throbber
® Check for updates
`
`Review
`
`Antiviral Chemistry & Chemotherapy 2011; 22:23-49 (doi: 10.3851/IMP1797)
`
`Nucleotide prodrugs for HCV therapy
`
`Michael J Sofia'*
`
`'Pharmasset, Inc., Princeton, NJ, USA
`
`*Corresponding author e-mail: michael.sofia@pharmasset.com
`
`
`HCV infection is a significant worldwide health problem
`and is a major cause of hepatocellular carcinoma. The
`current standard of care, interferon and ribavirin, is only
`effective against a proportion of the patient population
`infected with HCV. To address the shortcomings of exist-
`ing therapy, the development of direct acting antiviral
`agents is under investigation. The HCV RNA dependent
`RNA polymeraseis an essential enzyme for viral replica-
`tion and is therefore a logical target against which to
`develop novel anti-HCV agents. Nucleosides have been
`shown to be effective as antiviral agents for other viral
`diseases and therefore, have been investigated as inhibi-
`tors of HCV replication. The development of prodrugs
`of nucleoside 5’-monophosphates has been pursued
`
`to address limitations associated with poor nucleoside
`phosphorylation. This is required to produce the nucle-
`oside 5’-triphosphate which is the anabolite that is the
`actual
`inhibitor of the polymerase enzyme. Prodrugs of
`nucleoside 5’-monophosphates have been developed that
`enable their delivery into cells and in vivo into the liver.
`The implementation of these prodrug strategies has ulti-
`mately led to the identification of several prodrugs of
`nucleoside 5’-monophosphates that are potent
`inhibi-
`tors of HCV replication in vitro. They have progressed into
`the clinic and the early data demonstrate greatly reduced
`viral load levels in HCV-infected patients. This review will
`survey the state of nucleotide prodrugs for the treatment
`of HCV.
`
`Introduction
`
`HCV is known to have infected approximately 180
`million individuals worldwide [1]. It is estimated that
`of those infected with HCV approximately 80% will
`develop chronic liver disease and a significant propor-
`tion of those infected will eventually developliver cir-
`rhosis and subsequently hepatocellular carcinoma [2].
`HCVis a single-stranded, positive sense RNA virus of
`the Flaviviridae. Six major viral genotypes with over
`100 viral subtypes have beenidentified for HCV. Geno-
`types la and 1b are the most prevalent genotypes in
`the western world with genotypes 2 and 3 comprising
`20-30% of this population. HCV genotypes 2-6 pre-
`dominate in the developing world [3,4]. Because HCV
`replicates in the cytoplasm ofinfected cells by a mem-
`brane associated replication complex and the virus has
`an RNA genome with no DNAintermediate during
`replication, no genomic templates are stably integrated
`into the host genome. Therefore, virological cures are
`possible for HCV patients. This is in contrast to other
`viruses such as HIV and HBV wheretheviral genomeis
`integrated into the host DNA anda virological cureis
`considered remote. However, for HCV-infected patients
`virological cures are madedifficult due to the high rate
`of HCVviral replication and by the high spontaneous
`
`©2011 International Medical Press 1359-6535 (print) 2040-2066 (online)
`
`mutation rate of the virus. This high mutationrate is a
`result of poor replication fidelity exhibited by the HCV
`polymerase and an apparentlack of proof reading[4].
`The current therapy for treating chronic HCV infec-
`tion consists of regular injections of o-interferon (IFN)
`with daily oral administration of ribavirin (RBV). This
`standard of care (SOC) regimen does notact by directly
`attacking the virus but functions by boosting the host
`immuneresponse. For genotype 1 patients regular IFN/
`RBV treatments for 48 weeksresult in only 40-50% of
`patients achievinga sustained virological response (SVR)
`indicative of a cure [5,6]. However, for genotype 2 and
`3 patients the SVRrates can beas high as 75%. It is also
`knownthat subpopulations which include individuals of
`African ancestry tend to respond less well to IFN/RBV
`treatments [7]. Recent genome-wideassociation studies
`have shownthat a single nucleotide polymorphism 3kb
`upstream of the IL28B genecorrelates with a significant
`difference in response to IFN therapy [8]. IL28B which
`encodesthe typeIII interferon IFN-A-3, is knownto be
`upregulated by IFNs and by RNAviralinfections.It has
`been shown that HCVpatients who harbour a TT or TC
`allele in their IL28B genetend to respondless well to IFN/
`RBVtreatment than do those having the CC genotype.
`
`Gilead 2005
`I-MAK v. Gilead
`IPR2018-00125
`
`

