throbber
7202 J. Med. Chem. 2010, 53, 7202–7218
`DOI: 10.1021/jm100863x
`
`0
`-r-fluoro-2
`Discovery of a β-D-2
`-Deoxy-2
`Treatment of Hepatitis C Virus
`
`0
`
`0
`
`-β-C-methyluridine Nucleotide Prodrug (PSI-7977) for the
`
`Michael J. Sofia,* Donghui Bao, Wonsuk Chang, Jinfa Du, Dhanapalan Nagarathnam, Suguna Rachakonda,
`P. Ganapati Reddy, Bruce S. Ross, Peiyuan Wang, Hai-Ren Zhang, Shalini Bansal, Christine Espiritu, Meg Keilman,
`Angela M. Lam, Holly M. Micolochick Steuer, Congrong Niu, Michael J. Otto, and Phillip A. Furman
`
`Pharmasset, Inc., 303A College Road East, Princeton, New Jersey 08540
`
`Received July 10, 2010
`
`Hepatitis C virus (HCV) is a global health problem requiring novel approaches for effective treatment
`of this disease. The HCV NS5B polymerase has been demonstrated to be a viable target for the
`0
`0
`0
`-R-fluoro-2
`development of HCV therapies. β-D-2
`-β-C-methyl nucleosides are selective
`-Deoxy-2
`inhibitors of the HCV NS5B polymerase and have demonstrated potent activity in the clinic.
`0
`0
`0
`0
`-R-fluoro-2
`-phosphate derivative of the β-D-2
`-β-C-
`Phosphoramidate prodrugs of the 5
`-deoxy-2
`methyluridine nucleoside were prepared and showed significant potency in the HCV subgenomic
`replicon assay (<1 μM) and produced high levels of triphosphate 6 in primary hepatocytes and in the
`livers of rats, dogs, and monkeys when administered in vivo. The single diastereomer 51 of diastereomeric
`mixture 14 was crystallized, and an X-ray structure was determined establishing the phosphoramidate
`stereochemistry as Sp, thus correlating for the first time the stereochemistry of a phosphoramidate
`prodrug with biological activity. 51 (PSI-7977) was selected as a clinical development candidate.
`
`Introduction
`The hepatitis C virus (HCVa) presents a global health
`problem with approximately 180 million individuals infected
`worldwide with 80% of those progressing to chronic HCV
`infection.1 Of those chronically infected individuals, approxi-
`mately 30% will develop liver cirrhosis and 10% will go on to
`develop hepatocellular carcinoma.2 The current standard of
`care (SOC) for HCV infected patients consists of regular
`injections of pegylated interferon (IFN) and oral ribavirin
`(RBV) administration. However, SOC has proven to be
`effective in producing a sustained virological response in only
`40-60% of patients treated, dependent on viral genotype and
`other predictors of host immune responsiveness. In addition,
`drug discontinuations may be high because of adverse side
`effects associated with the SOC treatment regimen.3,4 Conse-
`quently, the development of alternative treatment options is
`greatly needed. The search for novel therapies for the treat-
`ment of HCV infection has focused on the development of
`direct acting antiviral agents (DAAs).5,6
`
`*To whom correspondence should be addressed. Phone: (609) 613-
`4123. Fax: 609-613-4150. E-mail: michael.sofia@pharmasset.com.
`a Abbreviations: HCV, hepatitis C virus; SOC, standard of care; IFN,
`interferon; RBV, ribavirin; DAA, direct acting antiviral; RdRp, RNA
`dependent RNA polymerase; b.i.d., twice daily; q.d., once daily; YMPK,
`uridine-cytidine monophosphate kinase; NDPK, nucleoside diphos-
`phate kinase; SAR, structure-activity relationship; NMI, N-methyl-
`imidazole; DCM, dichloromethane; EC90, compound concentration that
`returns 90% of inhibition; SGF, simulated gastric fluid; SIF, simulated
`intestinal fluid; NTP, nucleoside triphosphate; PK, pharmacokinetic;
`Cmax, maximum concentration; AUC, area under the curve; tmax, time at
`maximum concentration; PAMPA, parallel artificial membrane perme-
`ability assay; IC50, compound concentration that returns 50% of
`inhibition; NOAEL, no observed adverse effect level; TFA, trifluoro-
`acetic acid, THF, tetrahydrofuran; IPE,
`isopropyl ether; DMSO,
`dimethylsulfoxide.
`
`pubs.acs.org/jmc
`
`Published on Web 09/16/2010
`
`HCV is a plus strand RNA virus of the Flaviviridea family
`with a 9.6 kb genome encoding for 10 proteins: three structural
`proteins and seven nonstructural proteins. The nonstructural
`proteins, which include the NS5B RNA dependent RNA
`polymerase (RdRp), provide several attractive targets for
`the development of anti-HCV therapy.7,8 The HCV RdRp is
`part of a membrane associated replication complex that is
`composed of other viral proteins, viral RNA, and altered
`cellular membranes.3 The NS5B polymerase is responsible for
`replicating the viral RNA genome and thus is absolutely
`required for HCV replication. As in the case of other viral
`polymerases, two approaches have been pursued to identify
`small molecule HCV NS5B polymerase inhibitors. These
`approaches include the identification of nucleoside analogues
`that function as alternative substrate inhibitors that induce a
`chain termination event and non-nucleoside inhibitors that
`bind to allosteric sites on the polymerase leading to a non-
`functional enzyme.5,6
`Several nucleoside classes have been or continue to be in
`development as inhibitors of HCV. These classes include the
`0
`0
`0
`0
`-R-F-2
`β-D-2
`-β-C-methylribose, the β-D-2
`-β-
`-deoxy-2
`0
`-azidoribose classes.9-13 These classes
`methylribose, and the 4
`are represented by the clinical candidates 1 (RG7128), 2 (NM-
`283), and 3 (R1626), respectively (Figure 1).
`0
`0
`Compound 1 is a 3
`,5
`-diisobutyrate ester prodrug of the
`cytidine nucleoside 4. In clinical studies when administered at
`1000 mg b.i.d., 1 demonstrated efficacy in genotype 1 infected
`patients (reduction of HCV RNA levels) in a 14-day mono-
`therapy study (-2.7 log10 decrease in HCV RNA) and
`produced a 88% RVR in a 4-week combination study with
`SOC-pegylated interferon plus ribavirin.14,15 In addition, 1
`was shown to be efficacious in HCV genotype 2,3 patients who
`had not responded to prior therapy, the first direct-acting
`
`r 2010 American Chemical Society
`
`Gilead 2004
`I-MAK v. Gilead
`IPR2018-00125
`
`

