) gene in the GAA knockout mice: implications for therapy. Hum Mol Genet2001;10:2039–2047.
  • Raben N, Nagaraju K, Lee Eet al.Targeted disruption of the acid alpha-glucosidase gene in mice causes an illness with critical features of both infantile and adult human glycogen storage disease type II. J Biol Chem1998;273:19086–19092.
  • Durantel D.Celgosivir, an alpha-glucosidase I inhibitor for the potential treatment of HCV infection. Curr Opin Investig Drugs2009;10:860–870.
  • Fischl MA, Resnick L, Coombs Ret al.The safety and efficacy of combination N-butyl-deoxynojirimycin (SC-48334) and zidovudine in patients with HIV-1 infection and 200–500 CD4 cells/mm3. J Acquir Immune Defic Syndr1994;7:139–147.
  • Wu SF, Lee CJ, Liao CL, Dwek RA, Zitzmann N, Lin YL.Antiviral effects of an iminosugar derivative on flavivirus infections. J Virol2002;76:3596–3604.
  • Bridges CG, Brennan TM, Taylor DL, McPherson M, Tyms AS.The prevention of cell adhesion and the cell-to-cell spread of HIV-1 in vitro by the alpha-glucosidase 1 inhibitor, 6-O-butanoyl castanospermine (MDL 28574). Antiviral Res1994;25:169–175.
  • Moorthy NS, Ramos MJ, Fernandes PA.Studies on alpha-glucosidase inhibitors development: magic molecules for the treatment of carbohydrate mediated diseases. Mini Rev Med Chem2012;12:713–720.
  • Hakamata W, Kurihara M, Okuda H, Nishio T, Oku T.Design and screening strategies for alpha-glucosidase inhibitors based on enzymological information. Curr Top Med Chem2009;9:3–12.
  • Kajimoto T, Node M.Inhibitors against glycosidases as medicines. Curr Top Med Chem2009;9:13–33.
  • Cox T, Lachmann R, Hollak C, Aerts Jet al.Novel oral treatment of Gaucher's disease with N-butyldeoxynojirimycin (OGT 918) to decrease substrate biosynthesis. Lancet2000;355:1481–1485.
  • Oulaidi F, Front-Deschamps S, Gallienne Eet al.Second-generation iminoxylitol-based pharmacological chaperones for the treatment of Gaucher disease. Chem Med Chem2011;6:353–361.
  • Jacob GS, Bryant ML.Iminosugar glycosylation inhibitors as anti-HIV agents. Perspect Drug Discov Des1993;1:211–224.
  • Vaughn DW, Green S, Kalayanarooj Set al.Dengue viremia titer, antibody response pattern, and virus serotype correlate with disease severity. J Infect Dis2000;181:2–9.
  • Block TM, Jungkind D, Crowell R, Denison M, Walsh LR ( ed.). Innovations in antiviral development and the detection of virus infections.New York: Plenum Press, 1992.
  • Huovila AP, Eder AM, Fuller SD.Hepatitis B surface antigen assembles in a post-ER, pre-Golgi compartment. J Cell Biol1992;118:1305–1320.
  • Penin F, Dubuisson J, Rey FA, Moradpour D, Pawlotsky JM.Structural biology of hepatitis C virus. Hepatology2004;39:5–19.
  • Simon K, Lingappa VR, Ganem D.Secreted hepatitis B surface antigen polypeptides are derived from a transmembrane precursor. J Cell Biol1988;107( 6 Pt 1):2163–2168.
  • Werr M, Prange R.Role for calnexin and N-linked glycosylation in the assembly and secretion of hepatitis B virus middle envelope protein particles. J Virol1998;72:778–782.
  • Dubuisson J, Rice CM.Hepatitis C virus glycoprotein folding: disulfide bond formation and association with calnexin. J Virol1996;70:778–786.
  • Branza-Nichita N, Durantel D, Carrouee-Durantel S, Dwek RA, Zitzmann N.Antiviral effect of N-butyldeoxynojirimycin against bovine viral diarrhea virus correlates with misfolding of E2 envelope proteins and impairment of their association into E1-E2 heterodimers. J Virol2001;75:3527–3536.
  • Branza-Nichita N, Lazar C, Durantel D, Dwek RA, Zitzmann N.Role of disulfide bond formation in the folding and assembly of the envelope glycoproteins of a pestivirus. Biochem Biophys Res Commun2002;296:470–476.
  • Norton PA, Menne S, Sinnathamby Get al.Glucosidase inhibition enhances presentation of de-N-glycosylated hepatitis B virus epitopes by major histocompatibility complex class I in vitro and in woodchucks. Hepatology2010;52:1242–1250.