`

`MJ Sofia
`
`Figure 1. Model of HCV NS5B RNA complex
`
`
`
`Ribbon structure of the HCV NS5B RNA dependent RNA polymerase showing the
`Palm, Finger and Thumb domainstypical of a RNA polymerase.Also depicted is a
`bound template-primer strand of RNA andthe nucleotide bindingsite.
`
`Patients who choose to undergo IFN/RBV therapy face
`not only the possibility of not responding to treatment
`but also must contendwith the potential for multiple and
`sometimes serious side-effects that include influenza-like
`symptoms, fatigue, hemolytic anaemia and depression.
`The intolerable side effects can result in a high rate of
`drug discontinuations. Consequently, the modest cure
`rates and subpopulation differences combined with the
`side-effect profile for SOC have prompted an urgency to
`develop alternative novel, safe and effective therapies.
`As in the case of other viral diseases, the development
`of direct acting antivirals (DAAs) has become a focus.
`Development of small molecule agents to attack essen-
`tial viral proteins has the potential benefit of reducing
`toxicities and side effects associated with manipulating
`host functions and hopefully such DAA therapies will
`not have the intolerable side effects exhibited by cur-
`rent SOC.
`The push to identify small molecule DAAshasalso
`promptedthe discussion aroundthe possibility of elimi-
`nating or at least reducing the use of IFN/RBV from
`treatment regimens. Although clinical development of
`first generation DAAs has focused on combinations
`with SOC in the hope of both shortening duration of
`treatment and increasing the cure rate, the long-term
`desire is to completely eliminate the use of IFN/RBV
`from treatment regimens. Clearly this is an aspirational
`goal and a goalthat can only be achieved if an immune
`component of therapy is not absolutely required to
`eliminate those vestiges of undetectable virus
`[9].
`In addition, such a lofty goal can be realised if small
`
`24
`
`molecule DAAseither alone or more probably in com-
`bination can drive viral loads to undetectable limits and
`maintain those undetectable levels over the necessary
`course of therapy andafter cessation of treatment with-
`out having viral breakthrough resulting from the emer-
`genceofresistant virus. As has been shown with HIV
`highly active antiretroviral therapy (HAART), the HCV
`treatment paradigm will likely require combinations of
`anti-HCV agents [7]. The desire is to suppress virus as
`rapidly and completely as possible in order to give the
`body’s natural immunesystem the opportunity to clear
`residual virus and to hold back emergence of resistant
`virus. However, what will comprise those ideal com-
`binations of DAAsis yet to be determined and studies
`to clarify this question are under active discussion and
`investigation.
`HCVhasa 9.6 kb genomeof positive-stranded RNA.
`This genome encodes a precursor polyprotein thatis
`processed into 10 functional proteins: three structural
`proteins and seven non-structural proteins [10]. Sev-
`eral of the non-structural proteins have been the focus
`of intensive efforts to identify small molecule DAA
`agents as inhibitors of HCV replication. Of the seven
`non-structural proteins, molecules that inhibit the func-
`tions of the NS3/4 protease, NS4A, NS4B, NSSA and
`NSSB RNA dependent RNA polymerase (RdRp) have
`advanced to the clinic [11-15]. The most advanced
`agents are the NS3/4 protease inhibitors telaprevir and
`boceprevir. Each of these compounds has completed
`Phase III clinical
`investigation and both have been
`shownto be efficaciousin treating HCV infection when
`given in combination with SOC. However, each of these
`first generation protease inhibitors suffers from the lack
`of genotype coverage, undesired side effects, that may
`limit their usage, and the early emergence of resistant
`virus. It is therefore not surprising that even with the
`potential benefits of these first generation DAAs, ware-
`housingof patients by physicians occursin order to wait
`for approval of moreeffective and tolerable agents.
`The HCV RNA dependent RNA polymerase is an
`~68 kd protein that has the typical palm-finger-thumb
`structural motif found in many viral polymerases(Fig-
`ure 1) [16,17]. HCV polymeraseis an essential enzyme
`involved in RNA replication. Phylogenetic analysis
`shows a 65% homology of HCV RdRpacross genotypes
`and an 80% homologywithin a particular genotype [3].
`The HCV polymeraseactivesite is located in the palm
`domain where the conserved aspartic acid residue-con-
`taining GDD motifis located [18]. This conserved GDD
`motif is commonto viral polymerases in general [18].
`Through a divalent metal ion (Mg** or Mn**) the GDD
`motif functions to coordinate the bindingof the ribonu-
`cleoside triphosphate. The HCV polymerase catalyzes
`the addition of a single ribonucleoside triphosphate
`monomerto the 3’-end of the growing RNAchain by the
`
`©2011 International Medical Press
`
`