`

`Article
`
`Journal of Medicinal Chemistry, 2010, Vol. 53, No. 19
`
`7203
`
`antiviral to show multiple genotype coverage in the clinic.16
`However, even with the positive clinical attributes of 1, we
`were interested in investigating second generation agents with
`improved potency, enhanced pharmacokinetic properties
`(i.e., q.d. dosing), and the potential for generating high concen-
`trations of the active triphosphate in the liver to enable low
`doses and potentially fixed-dose combinations of DAAs. To
`0
`0
`0
`-R-F-2
`achieve this objective, we focused on β-D-2
`-deoxy-2
`-
`
`Figure 1. Structures of HCV nucleoside inhibitors 1, 2, 3, and 4.
`
`β-C-methyluridine (5) (Figure 2). Earlier studies had shown
`that while 5 was inactive in the HCV replicon assay, its
`triphosphate was a potent inhibitor of HCV NS5B with a
`Ki of 0.42 μM.17-19 In addition, metabolism studies with 4
`showed that the monophosphate of 4 can be deaminated to
`the uridine monophosphate derivative and subsequently ana-
`bolized to the triphosphate 6 by uridine-cytidine mono-
`phosphate kinase (YMPK) and nucleoside diphosphate kinase
`(NDPK) (Figure 2). This uridine triphosphate was shown to
`have an intracellular half-life of 38 h.17-19 Therefore, in order
`to leverage the desired attributes of the uridine derivative, we
`needed to deliver the monophosphate of uridine nucleoside 5.
`To accomplish this, we required a monophosphate prodrug
`that would bypass the nonproductive phosphorylation step
`and that would potentially accomplish our other objective: the
`delivery of high liver concentrations of the desired triphos-
`phate 6.
`Phosphoramidate prodrug strategies had been shown
`to enhance nucleoside potency in cell culture presumably
`by increasing intracellular concentrations of the active
`nucleotide.20-22 However, at the time we began our work
`there was no example where phosphoramidate prodrug tech-
`nology was applied to the inhibition of HCV. We speculated
`that application of the phosphoramidate prodrug method
`would be an ideal approach for delivering the desired uridine
`monophosphate to hepatocytes in an in vivo setting (Figure 3).
`We hoped to take advantage of first pass metabolism where
`the liver enzymes would hydrolyze the terminal carboxylic acid
`
`Figure 2. Metabolism of 4 leads to both the active triphosphate and the inactive nucleoside 5. The monophosphate metabolite of 4 is also
`metabolized to the uridine monophosphate derivative which is then further phosphorylated to the active uridine triphosphate 6.
`
`Figure 3. First pass metabolism of the phosphoramidate prodrug derivative of the monophosphate of 5 releases the monophosphate in the
`liver at the desired site of action.
`
`