`

`formation of a 3’,5’-phosphodiester linkage. To accom-
`plish this process, the polymerase must simultaneously
`bind a template RNAstrand, a primer RNAstrand and
`a ribonucleoside triphosphate monomer[19,20]. There-
`fore, investigation of nucleoside analoguesis a rational
`choice for the development of inhibitors of the HCV
`NSS5B polymerase.
`Ofall the DAAsunderclinical investigation, nucleo-
`side/nucleotide NS5B polymerase inhibitors hold the
`promise of pan-genotype coverage and a high barrier
`to developmentofresistant virus. As in the case of HIV
`infection where nucleosides have becomethe backbone
`of therapy (for example, TRUVADA® and Combivir®),
`HCVnucleosides/nucleotides are positioned to assume
`a similar role. To date, only nucleosides/nucleotides
`have demonstrated broad genotype coverage both in
`the laboratory and in humanclinical studies [21]. In
`addition, to date, no pre-emergentresistant virus has
`been detected in clinical studies [22]. It is for these
`reasons that nucleosides/nucleotides are positioned to
`play a prominentrole in developing HCV treatment
`paradigms.
`The HCV polymerase has been shown to be a
`uniquely selective polymeraseas it relates to the devel-
`opment of nucleoside/nucleotide inhibitors. Over
`the last 10 years only two broad classes of nucleo-
`sides have emerged as inhibitors of this polymerase
`[23-25]. These include the 2’-methyl and the 4’-azido
`classes (Figure 2). However,
`these classes and sub-
`groups within these classes have clearly differentiated
`themselves in preclinical and clinical studies. This
`differentiation is exemplified in their viral selectivity,
`viral resistance, overall safety andclinical efficacy pro-
`files. Resistance associated with the 2’-methyl class of
`nucleosides is associated with the $282T amino acid
`alteration located in the finger domain of the HCV
`polymerase [26-29]. This mutation has been shown to
`be difficult to raise in vitro and has not been detected
`as a pre-existing mutation in clinical
`isolates [22].
`Similarly, for the 4’-azido class, the S96T amino acid
`alteration has been identified in vitro but has not been
`observedin the clinic [27].
`Implementation of a prodrug strategy has played
`a prominent role in the development of a number of
`nucleosides and nucleotides for the treatment of HCV
`infection [23,30]. These prodrugs have been developed
`to overcome, not only, bioavailability and stability
`issues but also to address key anabolism limitations
`important to nucleoside activation. Simple ester prod-
`rugs of both the 2’-methylcytidine, 2’-c-fluoro-2’-B-C-
`methylcytidine and 4’-azidocytidine nucleosides were
`developed to overcome both bioavailability issues and
`to curb undesirable metabolism [21,31,32]. Prodrugs of
`the phosphate group of nucleoside 5’-monophosphates
`were developed to address not only bioavailability
`
`Antiviral Chemistry & Chemotherapy 22.1
`
`Nucleotide prodrugs for HCV therapy
`
`Figure 2. Two major classes of nucleosides that are known to
`be inhibitors of HCV polymerase.
`
`2’-Methyl class
`
`4’-Azido class
`
`HO
`
`2
`
`BASE
`
`HO
`
`=X
`
`Xe OH, F
`
`BASE
`
`Ho“
`
`\ 9
`
`N3*
`HO
`
`=X
`
`X = OH, F
`
`These classes include the 2’-methyl and the 4’-azido ribosides.
`
`issues but also poor in vitro and in vivo conver-
`sion of the parent nucleoside to the active nucleoside
`5’-triphosphate [33]. Because nucleosides must be con-
`verted to their 5’-triphospates to be active as inhibitors
`of the HCV polymerase, they need to undergo a series
`of phosphorylation steps catalyzed by three separate
`kinases (Figure 3). These kinases convert the nucleoside
`first to the monophosphate, then to the diphosphate
`and finally to the active triphosphate. However, it is
`not uncommonthat in the phosphorylation cascade, a
`nucleoside or its corresponding mono- or diphosphate
`is a poor substrate for one of the kinases. In particular,
`it is the first kinase in the phosphorylation cascade that
`is generally the most substrate selective. Therefore, it
`is not unusual that bypassingthefirst kinase results in
`achieving high levels of the active triphosphate. Because
`nucleoside monophosphates are enzymatically dephos-
`phorylated and negatively charged, they do notreadily
`enter cells and therefore are not desirable as drug can-
`didates. To overcomethe limitations of administering
`a nucleoside monophosphate-containing agent, pro-
`drugs of the 5’-monophosphate nucleoside have been
`employed. Prodrugs of nucleoside monophosphates
`have been known for many years and a number of
`phosphate prodrug strategies have been developed to
`address the need to deliver a 5’-monophosphate nucleo-
`side into the cell [33-35]. However, there have been few
`examples where a nucleotide prodrug has been shown
`to deliver the corresponding 5’-monophosphate in vivo
`to the desired site of action [33]. Often the prodrug
`moiety decomposes prior to achieving its objective
`because of either chemical or enzymatic instability in
`the gastrointestinal tract and/or plasma.
`The development of a nucleoside phosphate prod-
`rug useful for the treatment of HCV faces several chal-
`lenges. The nucleoside phosphate prodrug must have
`sufficient chemical stability to be formulated for oral
`
`25
`
`

`

`M$ Sofia
`
`Figure 3. Nucleoside kinase activation pathway
`
`BASE
`[BASE
`ONo.
`[BASE|
`o.
`HO
`
`
`™ od _Kinase2aKinase 1 ad
`be X
`\ bx
`\__
`Y
`HO
`OY
`
`oo
`
`
`
`HO
`
`|BASE]
`_Kinase33
`= Phosphate
`
`Nucteoside kinase activation pathwayresulting in the nucleoside triphosphate whichis the active substrate for a polymerase allowing incorporation of the nucleoside
`or nucleoside analogueinto the growing RNA chain andthusresulting in inhibition of virus replication. Examples of kinases 1, 2, and 3 include deoxycytidine kinase
`(dCk), nucleoside monophosphate kinase (YMPK) and nucleoside disphosphate kinase (NDPK), respectively,
`
`administration. It must be stable to conditions of the
`gastrointestinal tract such that the prodrug reaches the
`site of absorption intact. The prodrug must have good
`absorption properties and must not undergo apprecia-
`ble enzymatic degradation during the absorption phase.
`Once absorbed, the prodrug needsto havesufficient sta-
`bility in the bloodin order to reach the target organ: for
`example theliver in the case of HCV. The prodrug must
`then be transported into hepatocytes andrelease the free
`5’-monophosphate nucleoside which can subsequently
`be converted to the active triphosphate derivative. Since
`HCVis a disease of the liver, and the liver is the first
`organ the prodrug encounters after absorption, HCVis
`an ideal disease for which to develop a targeted nucle-
`otide prodrug strategy. Consequently, several of these
`nucleoside monophosphate prodrugs have advanced
`to the clinic and have demonstrated proof of concept
`for treating HCV. Here, the application of phosphate
`prodrugs in the development of nucleotide inhibitors
`of HCV andthe status of nucleotide prodrugs under
`investigation for the treatment of HCV infection will
`be reviewed.
`
`Nucleotide phosphoramidates
`Nucleotide phosphoramidates were first disclosed
`by McGuigan et al. [36] as a prodrug strategy to
`deliver a nucleoside 5’-monophosphateforthe treat-
`ment of HIV and cancer. The structure of a nucle-
`otide phosphoramidatetypically consists of a nucleo-
`side 5’-monophosphate where the phosphate group
`is masked by appending an aryloxy group (usually a
`phenol) and an o-amino acid ester (Figure 4); how-
`ever, other related constructs have also appeared. The
`
`26
`
`phosphate groupis ultimately revealed by a sequence
`of enzymatic and chemical steps that requires either
`carboxyesterase or cathepsin A to cleave the terminal
`amino acid ester, intramolecular displacement of the
`phosphate phenoland then enyzmatic cleavage of the
`amino acid moiety by a phosphoramidaseorhistidine
`triad nucleotide-binding protein 1 (HINT 1) [37-39].
`It
`is believed that
`the phosphoramidate prodrug
`construct
`increases lipophilicity of the nucleoside
`5’-monophosphate and therefore increases cellular
`permeability and ultimately intracellular nucleotide
`concentrations. Since the phosphoramidate prodrug
`moiety contains a chiral phosphorus centre,
`issues
`arise with regard to development of a compoundthat
`consists of a mixture of isomers with implications
`arising from differential activity of each of the iso-
`mers, pharmacokinetics and manufacturing optimi-
`zation, etc. In addition, the typical phosphoramidate
`contains a phenolic substituent that is released during
`metabolism to the free monophosphate. Successful
`development also considers the metabolic release of
`this phenolic substituent. Selective examples employ-
`ing the phosphoramidate strategy to achieve kinase
`bypass for nucleoside inhibitors of HIV reverse tran-
`scriptase (RT) inhibitors, for example, ddA, d4T and
`d4A, showed that in vitro whole cell enhancement
`in potency could be achieved [40-42]. Although the
`phosphoramidate strategy was explored extensively to
`deliver nucleotides for the treatment of HIV and colon
`cancer [33,36], proof of concept in the clinic has yet
`to be reported. However, the phosphoramidate prod-
`rug approach has provento be a valuable strategy in
`the development of HCV nucleotide therapy.
`
`©2011 International Medical Press
`
`