`

`7204 Journal of Medicinal Chemistry, 2010, Vol. 53, No. 19
`
`Sofia et al.
`
`Scheme 1a
`
`a (a) 70% aqueous acetic acid, 100 °C; (b) 25% methanolic ammonia, 0-15 °C.
`
`Scheme 2a
`
`a NMI, DCM, -5 to 5 °C; (b) NMI, DCM, 5-25 °C.
`
`ester of the phosphoramidate moiety triggering a cascade of
`chemical and enzymatic events that would produce the
`desired uridine monophosphate at the desired site of action,
`the liver. Subsequently, several reports demonstrated that
`phosphoramidates of several other anti-HCV nucleosides
`were able to improve potency, but these reports did not
`translate this improved in vitro potency into a clinical
`development candidate.23-25 Here we describe the discov-
`0
`0
`0
`-R-F-2
`-β-
`-deoxy-2
`ery of phosphoramidate prodrugs of 2
`0
`C-methyluridine 5
`-monophosphate and the selection of 14
`and ultimately of its single isomer 51 as clinical develop-
`ment candidates.
`
`Results and Discussion
`
`The development of phosphoramidate prodrugs of 5 began
`with the investigation of the anti-HCV SAR around the
`phosphoramidate portion of the molecule. The synthesis of
`0
`0
`0
`-R-F-2
`-β-C-methyluridine phosphoramidates
`-deoxy-2
`the 2
`began with the preparation of the uridine nucleoside 5. To
`obtain the uridine nucleoside 5, we started with the benzoyl
`0
`0
`0
`-R-F-2
`-β-C-methylcytidine (7). We
`-deoxy-2
`protected 2
`recently reported on the efficient synthesis of 7.26 From 7,
`preparation of the uridine nucleoside was efficiently accom-
`plished by a two-step process (Scheme 1). The benzoyl
`cytidine 7 was heated with 80% acetic acid overnight to afford
`the protected uridine 8, which was then treated at room
`temperature with methanolic ammonia to provide 5 in 78%
`yield.
`The phosphoramidate derivatives of the uridine nucleoside
`were prepared as shown in Scheme 2. The phosphoramidate
`
`moiety was appended by reacting 5 with a freshly prepared
`chlorophosphoramidate reagent 11 in the presence of NMI.20
`Each chlorophosphoramidate reagent was prepared by stir-
`ring an amino acid ester 9 with the appropriate phosphoro-
`dichloridate reagent 10 in the presence of an amine base (Et3N
`or NMI) in either THF or dichloromethane. The reaction
`0
`-phosphoramidates 12-49 as the major
`provided the desired 5
`0
`0
`0
`- and 3
`,5
`-phosphor-
`product with lesser amounts of the 3
`0
`-phosphoramidates were
`amidate derivatives. The desired 5
`purified by chromatography as a 1:1 mixture of diastereomers
`at the phosphorus center.
`The anti-HCV activity of these prodrugs was assessed using
`the clone A replicon and a quantitative real time PCR assay.12
`Each compound was simultaneously evaluated for cytotox-
`icity by assessing for the levels of cellular rRNA.12 The
`objective was to identify phosphoramidate prodrugs that
`exhibited submicromolar activity with the hope that this
`increased activity would translate into reduced drug load in
`the clinic relative to 1. A survey of the terminal carboxylic acid
`ester of the phosphoramidate moiety in which the amino acid
`was alanine and the phosphate ester was simply phenyl
`showed that small simple alkyl and branched alkyl groups
`provided the desired submicromolar activity; however, in the
`case of the n-butyl (15), 2-butyl (16), and n-pentyl (17) esters,
`cytotoxicity was observed (Table 1). Small cycloalkyl (18) and
`benzyl (22, 23) esters were also compatible; however, phenyl
`(21) and halogenated alkyl groups (19, 20) did not provide the
`desired potency enhancement.
`A survey of the phosphoramidate phosphate ester sub-
`stituent (Table 2) demonstrated that the 1-naphthyl ester 29
`
`