`

`Figure 4. The phosphoramidate prodrug decomposition pathwaythatresults in the release of the nucleoside 5'-monophosphate
`
`Nucleotide prodrugs for HCV therapy
`
`Oo
`
`Ro
`
`OQ
`
`Ra
`
`Carboxyesterase \ R
`\ G
`¢ oe) “se
`fHoe)
`Ry
`9
`Cathepsin A
`H
`id
`Ar
`Ar
`
`
`
`Nucleotide phosphoramidate
`
`oO
`
`Spontaneous
`
`alae\-4
`
`A
`
`oO
`
`
`
`HINT-1
`
`h
`;
`(Phosphoramidase)
`
`4
`HO-F=0
`OH
`
`Ro
`
`a
`HN=F-o
`O
`
`|H
`
`oO
`
`G
`H
`
`2’-C-Methyl ribonucleotide phosphoramidates
`The 2’-C-methylcytidine nucleoside, NM107 (1; Fig-
`ure 5), was shown to be an inhibitor of HCV in cell
`culture (50% effective concentration [EC,,] =1.23 UM)
`and its triphosphate (3) was demonstrated to be a
`potent inhibitor of the HCV polymerase enzyme (50%
`inhibitory concentration [IC,,] =0.09-0.18 4M)acting
`as a nonobligate chain terminator [43]. NM107 also
`showed broad antiviral activity against not only HCV
`but also bovine virus diarrhoea virus (BVDV), yellow
`fever virus, dengue virus and West Nile virus [31]. To
`overcome bioavailability issues the 3’-valinate ester
`prodrug, NM283(valopicitabine; 2) [31,44], was taken
`into clinical development. In a Phase I monotherapy
`study, NM283 demonstrated proof of conceptdeliver-
`ing an ~1.2 log, [U/mlreductionin viral load at a dose
`of 800 mg twice daily given over 14 days. Unfortu-
`nately, NM283 was discontinued because of significant
`gastrointestinal toxicity in Phase II studies [24,30].
`Subsequent work on the development of the 2’-C-
`methyl class of nucleosides focused on 5’-phosphate
`nucleotide prodrugs. It was observed that the 2’-C-
`methylcytidine triphosphate (3) was highly active as
`an inhibitor of the NSSB polymerase, yet the parent
`
`Antiviral Chemistry & Chemotherapy 22.1
`
`nucleoside NM107 (1) was only modestly activein the
`whole cell based replicon assay. Studies had shown that
`NM107-triphosphate (3) formation was inefficient,
`particularly because of poor conversion of the nucleo-
`side to its monophosphate by 2’-deoxycytidine kinase
`[45]. Consequently, to circumvent this phosphoryla-
`tion problem and potentially improve the therapeutic
`index by increasing nucleoside triphosphate levels in
`the liver, a phosphoramidate prodrug approach was
`investigated [45]. This effort lead to the identification
`of phosphoramidate derivative 4 (Figure 5; EC,,<0.5
`uM) showing substantial increases in potency rela-
`tive to NM283[45]. The activity of compound 4 cor-
`related with the levels of triphosphate produced in
`humanhepatocytes and these levels were shown to be
`muchhigher than that seen with NM283 (2). In vivo
`studies assessing liver nucleoside triphosphate levels
`after oral administration in hamsters showedlowtri-
`phosphate concentrations only twofold higher than
`obtained with NM283. Since substantial
`liver tri-
`phosphatelevels were seen after subcutaneous admin-
`istration iz vivo and the compounds were shown to
`be stable in simulated gastric fluid, it was concluded
`that low oral bioavailability or metabolic degradation
`
`27
`
`

`

`M3 Sofia
`
`Figure 5. 2’-C-Methylcytidine nucleosides and nucleotide phosphoramidate prodrug inhibitors of HCV replication
`
`7X
`|
`vrN
`—
`HO
`O NY
`HO OA
`0
`QO
`NH2
`C
`.
`-
`OH
`ge
`O
`
`HO=OH |
`
`NH»
`
`NH»
`
`O
`
`NM107
`EC,,=1.23 uM
`1
`
`NM283
`2
`
`NH»
`
`oO
`O
`HO—P—o—P—o—P—o-*
`l
`l
`OH
`OH
`OH
`
`.0. Ny
`VV ~~
`toes
`
`F
`
`HO
`
`OH
`
`IC,,=0.09-0.18 uM
`3
`
`NHz
`
`NH2
`
`EC,=0.22 uM, CC,,=7 uM
`4
`
`EC,,=0.24 uM
`5
`
`opcing
`
`OH
`
`oO
`
`HO
`
`OH
`
`NTP (human hepatocytes) AUC,,,,=190 uMeh
`
`7
`
`sat
`
`0
`
`HO
`
`OH
`
`EC,=8.2 uM, CC,,=>100 1M
`NTP (human hepatocytes) AUC.
`‘o-ay= 1720 uMeh
`6
`
`
`
`AUC,area under the curve; CC,,, 50% cytotoxic concentration; EC,,, 50%effective concentration; IC,,, 50% inhibitory concentration.
`
`28
`
`©2011 International Medical Press
`
`

`

`Nucleotide prodrugs for HCV therapy
`
`in the intestine was the reason for the lack of oral
`efficacy [45].
`Anotherseries of 2’-C-methylcytidine phosphorami-
`date prodrugs having an acyloxyethylamino phospho-
`ramidate promoiety was studied (Figure 5) [46]. The
`phosphoramidate prodrug 5 provided up to a 30-fold
`improvement in HCV replicon potency over NM283
`(2), and it was also shown that this activity correlated
`to levels of nucleoside triphosphate in rat and human
`hepatocytes. However, when administeredorally to rats
`these phosphoramidates demonstrated no improvement
`in production of triphosphate in rat liver relative to
`NM283(2). These results put into question the oral
`bioavailability and conversion of these prodrugs to the
`nucleoside triphosphatein theliver.
`Phosphoramidate monoesters of 2’-C-methylcytidine
`were also explored (Figure 5) [45]. In this case the ami-
`date moiety was either an o-amino acid (6) or an acy-
`loxyethylamino substituent (7). Although phosphorami-
`date monoester 6 was shownto haveinferior replicon
`potencyrelative to its phenolic ester counterpart (<200-
`fold) it showed higher levels of triphosphate formation
`in human hepatocytes. Formation of nucleoside triphos-
`phate levels were observed in hepatocytes of various spe-
`cies for both 6 and 7 but the phosphoramidate monoester
`
`6 was shownto besuperiorto 7. In bothcases,liver levels
`of nucleoside triphosphate 3 were achieved in vivo after
`subcutaneous administration but not after oral adminis-
`tration suggesting a lack of oral bioavailability.
`Other 2’-C-methyl nucleosides containing purine
`bases were also investigated as inhibitors of HCV,
`including the 7-deaza-2’-C-methyladenosine derivative
`MK0608(8) which showed potentinhibition of HCV
`replication in vitro (EC,,=0.25 4M) and demonstrated
`SVR in an HCV-infected chimpanzee animal model
`[47,48]. Although MK0608 (8) was never progressed
`into clinical development, it did demonstrate the poten-
`tial of purine nucleosides as inhibitors of HCV. Sub-
`sequently, phosphoramidate prodrugs of 2’-C-methyl
`purine analogues werestudied to determineif improve-
`ment in potency could be achieved by kinase bypass.
`Although application of the phosphoramidate prodrug
`strategy was not successfulfor the adenosine derivative
`11 (Figure 6), its application to the guanosine analogue
`12 resulted in an 84-fold increasein activity in the rep-
`licon assay relative to the parent nucleoside (Figure 7)
`[49]. This result was rationalized by the observation
`that the guanosine triphosphate (13; IC,,=0.13 MM)
`was more potent as an inhibitor of the HCV polymer-
`ase than the adenosine triphosphate (10; IC,,=1.9
`
`
`
`Figure 6. 2’-C-Methyladenosine nucleoside and nucleotide phosphoramidate prodrug inhibitors of HCV replication
`
`EC,=0.25 uM, CC,,=>50 uM
`8
`
`EC,,=0.3 uM, CC,,=>50 uM
`9
`
`HO
`
`OH
`
`NH2
`
`NH»
`i
`hs
`“ Xk
`9
`O
`OQ
`N
`N
`if
`i
`i
`U1
`neeas y=
`
`OH
`
`OH
`
`OH
`
`1
`
`al
`
`-
`
`HO
`
`OH
`
`IC,,=1.9 uM, EC,,=0.3 uM
`10
`
`Eto
`
`oO
`
`CHs
`
`a
`
`¢ jn
`Ayttoe
`TON i, Vy n=
`
`oe HO
`=.
`
`OH
`
`EC,,=0.25 pM, CC,,=>50 uM
`14
`
`
`
`CC,,, 50% cytotoxic concentration; EC,,, 50% effective concentration; IC,,, 50% inhibitory concentration.
`
`Antiviral Chemistry & Chemotherapy 22.1
`
`29
`
`