`

`Article
`
`Journal of Medicinal Chemistry, 2010, Vol. 53, No. 19
`
`7205
`
`Table 1. HCV Replicon Activity of Phosphoramidate Prodrugs 12-23: Modification of the Phosphoramidate Ester Moiety
`
`compd
`
`R2
`
`EC90 cloneA (μM)a
`3.9
`1
`0.91
`Me
`12
`0.98
`Et
`13
`i-Pr
`0.52
`14
`n-Bu
`0.09
`15
`0.06
`2-Bu
`16
`n-Pen
`>50
`17
`c-Hex
`0.25
`18
`FCH2CH2
`1.72
`19
`F2CHCH2
`6.80
`20
`18.50
`Ph
`21
`0.13
`Bn
`22
`0.24
`4-F-Bn
`23
`a Each value is a result of n = 2 determinations. b Clone A cells.
`
`inhibition of cellular rRNA replication at 50 μM (%)b
`
`0
`0
`36.9
`25.9
`79.6
`93.8
`92
`61
`43.8
`38.3
`0
`74.3
`0
`
`Table 2. HCV Replicon Activity of Phosphoramidate Prodrugs 12 and 24-30: Modification of the Phosphoramidate Phenolic Ester Substituent
`
`compd
`
`R3
`
`EC90 cloneA (μM)a
`0.91
`Ph
`12
`0.69
`4-F-Ph
`24
`0.58
`4-Cl-Ph
`25
`2.11
`4-Br-Ph
`26
`0.45
`3,4-Cl-Ph
`27
`0.69
`2,4-Cl-Ph
`28
`0.09
`1-Napth
`29
`>50
`Et
`30
`a Each value is a result of n = 2 determinations. b Clone A cells.
`
`inhibition of cellular rRNA replication at 50 μM (%)b
`
`0.0
`16.8
`62.8
`30.8
`63.7
`10.9
`95.4
`16.8
`
`provided the greatest potency and that mono- and dihalo-
`genated phenolic esters also gave inhibitors with submicromolar
`potency. The derivative with a simple alkyl phosphate ester
`(30) was not active against HCV. Although the 1-naphthol
`ester substitution produced the most potent HCV inhibitor,
`this substitution also led to substantial cytotoxicity and was
`therefore not considered a viable substituent.
`Study of the amino acid side chain demonstrated that a
`small alkyl group (12, 32) was accommodated, but R-sub-
`stitution larger than ethyl showed substantial reduction in
`potency (Table 3). R-Disubstituted amino acids that included
`an R-cyclopropanylamino acid derivative 39 did not provide
`the target submicromolar potency (Table 4). Additionally, it
`was shown that the natural L-amino acid was required for
`activity, since the D-alanine derivative 40 was inactive
`(Table 4).
`Results of the phosphoramidate moiety single substituent
`modifications showed that L-alanine was the preferred amino
`
`acid moiety, that methyl, ethyl, isopropyl, or cyclohexyl
`carboxylate esters provided the desired potency enhancement,
`and that the phosphate ester accommodated simple phenyl or
`halogenated phenyl substituents. Subsequently, select combi-
`nations of these preferred substitutions were prepared in
`which only the phenyl or para-halogenated phenyl phosphate
`ester analogues were examined. Polyhalogenated phosphate
`esters were excluded from further evaluation in order to
`preempt any potential toxicity issues that may arise from the
`release of polyhalogenated phenols upon conversion of the
`phosphoramidate to the desired nucleoside monophosphate
`(Table 5).27-29 The most dramatic difference observed in the
`SAR for the phosphoramidate substituent combinations was
`associated with the terminal carboxylic acid ester substituent
`where the cyclohexyl ester derivatives (18, 47-49) showed as
`much as a 10-fold improvement in potency relative to their
`methyl, ethyl, or isopropyl analogues. On the basis of replicon
`potency, initial cytotoxicity profile, and structural diversity,
`
`

`

`7206 Journal of Medicinal Chemistry, 2010, Vol. 53, No. 19
`
`Sofia et al.
`
`Table 3. HCV Replicon Activity of Phosphoramidate Prodrugs 12 and 31-37: Modification of the Amino Acid Side Chain
`
`compd
`
`R1
`
`EC90 clone A (μM)a
`22.11
`H
`31
`0.91
`Me
`12
`1.61
`Et
`32
`Me2CH
`>50
`33
`Me2CHCH2
`5.4
`34
`MeSCH2CH2
`60.13
`35
`PhCH2
`57.65
`36
`15.6
`indole-3-CH2
`37
`a Each value is a result of n = 2 determinations. b Clone A cells.
`
`inhibition of cellular rRNA replication at 50 μM (%)b
`
`0.0
`0.0
`0.0
`0.0
`0.0
`24.1
`20.6
`68.4
`
`Table 4. HCV Replicon Activity of Phosphoramidate Prodrugs 24 and 38-40: R-Disubstituted Amino Acid Side Chains
`
`compd
`
`EC90 clone A (μM)a
`0.69
`24
`2.20
`38
`>50
`39
`>50
`40
`a Each value is a result of n = 2 determinations. b Clone A cells.
`
`inhibition of cellular rRNA replication at 50 μM (%)b
`
`16.8
`0.0
`0.0
`0.09
`
`a select set of seven compounds (12, 14, 18, 41, 44, 45, and 47)
`was chosen for further evaluation.
`To achieve the objective of identifying a phosphoramidate
`0
`0
`-R-F-2
`-β-C-methyluridine monophosphate
`prodrug of the 2
`suitable for clinical studies as a treatment for HCV, the
`prodrug moiety would need to survive exposure in the gastro-
`intestinal tract and preferentially release the nucleotide mono-
`phosphate in the liver. Consequently, compounds 12, 14, 18,
`41, 44, 45, and 47 were further evaluated for gastrointestinal
`stability using simulated gastric fluid (SGF) and simulated
`intestinal fluid (SIF). In addition, stability in human plasma
`and stability on exposure to human liver S9 fraction were
`also evaluated (Table 6). The human liver S9 fraction was
`chosen as a surrogate in vitro model to test for liver stability.
`The ultimate objective was to select compounds that showed
`
`improved potency relative to 4 in the HCV replicon and
`stability in SGF, SIF, and plasma but showed a short half-
`life in liver S9, which could indicate rapid release in hepato-
`cytes. Table 6 shows the in vitro stability data for the key
`compounds selected from combinations of the preferred
`phosphoramidate substituents. Each of these compounds
`exhibited prolonged stability (t1/2>15 h) in SGF, SIF, and
`human plasma but decomposed quickly to the monophos-
`phate when incubated with human liver S9 fraction.
`Having the desired target activity and stability profile,
`compounds 12, 14, 18, 41, 44, 45, and 47 were evaluated in
`vivo to determine liver levels of the active uridine triphosphate
`6 after oral administration. Since HCV replicates in liver cells,
`measurable levels of nucleoside triphosphate (NTP) should be
`a strong indication of in vivo efficacy, the assumption being
`
`

`

`Article
`
`Journal of Medicinal Chemistry, 2010, Vol. 53, No. 19
`
`7207
`
`Table 5. HCV Replicon Activity of Phosphoramidate Prodrugs: Simultaneous Carboxylate and Phenolic Ester Modification of the
`Phosphoramidate Moiety
`
`compd
`
`R2
`
`R3
`
`Ph
`Me
`12
`4-F-Ph
`Me
`24
`4-Cl-Ph
`Me
`25
`4-Br-Ph
`Me
`26
`Ph
`Et
`13
`4-F-Ph
`Et
`41
`4-Cl-Ph
`Et
`42
`4-Br-Ph
`Et
`43
`i-Pr
`Ph
`14
`i-Pr
`4-F-Ph
`44
`i-Pr
`4-Cl-Ph
`45
`i-Pr
`4-Br-Ph
`46
`c-Hex
`Ph
`18
`c-Hex
`4-F-Ph
`47
`c-Hex
`4-Cl-Ph
`48
`c-Hex
`4-Br-Ph
`49
`a Each value is a result of n = 2 determinations. b Clone A cells.
`
`EC90 cloneA (μM)a
`1.62
`0.69
`0.58
`2.11
`0.98
`0.76
`0.39
`0.36
`0.52
`0.77
`0.42
`0.57
`0.25
`0.04
`0.054
`0.039
`
`inhibition of cellular rRNA replication at 50 μM (%)b
`
`0.0
`16.8
`62.8
`30.8
`36.9
`55.3
`0.0
`80.5
`25.9
`0.0
`0.0
`0.0
`61.1
`52.1
`66.9
`91.5
`
`Table 6. Stability Assessment in SGF, SIF, Human Plasma, and
`Human Liver S9 Fraction for Compounds 12, 14, 18, 41, 44, 45, and 47
`
`compd
`
`SGFa
`
`SIFb
`
`stability t1/2 (h)
`human plasmac
`
`human S9d
`
`0.18
`16.7
`>20
`15.5
`12
`0.57
`>24
`>24
`22
`14
`1.4
`>24
`>20
`17
`18
`0.23
`>8
`>20
`17
`41
`0.42
`>24
`>20
`>20
`44
`0.35
`>24
`>20
`>20
`45
`0.18
`>24
`>20
`20
`47
`a SGF = simulated gastric fluid, pH 1.2, 50 μg/mL concentration,
`37 °C, 20 h. b SIF = simulated intestinal fluid, pH 7.5, 50 μg/mL concen-
`tration, 37 °C, 20 h. c 100 μM, 37 °C, 24 h. d 100 μM, 37 °C, 24 h, pH 7.4.
`
`the larger the amount of NTP, the greater the potential
`efficacy. Therefore, each of the seven key compounds was
`evaluated in a screening rat PK study where each compound
`was administered as a single 50 mg/kg oral dose, livers were
`removed, and liver extracts were assayed for levels of the
`0
`0
`0
`-R-F-2
`-β-C-methyluridine triphosphate. The rela-
`-deoxy-2
`2
`tive levels of triphosphate found in the liver samples would be
`an indication of anticipated in vivo potency and would be
`used to select which of the key compounds would progress
`further. Table 7 shows the PK parameters for each of the key
`compounds. Among the seven compounds, compounds 12,
`14, and 47 produced the highest Cmax and AUC values. These
`results strongly suggest that each of these compounds was able
`to traverse the GI tract, remain intact during the absorption
`phase, and arrive intact at the target organ, ultimately result-
`ing in high drug exposure in the liver.
`Since little is known about which species is predictive
`of human exposure for nucleoside phosphoramidates, addi-
`tional in vivo PK assessment was undertaken in dog and
`cynomolgus monkeys to provide a cross-species comparison
`
`Table 7. PK Parameters of Uridine Triphosphate 6 in Rat Liver after an
`Oral Dose of 50 mg/kg Phosphoramidate Prodrugs 12, 14, 18, 41, 44, 45,
`and 47a
`
`compd
`
`Cmax
`(ng/g)
`
`tmax
`(h)
`
`AUC(0-t)
`AUC(inf)
`(ng 3 h/g)
`(ng 3 h/g)
`18968
`14206
`6.00
`1985
`12
`18080
`16796
`4.00
`1934
`14
`8831
`6487
`2.00
`557
`18
`5423
`4191
`4.00
`291
`41
`7375
`6140
`6.00
`519
`44
`8468
`5143
`1.00
`339
`45
`9888
`8937
`4.00
`716
`47
`a Livers were removed at time points 0.5, 1, 2, 4, 6, and 12 h postdose.
`
`of liver exposure to determine whether a dramatic species
`difference in PK behavior existed among compounds 12, 14,
`and 47. Comparison of the in vivo PK characteristic in dogs
`and cynomolgus monkeys was accomplished by evaluating
`both plasma and liver exposures upon oral q.d. dosing over
`4 days with a 50 mg/kg daily dose. Plasma samples were taken
`on day 3, and liver samples were taken on day 4. Liver levels
`were determined at a single time point 4 h postdose on day 4
`and therefore reflect a single concentration at a single point in
`time. Plasma and liver levels of phosphoramidate prodrug and
`liver triphosphate levels were analyzed by LC/MS/MS. In the
`dog study, compound 14 showed a 16- and 110-fold higher
`overall plasma exposure (AUC) of the parent prodrug than
`compounds 12 and 47, respectively (Table 8). In the monkey
`compound 14 also provided greater (>3-fold) plasma expo-
`sure than did compounds 12 and 47 (Table 9). Similarly, liver
`exposures in dog and monkey of the parent prodrugs were
`higher for compound 14 than for compounds 12 and 47.
`Triphosphate levels in the liver demonstrated the same rela-
`tive trends. In dog and monkey compound 14 produced
`higher triphosphate levels relative to compounds 12 and 47.
`
`

`

`7208 Journal of Medicinal Chemistry, 2010, Vol. 53, No. 19
`
`Sofia et al.
`
`Table 8. Dog Plasma and Liver PK Profile after Oral Administration of Compounds 12, 14, and 47
`plasma (prodrug)b
`
`liverc
`
`compda
`
`dose (mg/kg)
`
`Cmax (ng/mL)
`
`Tmax (h)
`
`AUC(0-t)
`AUC(inf)
`(ng 3 h/mL)
`(ng 3 h/mL)
`4960
`5.24
`418
`420
`1.00
`317
`50
`12
`10560
`612
`6894
`6903
`0.50
`6179
`50
`14
`476
`8.72
`54
`62
`0.25
`36
`50
`47
`a Dose of 50 mg/kg for 4 consecutive days. b Blood samples collected at 1, 2, 4, 6, 12, and 24 h postdose on day 3. c Livers removed at 4 h time point
`postdose on day 4.
`
`prodrug
`(ng/g liver)
`
`uridine triphosphate 6
`(ng/g liver)
`
`Table 9. Cynomolgus Monkey Plasma and Liver PK Profile after Oral Administration of Compounds 12, 14, and 47
`plasma (prodrug)b
`
`liverc
`
`compda
`
`dose (mg/kg)
`
`Cmax (ng/mL)
`
`Tmax (h)
`
`AUC(0-t)
`AUC(inf)
`(ng 3 h/mL)
`(ng 3 h/mL)
`26
`4.66
`27
`34
`0.25
`19
`50
`12
`57
`177
`86
`170
`1.00
`33
`50
`14
`NA
`13
`NA
`NA
`6.00
`1.8
`50
`47
`a Dose of 50 mg/kg for 4 consecutive days. b Blood samples collected at 1, 2, 4, 6, 12, and 24 h postdose on day 3. c Livers removed at 4 h time point
`postdose on day 4.
`
`prodrug
`(ng/g liver)
`
`uridine triphosphate 6
`(ng/g liver)
`
`Table 10. AlogP and PAMPA Profile of Compounds 12, 14, and 47
`
`compd
`
`AlogP a
`
`PAMPA permeability (nm/s)b
`
`0.19
`12
`0.92
`14
`2.26
`47
`a Reference 31. b pH 7.4, incubated for 12-24 h.
`
`0.07
`0.46
`4.88
`
`generated by cells when incubated with cytidine nucleoside 4.
`These enhanced triphosphate levels in clone A cells are
`consistent with the increased potency observed for com-
`0
`0
`-C-methyl-
`-F,2
`pounds 12, 14, and 47 when compared to 4. 2
`0
`-triphosphate levels in primary hepatocytes of
`uridine 5
`human, rat, dog, and monkey incubated with compounds 12,
`14, and 47 were also shown to be high, whereas in each case
`the triphosphate levels following incubation with 4 were
`shown to be below the limit of detection. Compound 47
`consistently showed the lowest triphosphate levels of the three
`phosphoramidate analogues. Compound 14 demonstrated 3-
`to 4-fold higher triphosphate levels in human primary hepa-
`tocytes relative to compounds 12 and 47. Compounds 12 and
`14 produced comparable amounts of triphosphate in monkey
`and rat hepatocytes, whereas in dog hepatocytes compound
`12 showed 2-fold higher levels than 14. The interspecies
`differences observed regarding conversion of phosphor-
`amidates 12, 14, and 47 to the active triphosphate 6 may be
`attributed to either cross-species differences in the level of
`enzymes needed to convert the phosphoramidates to the
`intermediate monophosphate or to differences in cell penetra-
`tion. Similarly, since each of the phosphoramidates 12, 14, and
`47 is converted to the same triphosphate metabolite, intra-
`species differences could be attributed to the selectivity of
`processing enzymes or to the ability to penetrate the species
`specific hepatocytes by each of the different analogues. To
`assess relative membrane permeability characteristics, both
`calculated AlogP values and parallel artificial membrane
`permeability (PAMPA) were evaluated. Both AlogP and
`PAMPA results indicate that the predicted order of cell
`permeability would be 47 > 14 >12, in line with lipophilic
`character (Table 10).31,32 This relative order tracks well with
`the relative replicon EC90 values; however, the order does not
`necessarily correlate to triphosphate levels observed in hepato-
`cytes. Therefore, it is possible that other factors such as
`
`Figure 4. Uridine triphosphate (6) levels in human, rat, dog, and
`monkey primary hepatocytes when treated with 4 and uridine
`phosphoramidates 12, 14, and 47.
`
`However, it should be noted that the liver triphosphate data
`were only used for an intraspecies comparison of compounds.
`One should not compare the relative dog to monkey liver
`triphosphate levels because of the liver single time point
`sampling protocol.
`Since the generation of triphosphate levels is critical to
`predicting in vivo potency of nucleos(t)ide analogues, in vitro
`triphosphate production in primary human hepatocytes was
`examined and compared to in vitro triphosphate levels deter-
`mined in primary hepatocytes from rat, dog, and monkey
`when incubated with compounds 12, 14, and 47. An analysis
`of uridine triphosphate (6) production in these primary
`hepatocyte studies would provide a comparison of the inher-
`ent capacity of each nucleotide phosphoramidate to both
`enter hepatocytes and be converted to the target triphosphate.
`In vitro analysis of triphosphate production was accom-
`plished by incubating each compound for 48 h with primary
`hepatocytes from rat, dog, monkey, and human and then
`extracting the cells and analyzing the extracts by HPLC.19,30
`The triphosphate levels generated in the primary hepatocytes
`were compared to triphosphate levels generated in clone A
`replicon cells and to the triphosphate levels generated on
`exposure to a known clinically efficacious agent, 4. Figure 4
`0
`0
`-C-methylur-
`-F,2
`shows that for compounds 12, 14, and 47 2
`0
`-triphosphate levels in clone A cells were 6- to 16-fold
`idine 5
`0
`0
`0
`-C-methylcytidine 5
`-F,2
`-triphosphate levels
`greater than 2
`
`

`

`Article
`
`Journal of Medicinal Chemistry, 2010, Vol. 53, No. 19
`
`7209
`
`selectivity for processing enzymes or relative levels of proces-
`sing enzymes in hepatocytes from different species are con-
`tributing to the relative difference in cellular triphosphate
`levels observed.
`Nucleos(t)ide toxicity can be correlated to several cellular
`effects. Mitochondrial toxicity has been reported to be asso-
`ciated with the long-term use of certain nucleos(t)ide ana-
`logues resulting in myopathy, peripheral neuropathy, and
`pancreatitis.33,34 Hematotoxicity, which can lead to neutro-
`penia, severe anemia, and thrombocytopenia, has also been
`associated with several nucleoside analogues including the
`HCV polymerase inhibitor 3.35,36 One cause of hematotoxicity
`is bone marrow toxicity.35,36 To examine for these possible
`toxicities, compounds 12, 14, and 47 were evaluated for
`general cytotoxicity against an expanded cell panel and
`evaluated in vitro for both mitochondrial toxicity and toxicity
`to bone marrow progenitor cells. In an expanded cell panel
`that included two human hepatocyte cell lines, Huh7 and
`HepG2, a human pancreatic cell line, BxBC3, and a human T
`lymphoblast cell line, CEM, compounds 12, 14, and 47 were
`found to show no cytotoxicity up to 100 μM, the highest
`concentration tested. Compounds 12, 14, and 47 were assessed
`for mitochondrial toxicity in both CEM and HepG2 cells by
`incubating each compound for 14 days. Percentage inhibition
`of mitochondrial DNA production was determined relative to
`a no drug control. No inhibition of mitochondrial DNA
`synthesis up to the highest concentration tested (50 μM) was
`observed for the three compounds. To evaluate the potential
`for bone marrow toxicity, compounds 12, 14, and 47 were
`screened from 0.1 to 50 μM for their effect on human
`erythroid and myeloid progenitor cell colony proliferation.
`Differentiation of hematopoietic progenitors into erythroid or
`granulocyte-myeloid cell lineages over 14 days was measured.
`The results showed that compounds 12 and 14 had IC50 values
`of >50 μM for both erythroid and myeloid progenitor cells.
`Erythroid and myeloid progenitor cells were more sensitive to
`compound 47 which showed IC50 values of 37 ( 5 μM and
`30 ( 5 μM for erythroid and myeloid progenitor cells, res-
`pectively. Since each phosphoramidate derivative leads to the
`same metabolic intermediates, it is unlikely that the metabolic
`intermediates leading to the triphosphate active metabolite
`contribute to the effects on erythroid or myeloid progenitor
`cells. Therefore, since the only difference between compounds
`12, 14, and 47 resides in the nature of the carboxylic acid ester
`and the phenolic ester substituents, it would appear that the
`character of the carboxylate and/or phenolic esters contributes
`significantly to the effect on erythroid and myeloid progenitor
`cells and therefore the potential for bone marrow toxicity. The
`effect on progenitor cells observed for compound 47 could be a
`result of the phosphoramidate itself or from the released ester
`moieties after prodrug metabolism.
`A comparison of in vivo acute toxicity of compounds 12, 14,
`and 47 was undertaken by single dose oral administration in rats
`with a 14-day postdose observation period. Rats (three males
`and three females) were dosed with each compound at doses of
`50, 300, and 1800 mg/kg. Fourteen days after dose administra-
`tion, all rats were euthanized and daily clinical observations,
`body weights, macroscopic pathology including kidney and
`liver weights were assessed. For each of the compounds, no test-
`article-related mortality, clinical signs of toxicity, body weight
`changes, macroscopic pathology, or organ weight changes for
`liver and kidney were observed in any of the treatment groups.
`Consequently, for each of the compounds studied the NOAEL
`was established at >1800 mg/kg.
`
`Figure 5. X-ray crystal structure of 51 crystallized from CH2Cl2.
`
`In vitro hepatocyte triphosphate levels and in vivo PK
`profiles and effects on bone marrow progenitor cells were
`factors that differentiated compounds 12, 14, and 47. Com-
`pound 14 consistently produced high levels of triphosphate
`across all species and in particular showed the greatest triphos-
`phate levels in primary human hepatocytes by as much as
`3-fold over compounds 12 and 47. The in vivo PK assessment
`showed that compound 14 consistently demonstrated the
`highest liver triphosphate levels across species. Bone marrow
`toxicity studies showed that no difference in safety profile was
`observed between compounds 12 and 14; however, bone
`marrow progenitor cells were more sensitive to the presence
`of compound 47. On the basis of the overall profile, com-
`pound 14 (PSI-7851) was selected for further development.37
`14 is a 1:1 mixture of diastereomers at the phosphorus
`center of the phosphoramidate moiety and is a low melting
`point (mp = 66-75 °C) amorphous solid. The diastereomers
`of 14 were separated by HPLC chromatography to give the
`two pure diastereomers 50 (fast moving isomer) and 51 (slow
`moving isomer). In the clone A replicon assay, compounds 50
`and 51 produced anti-HCV activity with EC90 values of 7.5
`and 0.42 μM, respectively, thus demonstrating a >10-fold
`difference in activity between the two isomers. Diastereomer
`51 was subsequently crystallized using methylene chloride as
`the solvent, and a single crystal X-ray structure of 51 was
`obtained, definitively establishing the configuration of the
`phosphorus center as Sp and by corollary the configuration of
`50 as Rp (Figures 5 and 6). This is the first demonstrated
`crystallization and X-ray structure determination of a phos-
`phoramidate nucleotide prodrug and the first example where
`stereochemistry at phosphorus could be correlated unequiv-
`ocally to nucleotide phosphoramidate activity.
`Each diastereomer was also evaluated against repli

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