`

`MJ Sofia
`
`Figure 7. 2’-C-Methylguanosine nucleoside and nucleotide phosphoramidate prodrug inhibitors of HCV replication
`
`a
`
`O°
`N
`Cw
`NN
`at 2
`
`Lo.
`
`8
`e
`HO—P—O—P—o-P—o-*%
`
`OH
`
`OH
`
`OH
`
`0I
`
`O N
`
`H
`
`NC
`
`J
`iVey,
`
`HO~
`
`O
`
`HO
`
`OH
`
`EC,,=3.5 uM
`
`HO
`
`OH
`
`IC,,=0.13 uM
`13
`
`x
`0.
`
`CH
`
`pe,
`
`|
`
`;
`
`oc
`“Nn
`Ph
`=
`
`N
`
`¢ /
`
`N
`
`H
`
`f
`,
`4
`Oo
`Oo
`“Tt HNO"Ne
`co" °
`
`14 R.=CH,CH,Ph, R,=Ala EC,,=0.08 1M, CC,,=>100 uM
`15 R.=0-CIPh, R,=Val EC,,=0.43 uM, CC,,=>100 uM
`
`INX-08189
`
`EC,,=0.010 uM, CC,,=7 uM
`16
`
`IDX184
`
`EC,,=0.4 uM, CC,,>100 uM
`
`
`
`CC,,, 50% cytotoxic concentration; EC,,, 50% effective concentration; IC, 50% inhibitory concentration.
`
`uM), yet in the whole cell replicon assay the adenosine
`analogue (9; EC,,=0.3 4M; Figure 6) was more potent
`than the guanosine analogue (12; EC,,=3.5 1M;Figure
`7). Therefore, this finding coupled with the observa-
`tion that low levels of triphosphate were detected in
`cells for the guanosine nucleoside relative to that for
`the adenosine nucleoside hinted at poor phosphoryla-
`tion in the case of the guanosine nucleoside. The devel-
`opmentof the 2’-C-methylguanosine phosphoramidate
`derivative was further explored by systematically eval-
`uating structural modifications to the phosphorami-
`date moiety in an effort to achieve increased HCVrep-
`licon potency, plasma stability across multiple species,
`and appropriaterelative stability in intestinal andliver
`$9 preparations. Ultimately,
`these studies were able
`to identify 2’-C-methylguanosine phosphoramidate
`
`30
`
`prodrugs 14 and 15 (Figure 7) [50]. These prodrugs
`contained benzylor alkyl L-alanine and t-valine amino
`acid ester moieties and a naphthyl phosphate ester and
`exhibited a 10-30-fold enhancement in HCV repli-
`con potency with acceptable plasma andintestinal $9
`stability suitable for progression into in vivo studies.
`However, when mice were dosed orally in order to
`assessliver levels of the nucleoside triphosphate 13, the
`2’-C-methylguanosine phosphoramidates 14 and 15
`(Figure 7) produced substantial liver levels of triphos-
`phate but these levels were notsignificantly improved
`over that observed when mice were dosed with the par-
`ent guanosine nucleoside 12.
`Further investigation of the 2’-C-methylguanosine
`phosphoramidateseries led to the evaluation of substi-
`tution at the C-6-position of the guanosine base with the
`
`©2011 International Medical Press
`
`

`

`Nucleotide prodrugs for HCV therapy
`
`reported to date. In the whole cell HCV replicon assay
`intention of increasing lipophilicity and thus improving
`IDX184 (17) was shownto be a potentinhibitor of HCV
`cellular uptakerelative to the natural guanosine deriva-
`replication (EC,,=0.4 1M; CC,,>100 mM)and was also
`tive 12. This led to the identification of the C-6-O-methy!
`active in the genotype 2a JFH1 replicon (EC,,=0.6—11
`derivative INX-08189 (16; Figure 7) containing both a
`LM)[59]. At a concentration of 2.5 1M it cleared HCV
`neopentyl ester on the L-alanyl amino acid moiety and
`replicon RNA after 14 days of treatment. Like other
`a naphthyl ester on phosphorus of the phosphorami-
`2’-C-methyl nucleosides it was shown to select for the
`date {51]. In INX-08189 (16), in addition to metabolic
`NSSB $282T mutation in the HCVreplicon. In combi-
`conversion of the phosphoramidate pro-moiety to the
`nation with the protease inhibitor IDX320, IFN-o, or
`5’-monophosphate, the 6-O-methy] group of the purine
`RBV, IDX184 exhibited an additive or synergistic profile
`base is metabolized to the guanine base. INX-08189
`[60]. When administered orally to cynomolgus monkeys,
`(16) demonstrated exceptional potency in the HCV 1b
`IDX184 (17) produced high live triphosphate levels
`replicon assay (EC,,=0.01 uM; CC,,=7 UM), was active
`relative to oral administration of the parent nucleoside
`against genotype 1a and 2 replicons and produced sub-
`with what appears to be high hepatic extraction. Sub-
`stantial levels of the guanosine triphosphate 13 in pri-
`sequent studies in a HCV-1-infected chimpanzee model
`mary human hepatocytes over 48 h. The known 2’-C-
`showed that oral administration of IDX184 (17) at 10
`methyl nucleoside $282T mutant replicon was shown
`mg/kg over 3 days produced a medianviral load decline
`to be moderately resistant (3- to 10-fold) to INX-08189
`of approximately -2.3 log,, at day 3 and 4 [61]. Conse-
`(16) [52]. No difference in HCV replicon potency was
`quently, IDX184 (17) was progressed into the clinic and
`observed for each of the individual diastereoisomers of
`in a PhaseIa single ascending dose study was shown to
`INX-08189 (16), thus INX-08189 was advanced into
`be generally safe and well tolerated at oral doses from 5
`clinical development as a mixture of isomers at the
`mg to 100 mg [56]. PK assessment supportedalivertar-
`phosphoruscentre of the prodrug.
`geting mechanism for the compound.In a Phase Ib mon-
`In a single-ascending-dose Phase Ia study in healthy
`otherapy study in HCV genotype-1-infected patients,
`volunteers administered doses ranging from 3 mg to
`IDX184 (17) was administered at doses from 25 to 100
`100 mg, INX-08189 (16) was shown to be generally
`mg once a day for 3 days. Day 4 viral load assessment
`well tolerated at all doses with no drug-related serious
`showedthatat the highest dose of 100 mg,a -0.74 log,,
`adverse events and pharmacokinetics (PK) supporting
`IU/ml reduction in viral load was observed [62]. Fol-
`once daily oral dosing [53,54]. INX-08189 was pro-
`lowing the positive Phase I clinical results, a Phase II
`gressed into a Phase Ib study in treatment-naive geno-
`study wasinitiated in which IDX184 (17), at doses from
`type 1 HCV patients dosed once daily at either 9 mg
`50-200 mg, was combined with pegylated IFN and RBV
`or 25 mg. Antiviral activity was observed with a mean
`for 14 days in treatment-naive HCV genotype i patients
`HCV RNAreduction of -0.71 and -1.03 log,, TU/ml,
`[63]. At day 14 viral load reductionsof-2.7 to -4.1 log,,
`respectively [54,55]. These early human data demon-
`TU/ml were achieved with no adverse events attributed
`strated clinical proof of concept for INX-08189 and
`to IDX184 (17) and with no detection of resistant virus
`consequently, the compound is continuing to undergo
`[64]. However, in an attempt to evaluate the efficacy
`clinical evaluation.
`of a DAA combination of IDX184 (17) with the NS3
`Another application of phosphoramidate prodrug
`protease inhibitor IDX320, three serious adverse events
`technology for
`delivering a
`2’-C-methylguanosine
`were reported in a drug—druginteraction study in healthy
`5’-monophosphate is exemplified by IDX184 (17; Figure
`volunteers and consequently, the programmewasplaced
`7) [56]. In this case the phosphoramidate prodrug moi-
`onclinical hold in September 2010, awaiting resolution
`ety utilized a benzyl! aminein place of an amino acid for
`of the cause of the adverse events [65,66]. In February
`the amine substituent and an S-acety]-2-thioethyl moiety
`2011,
`the US Food and Drug Administration (FDA)
`(SATE)as the phosphate ester substituent in place of the
`removed full clinical hold for IDX184 (17) owing to
`more commonaryl group. The SATE group is a known
`evidence that the toxicity waslikely caused by IDX320,
`phosphate ester prodrug construct [33,57,58]. Mecha-
`and the programme wasplaced onpartial clinical hold.
`nism for release of the desired 2’-C-methylguanosine
`Initiation of a Phase [lb 12-weektrial of IDX184 (17)in
`5’-monophosphateis believed to involve both CYP450-
`combination with pegylated IFN and RBVis

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket