throbber
Downloaded from
`
`http://www.jbc.org/
`
` by guest on January 13, 2015
`
`THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 282, NO. 41, pp. 29812–29820, October 12, 2007
`© 2007 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in the U.S.A.
`
`Characterization of the Metabolic Activation
`of Hepatitis C Virus Nucleoside Inhibitor
`␤-D-2ⴕ-Deoxy-2ⴕ-fluoro-2ⴕ-C-methylcytidine
`(PSI-6130) and Identification of a Novel Active
`5ⴕ-Triphosphate Species*
`
`Received for publication, June 27, 2007, and in revised form, August 6, 2007 Published, JBC Papers in Press, August 13, 2007, DOI 10.1074/jbc.M705274200
`Han Ma1, Wen-Rong Jiang, Nicole Robledo, Vincent Leveque, Samir Ali, Teresa Lara-Jaime,
`Mohammad Masjedizadeh, David B. Smith, Nick Cammack, Klaus Klumpp, and Julian Symons
`From the Roche Palo Alto LLC, Palo Alto, California 94304
`
`␤-D-2ⴕ-Deoxy-2ⴕ-fluoro-2ⴕ-C-methylcytidine (PSI-6130) is a
`potent inhibitor of hepatitis C virus (HCV) replication in the
`subgenomic HCV replicon system, and its corresponding
`5ⴕ-triphosphate is a potent inhibitor of the HCV RNA polymer-
`ase in vitro. In this study the formation of PSI-6130-triphos-
`phate was characterized in primary human hepatocytes. PSI-
`6130 and its 5ⴕ-phosphorylated derivatives were identified, and
`the intracellular concentrations were determined. In addition,
`the deaminated derivative of PSI-6130, ␤-D-2ⴕ-deoxy-2ⴕ-fluoro-
`2ⴕ-C-methyluridine (RO2433, PSI-6026) and its corresponding
`phosphorylated metabolites were identified in human hepato-
`cytes after incubation with PSI-6130. The formation of the
`5ⴕ-triphosphate (TP) of PSI-6130 (PSI-6130-TP) and RO2433
`(RO2433-TP) increased with time and reached steady state lev-
`els at 48 h. The formation of both PSI-6130-TP and RO2433-TP
`demonstrated a linear relationship with the extracellular con-
`centrations of PSI-6130 up to 100 ␮M, suggesting a high capacity
`of human hepatocytes to generate the two triphosphates. The
`mean half-lives of PSI-6130-TP and RO2433-TP were 4.7 and
`38 h, respectively. RO2433-TP also inhibited RNA synthesis by
`the native HCV replicase isolated from HCV replicon cells and
`the recombinant HCV polymerase NS5B with potencies compa-
`rable with those of PSI-6130-TP. Incorporation of RO2433-5ⴕ-
`monophosphate (MP) into nascent RNA by NS5B led to chain
`termination similar to that of PSI-6130-MP. These results dem-
`onstrate that PSI-6130 is metabolized to two pharmacologically
`active species in primary human hepatocytes.
`
`Hepatitis C is a major health problem affecting ⬃170 million
`people worldwide of which around 3 million chronically
`infected patients reside within the United States (1). The cur-
`rent standard treatment for hepatitis C consisting of pegylated
`interferon-␣and ribavirin only results in about a 50% sustained
`virological response in patients infected with genotype 1 hepa-
`
`* The costs of publication of this article were defrayed in part by the payment
`of page charges. This article must therefore be hereby marked “advertise-
`ment” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.
`1 To whom correspondence should be addressed: Roche Palo Alto LLC, 3431
`Hillview Ave., Palo Alto, CA 94304. Tel.: 650-852-3190; Fax: 650-354-7554;
`E-mail: han.ma@roche.com.
`
`titis C virus (HCV),2 the most predominant genotype in the
`United States and Europe (2–4). New treatment options with
`improved clinical efficacy and greater tolerability are urgently
`needed. Novel antiviral agents targeting essential processes of
`HCV replication as part of optimized combination regimens
`could achieve increased clinical efficacy and potentially
`improved adverse event profiles as well as shortened treatment
`duration as compared with the current standard of care.
`HCV RNA replication is mediated by a membrane-associ-
`ated multiprotein replication complex (5, 6). The HCV NS5B
`protein, the RNA-dependent RNA polymerase, is the catalytic
`subunit of the HCV replication complex and is responsible for
`the synthesis of the RNA progeny and, hence, is a prime target
`of anti-viral inhibition. Nucleoside analogs have been estab-
`lished as successful antiviral agents targeting the active site of
`DNA polymerases for the treatment of other viral diseases,
`including human immunodeficiency virus, hepatitis B virus,
`and herpes simplex virus (7). The majority of marketed antiviral
`nucleoside analogs need to be converted to the active
`5⬘-triphosphate forms in the target cells. These nucleotide
`triphosphate analogs then serve as alternative substrates for the
`viral DNA polymerases and compete with the incorporation of
`the corresponding natural nucleotide triphosphates. Upon
`incorporation by the viral DNA polymerases, the lack of the
`3⬘-hydroxyl group in the deoxyribose moiety leads to the ter-
`mination of the nascent viral DNA (chain termination).
`In the past few years a number of ribonucleoside analogs with
`2⬘-C-methyl, 2⬘-O-methyl, or 4⬘-azido substituents on the
`ribose moiety have been reported to be inhibitors of HCV rep-
`lication in the subgenomic replicon system (8–13). Prodrugs of
`two nucleoside analogs, 2⬘-C-methylcytidine (NM107) and
`4⬘-azidocytidine (R1479), have successfully progressed into
`clinical development and shown efficacy in HCV-infected
`patients (14, 15). The corresponding nucleotide triphosphate
`analogs are substrates for HCV polymerase NS5B and inhibit
`RNA synthesis activity of HCV NS5B in vitro. The incorpora-
`tion of the nucleotide analogs into nascent HCV RNA strongly
`reduces the efficiency of further RNA elongation by NS5B,
`
`2 The abbreviations used are: HCV, hepatitis C virus; MP, 5⬘-monophosphate;
`DP, 5⬘-diphosphate; TP, 5⬘-triphosphate; HPLC, high performance liquid
`chromatography.
`
`29812 JOURNAL OF BIOLOGICAL CHEMISTRY
`
`VOLUME 282 • NUMBER 41 • OCTOBER 12, 2007
`
`IPR2018-00119
`
`Page 1 of 10
`
`I-MAK 1005
`
`

`

`Downloaded from
`
`http://www.jbc.org/
`
` by guest on January 13, 2015
`
`Metabolism and Mechanism of Action of ␤-D-2ⴕ-Deoxy-2ⴕ-fluoro-2ⴕ-C-methylcytidine
`
`resulting in termination of the nascent RNA product. There-
`fore, these nucleoside analogs are non-obligatory chain termi-
`nators despite the presence of a 3⬘-hydroxyl group.
`Recently, ␤-D-2⬘-deoxy-2⬘-fluoro-2⬘-C-methylcytidine (PSI-
`6130) has been identified as a potent and selective inhibitor of
`HCV replication in the subgenomic replicon system with little
`or no cytotoxicity in various human cell lines or bone marrow
`precursor cells (16). The corresponding triphosphate of PSI-
`6130 is an inhibitor of HCV NS5B competitive with natural
`CTP (17). Conversion to the active 5⬘-triphosphate form by
`cellular kinases is an important part of the mechanism of action
`for nucleoside analogs. In this study we determined the metab-
`olism of ␤-D-2⬘-deoxy-2⬘-fluoro-2⬘-C-methylcytidine in pri-
`mary human hepatocytes isolated from several donors. We
`show that ␤-D-2⬘-deoxy-2⬘-fluoro-2⬘-C-methylcytidine was
`converted to ␤-D-2⬘-deoxy-2⬘-fluoro-2⬘-C-methylcytidine
`5⬘-triphosphate and ␤-D-2⬘-deoxy-2⬘-fluoro-2⬘-C-methyluri-
`dine 5⬘-triphosphate via deamination of the phosphorylated
`cytidylates. Furthermore, we determined the kinetics of the for-
`mation of the two active triphosphates and the potency of the
`two triphosphates against the native HCV replicase and NS5B
`as well as the molecular mechanism of action of the two
`triphosphates.
`
`EXPERIMENTAL PROCEDURES
`Compounds—␤-D-2⬘-Deoxy-2⬘-fluoro-2⬘-C-methylcytidine
`(PSI-6130) was provided by Pharmasset, Inc. (18). A stock solu-
`tion of 10 mM PSI-6130 was prepared in Dulbecco’s phosphate-
`buffered saline and stored at ⫺20 °C. Tritium-labeled PSI-6130
`was synthesized at Roche Palo Alto LLC. The tritiated com-
`pound was dissolved in 50% (v/v) ethanol at the concentration
`of 0.97 mCi/ml with a specific activity of 25.78 Ci/mmol. The
`stock solution was stored at ⫺20 °C. The phosphorylated deriv-
`atives of PSI-6130, namely PSI-6130-MP, -DP, and -TP, were
`provided by Pharmasset, Inc. ␤-D-2⬘-Deoxy-2⬘-fluoro-2⬘-C-
`methyluridine (RO2433) was synthesized at Roche Palo Alto
`LLC. RO2433-MP, -DP, and -TP were synthesized by TriLink
`BioTechnologies (San Diego, CA). Compound stock solutions
`were prepared in nuclease-free H2O and stored at ⫺20 °C.
`3⬘-dCTP was purchased from TriLink BioTechnologies.
`Cell Culture of Primary Human Hepatocytes—Plated fresh
`human hepatocytes or hepatocyte suspensions were obtained
`either from CellzDirect, Inc. or from In Vitro Technologies,
`Inc. Fresh human hepatocytes obtained from each company
`were plated or cultured on 6-well collagen coated plates (BD
`Biosciences #356400) at 1.5 million cells per well using com-
`plete serum containing medium obtained from the respective
`companies. Cells were allowed to recover for at least 18 h before
`the addition of the compound. All incubations were carried out
`at 37 °C in a humidified 5% CO2 atmosphere.
`To determine the time course of uptake and phosphorylation
`of PSI-6130, human primary hepatocytes were incubated with
`3H-labeled PSI-6130 at a final concentration of 2 ␮M and 10
`␮Ci/ml. The compound was added 72, 48, 24, 16, 6 and 1 h
`before cell harvesting. All time points and untreated cell con-
`trols were set up in duplicates.
`To determine the dose response of the phosphorylation of
`PSI-6130, human primary hepatocytes were incubated with
`
`3H-labeled PSI-6130 at 0, 2, 10, 25, 50, 100, and 250 ␮M for 24 h.
`Final concentrations of PSI-6130 were achieved by supple-
`menting 3H-labeled PSI-6130 with non-radiolabeled PSI-6130.
`Duplicate cell samples were harvested after 24 h of incubation.
`To determine the half-life of the triphosphates of PSI-6130
`and RO2433, human primary hepatocytes were incubated for
`24 h with 3H-labeled PSI-6130 at 2 ␮M and 10 ␮Ci/ml. The cell
`monolayer was washed once with the cell culture medium with-
`out PSI-6130 and then incubated with fresh medium without
`PSI-6130 at 0-, 0.5-, 1-, 2-, 4-, 6-, 8-, 24-, 48-, and 72-h time
`points after the removal of PSI-6130. Duplicate cell samples
`were set up for each time point. The viable cell numbers of the
`untreated cell controls for each experiment were determined at
`the end of the experiment using the trypan blue exclusion
`method.
`Preparation of Cell Extract for High Performance Liquid
`Chromatography (HPLC) Analysis—At the time of cell harvest
`the cell culture medium was aspirated, and the cells were
`washed once with cold phosphate-buffered saline. The cells
`were scraped into 1 ml of pre-chilled 60% (v/v) methanol and
`extracted in methanol for 24 h at ⫺20 °C. The extracted sam-
`ples were then centrifuged at 10,000 ⫻ g for 15 min to remove
`cell debris. The supernatant was transferred to new tubes and
`evaporated in a speed vacuum at room temperature. The pellets
`were stored at ⫺80 °C until analysis.
`The dried pellets of cell extracts were dissolved in H2O and
`filtered though a nanosep MF centrifugal device (Pall Life Sci-
`ences #ODM02C34). Before HPLC analysis, cell extract sam-
`ples were spiked with unlabeled reference standards PSI-6130,
`RO2433, and their phosphorylated derivatives.
`HPLC—The phosphorylated derivatives of PSI-6130 were
`separated by ion exchange HPLC with a Whatman Partisil 10
`SAX (4.6 ⫻ 250 mm) column coupled to a radiometric detector
`(␤-RAM, IN/US Systems, Inc.). The mobile phase gradient
`changed linearly from 0% buffer A (H2O) to 100% buffer B (0.5
`M KH2PO4 ⫹ 0.8 M KCl) between 4 and 8 min. 100% buffer B ran
`from 8 to 18 min and changed back to 100% A in 1 min. Buffer
`A ran until 25 min. The flow rate was 1 ml/min. A ratio of 5:1 Flo
`Scint IV or Ultima-FloTM AP (PerkinElmer Life Sciences) to
`column eluent was used for the detection of radiolabeled spe-
`cies in the ␤-RAM detector (IN/US Systems, Inc.).
`The separation of PSI-6130 and RO2433 was performed by
`reverse phase chromatography with a Zorbax SB-C8 column
`(4.6 ⫻ 250 mm, 5 ␮m) coupled to a radiometric detector
`(␤-RAM). The gradient changed linearly from 100% buffer A
`(0.01 M heptane sulfonic acid, sodium salt, 0.1% (v/v) acetic acid
`in water) to 10% buffer B (0.01 M heptane sulfonic acid sodium
`salt, 0.1% (v/v) acetic acid in 1:1 methanol water) between 0 and
`3 min and then changed linearly from 10% buffer B to 95%
`buffer B between 3 and 18 min. 95% buffer B ran from 18 to 22
`min and changed back to 100% A in 0.1 min. Buffer A ran until
`25 min. The flow rate was 1 ml/min. PSI-6130 and its intercel-
`lular metabolites were identified by comparison of the reten-
`tion times of the intracellular species in the radiochromato-
`gram with the retention times of nonradioactive reference
`standards spiked in the cell extract samples and detected by UV
`absorption at 270 nm.
`
`OCTOBER 12, 2007 • VOLUME 282 • NUMBER 41
`
`JOURNAL OF BIOLOGICAL CHEMISTRY 29813
`
`IPR2018-00119
`
`Page 2 of 10
`
`I-MAK 1005
`
`

`

`Downloaded from
`
`http://www.jbc.org/
`
` by guest on January 13, 2015
`
`Metabolism and Mechanism of Action of ␤-D-2ⴕ-Deoxy-2ⴕ-fluoro-2ⴕ-C-methylcytidine
`
`Acid Phosphatase Treatment of Cell Extracts—Hepatocyte
`cell extracts were incubated with acid phosphatase (Sigma
`#P-0157) at a final concentration of 0.05 mg/ml (23.9 units/ml)
`at 37 °C for 2.5 h to dephosphorylate any phosphorylated
`metabolites of PSI-6130. After digestion the samples were ana-
`lyzed by reversed phase HPLC.
`HCV Replicon Assay—The 2209-23 cell line containing a
`bicistronic HCV subgenomic replicon (genotype 1b, Con1
`strain), which expresses a Renilla luciferase reporter gene as an
`index of HCV RNA replication, has been described before (9).
`The analysis of inhibition of HCV replication by nucleoside
`analogs and IC50 determinations were performed as described
`(12).
`HCV Replicase Assay—The membrane-associated native
`HCV replication complexes were isolated from 2209-23 repli-
`con cell lines as described (6). The inhibition of the RNA syn-
`thesis activity of the HCV replicases by PSI-6130-TP was deter-
`mined as described (6) except that 5 ␮l of cytoplasmic replicase
`complex (2.5 ⫻ 106 replicon cell equivalent) was added to a
`20-␮l reaction for 60 min. The inhibition of the RNA synthesis
`activity of the HCV replicases by RO2433-TP was determined
`in reactions containing 6.25 ␮l of cytoplasmic replicase com-
`plex (3.1 ⫻ 106 replicon cell equivalent), 50 mM HEPES, pH 7.5,
`10 mM KCl, 10 mM dithiothreitol, 5 mM MgCl2, 20 ␮g/ml acti-
`nomycin D, 1 mM ATP, GTP, and CTP, 24 ␮Ci of (0.4 ␮M)
`[␣-33P]UTP (PerkinElmer #NEG607H), 1 units/␮l SUPERase.
`In (Ambion), 10 mM creatine phosphate, 200 ␮g/ml creatine
`phosphokinase with or without the nucleotide triphosphate
`inhibitor in a final volume of 20 ␮l for 90 min.
`HCV Polymerase Assay—The inhibition potency of PSI-
`6130-TP on the RNA-dependent RNA polymerase activity of
`recombinant NS5B570-Con1 (genotype 1b, GenBankTM acces-
`sion number AJ242654) was measured as the incorporation of
`radiolabeled nucleotide monophosphate into acid-insoluble
`RNA products as described (6) with the following modifica-
`tions; IC50 determinations were carried out using 200 nM in
`vitro transcribed complementary internal ribosome entry site
`RNA template, 1 ␮Ci of tritiated UTP (42 Ci/mmol), 500 ␮M
`ATP, 500 ␮M GTP, 1 ␮M CTP, 1⫻ TMDN buffer (40 mM Tris-
`HCl, pH 8.0, 4 mM MgCl2, 4 mM dithiothreitol, 40 mM NaCl)
`and 200 nM NS5B570-Con1. The inhibition potency of
`RO2433-TP was determined as described above with the fol-
`lowing modification of NTP concentrations: 1 ␮Ci of tritiated
`CTP (39 Ci/mmol), 500 ␮M ATP, 500 ␮M GTP, 1 ␮M UTP. The
`compound concentration at which the enzyme-catalyzed rate is
`reduced by 50% (IC50) was calculated using equation,
`共% Max ⫺ % Min兲
`X
`
`Y ⫽ % Min ⫹
`
`冉1 ⫹
`
`共IC50兲冊
`
`(Eq. 1)
`
`where Y corresponds to the relative enzyme activity, % Min is
`the residual relative activity at saturating compound concentra-
`tion, % Max is the relative maximum enzymatic activity, and X
`corresponds to the compound concentration.
`The apparent Michaelis constants (Km(app)) for UTP or CTP
`were measured using assay conditions above with the following
`modifications; Km(app) for CTP was measured using 2 ␮Ci of
`
`tritiated UTP (0.93 ␮M), 4.07 ␮M unlabeled UTP, 50 ␮M ATP, 50
`␮M GTP, and 5 nM to 50 ␮M CTP; Km(app) for UTP was meas-
`ured using 2 ␮Ci of tritiated CTP (1.67 ␮M), 3.33 ␮M unlabeled
`CTP, 50 ␮M ATP, 50 ␮M GTP, and 5 nM to 50 ␮M UTP. Appar-
`ent Km(app) values were calculated by nonlinear fitting using
`Equation 2,
`
`Y ⫽
`
`共Vmax共app兲兲 X
`Km共app兲 ⫹ X
`where Y corresponds to the rate of RNA synthesis by NS5B,
`Vmax(app) is the maximum rate or RNA synthesis at saturating
`substrate concentration, and X corresponds to CTP or UTP
`concentration.
`Ki values were derived from the Cheng-Prusoff Equation
`(Equation 3) for competitive inhibition,
`
`(Eq. 2)
`
`(Eq. 3)
`
`Ki ⫽
`
`冉1 ⫹
`
`冊
`
`IC50
`关NTP兴
`Km共app兲
`where [NTP] is the concentration of CTP or UTP, and Km(app) is
`the apparent Michaelis constant for CTP or UTP. Mean Ki
`values were averaged from independent measurements at
`0.2, 1, 5, and 25 ␮M CTP or UTP concentrations in triplicate
`experiments.
`Gel-based Nucleotide Incorporation Assay—The RNA tem-
`plate-directed nucleotide incorporation and extension of
`nucleotide triphosphates and nucleotide triphosphate analogs
`by HCV polymerase was performed with a 19-nucleotide RNA
`oligo (5⬘-AUGUAUAAUUAUUGUAGCC-3⬘) under assay
`conditions as described (9). The incorporation and extension of
`CTP and CTP analogs were determined with 5⬘-end-radiola-
`beled GG primer and nucleotide triphosphates at the indicated
`concentrations. The incorporation and extension of UTP and
`UTP analogs were performed similarly with the same RNA
`oligo template, 5⬘-end-radiolabeled GGC primer and nucleo-
`tide triphosphates at the indicated concentrations. The radio-
`labeled RNA products were separated on a TBE-urea acrylam-
`ide gel and analyzed using phosphorimaging (GE Healthcare).
`
`RESULTS
`Metabolic Profile of PSI-6130—Cellular extracts from
`primary human hepatocytes incubated with tritium-labeled
`␤-D-2⬘-deoxy-2⬘-fluoro-2⬘-C-methylcytidine (PSI-6130) were
`resolved by ion exchange HPLC. PSI-6130 and metabolites
`derived from PSI-6130 were identified by comparing the reten-
`tion times of radiolabeled species with the retention times of
`unlabeled reference compounds (Fig. 1A). As shown in Fig. 1B,
`PSI-6130 (3.0 min) and the 5⬘-phosphorylated derivatives PSI-
`6130-DP (13.2 min) and PSI-6130-TP (16.8 min) were identi-
`fied in human hepatocytes incubated with PSI-6130. In addi-
`tion, metabolites with retention times corresponding to
`those of the deaminated product of PSI-6130, ␤-D-2⬘-deoxy-
`2⬘-fluoro-2⬘-C-methyluridine (RO2433, 3.8 min) and its cor-
`responding 5⬘-phosphates RO2433-DP (12.5 min)
`and
`RO2433-TP (14.8 min), were detected (Fig. 1B). The mono-
`phosphates of the cytidine and uridine analogs PSI-6130-MP
`and RO2433-MP were not separated sufficiently under the
`
`29814 JOURNAL OF BIOLOGICAL CHEMISTRY
`
`VOLUME 282 • NUMBER 41 • OCTOBER 12, 2007
`
`IPR2018-00119
`
`Page 3 of 10
`
`I-MAK 1005
`
`

`

`Metabolism and Mechanism of Action of ␤-D-2ⴕ-Deoxy-2ⴕ-fluoro-2ⴕ-C-methylcytidine
`
`Downloaded from
`
`http://www.jbc.org/
`
` by guest on January 13, 2015
`
`FIGURE 2. Identification of PSI-6130 and its metabolites by reversed
`phase HPLC and acid phosphatase treatment. A, reversed phase HPLC sep-
`aration of standard compounds: PSI-6130 (a, 20 min), RO2433 (b, 11.7 min),
`PSI-6130-MP (c, 7.5 min), (d, PSI-6130-DP, PSI-6130-TP, RO2433-MP, RO2433-
`DP, and RO2433-TP). B, reversed phase HPLC profile of an extract from hepa-
`tocytes incubated with 25 ␮M extracellular PSI-6130 for 24 h. C, reversed
`phase HPLC profile of the same extract after acid phosphatase treatment.
`
`In Vitro Potency of RO2433 and RO2433-TP—As described
`above,
`incubation of human hepatocytes with PSI-6130
`resulted in the formation of substantial concentrations of the
`triphosphate of its uridine analog, RO2433-TP. Next we deter-
`mined whether the PSI-6130-derived uridine analog RO2433
`could inhibit HCV replication targeting NS5B polymerase.
`Huh7 cells containing a subgenomic genotype 1b Con1 strain
`HCV replicon were incubated with RO2433 or PSI-6130 for
`72 h, and dose-dependent inhibition of luciferase reporter
`activity was determined. RO2433 did not inhibit the HCV rep-
`lication in the HCV subgenomic replicon system at concentra-
`tions up to 100 ␮M, whereas PSI-6130 inhibited HCV replica-
`tion with a mean IC50 of 0.6 ␮M under the same assay
`conditions (Table 1). The lack of potency in the replicon could
`be related to inefficient compound phosphorylation. To
`address whether the triphosphate of RO2433 directly inhibits
`the HCV RNA polymerase, the RNA synthesis activity of the
`native membrane-associated HCV replication complexes iso-
`lated from the same replicon cells was tested in the presence of
`RO2433-TP. RO2433-TP inhibited the RNA synthesis activity
`of HCV replicase with a mean IC50 of 1.19 ␮M, whereas PSI-
`6130-TP inhibited HCV replicase with a mean IC50 of 0.34 ␮M
`(Table 1). RO2433-TP also inhibited the RNA synthesis activity
`of the recombinant HCV Con1 NS5B on a heteropolymeric
`RNA template derived from the 3⬘-end of the negative strand of
`the HCV genome with an IC50 of 0.52 ␮M and Ki of 0.141 ␮M, as
`compared with an IC50 of 0.13 ␮M and Ki of 0.023 ␮M for PSI-
`6130-TP under the same assay conditions (Table 1). These
`results established that both RO2433-TP and PSI-6130-TP
`are intrinsically potent inhibitors of RNA synthesis by HCV
`polymerase.
`
`FIGURE 1. Ion exchange HPLC profile of an extract of primary human
`hepatocytes incubated with PSI-6130. A, HPLC separation and retention
`times of reference compounds PSI-6130 (a, 2.5 min), RO2433 (b, 3.4 min),
`RO2433-MP (c, 10.2 min), PSI-6130-MP (d, 10.4 min), RO2433-DP (e, 12.2 min),
`PSI-6130-DP (f, 12.8 min), RO2433-TP (g, 14.4 min), and PSI-6130-TP (h, 16.3
`min). B, HPLC profile of an extract from hepatocytes incubated with 3H-la-
`beled PSI-6130 at 2 ␮M for 24 h. The retention times in min of the intracellular
`species are indicated above the radioactive peaks. Intracellular PSI-6130 and
`its metabolites were identified by comparing the retention times of the radio-
`active peaks with those of the UV absorption peaks of reference compounds.
`There is a calibrated 0.3– 0.4-min delay in the retentions times of radioactive
`trace compared with those of UV trace due to sample traveling from UV
`detector to radiometric detector.
`
`chromatography conditions and, therefore, co-eluted as a sin-
`gle radioactive peak at 10.6 min. It has been reported that 2⬘-O-
`methylcytidine was extensively metabolized to CTP and UTP
`due to deamination coupled with demethylation of the 2⬘-sub-
`stituent or base swapping after glycosidic bond cleavage (19).
`None of the intracellular metabolites of PSI-6130 was eluted
`with retention time corresponding to those of 2⬘-C-methyl-
`CTP, CTP, and UTP (data not shown). Therefore, there was no
`evidence for metabolism of PSI-6130 at the 2⬘-position or evi-
`dence for hydrolysis at the glycosidic bond. These data suggest
`that the primary routes of PSI-6130 metabolism in human
`hepatocytes were phosphorylation at the 5⬘-position and
`deamination at the base.
`The hepatocyte extracts were also analyzed by reversed
`phase HPLC to identify unphosphorylated metabolites of PSI-
`6130. Two unphosphorylated species with retention times cor-
`responding to PSI-6130 (20.3 min) and its uridine metabolite
`RO2433 (11.9 min) were observed, with PSI-6130 being the
`predominant intracellular species (Fig. 2B). There was no evi-
`dence of formation of uracil, uridine, cytosine, cytidine, or
`2⬘-deoxycytidine (data not shown). These data agree well with
`the ion exchange HPLC analysis result and suggest the absence
`of metabolism at the 2⬘ position and at the glycosidic bond. The
`phosphorylated metabolites, with the exception of PSI-6130-
`MP, were not resolved by reversed phase HPLC and co-eluted
`early as a single broad peak (Fig. 2, A, and B). Acid phosphatase
`treatment completely converted all the intracellular metabo-
`lites to PSI-6130 and RO2433. Therefore, all detected intracel-
`lular metabolites represent phosphorylated derivatives of PSI-
`6130 and RO2433. These results established that PSI-6130
`could be phosphorylated to its pharmacologically active
`5⬘-triphosphate analog and that PSI-6130 and/or its phos-
`phates could be deaminated to the corresponding uridine ana-
`logs in primary human hepatocytes.
`
`OCTOBER 12, 2007 • VOLUME 282 • NUMBER 41
`
`JOURNAL OF BIOLOGICAL CHEMISTRY 29815
`
`IPR2018-00119
`
`Page 4 of 10
`
`I-MAK 1005
`
`

`

`Metabolism and Mechanism of Action of ␤-D-2ⴕ-Deoxy-2ⴕ-fluoro-2ⴕ-C-methylcytidine
`
`TABLE 1
`Potency of PSI-6130 and RO2433 and their 5ⴕ-triphosphates
`Values presented are the mean ⫾ S.D. from greater than three independent experiments. ND, not determined.
`IC50
`Compound
`NS5B GT1b
`␮M
`ND
`ND
`0.13 ⫾ 0.01
`0.52 ⫾ 0.11
`
`PSI-6130
`RO2433
`PSI-6130-TP
`RO2433-TP
`
`Replicon GT1b
`
`0.6 ⫾ 0.04
`⬎100
`ND
`ND
`
`Downloaded from
`
`http://www.jbc.org/
`
` by guest on January 13, 2015
`
`Ki NS5B GT1b
`
`Km NS5B GT1b
`
`␮M
`ND
`ND
`0.023 ⫾ 0.002
`0.141 ⫾ 0.03
`
`␮M
`ND
`ND
`0.22 (CTP)
`0.37 (UTP)
`
`Replicase GT1b
`
`ND
`ND
`0.34 ⫾ 0.1
`1.19 ⫾ 0.09
`
`FIGURE 3. PSI-6130-TP and RO2433-TP are substrates of HCV polymerase
`and chain-terminators. A, sequence of the RNA template and primers. B, the
`incorporation of CTP and CTP analogs was initiated with GG primer according
`as described under “Experimental Procedures.” The nucleotide triphosphates
`included in the reactions were as follows: 10 ␮M CTP, ATP, and UTP (lane 1); 2,
`10, 100, and 1000 ␮M UTP (lanes 2–5); 10 ␮M CTP without UTP (lane 6) or with
`2, 10, 100, and 1000 ␮M UTP (lanes 7–10); 10 ␮M PSI-6130-TP without UTP (lane
`11) or with 2, 10, 100, and 1000 ␮M UTP (lanes 12–15); 10 ␮M 3⬘-dCTP without
`UTP (lane 16) or with 2, 10, 100, and 1000 ␮M UTP (lane 17–20). FL, full-length.
`C, incorporation of UTP and UTP analogs initiated with GGC primer. The
`nucleotide triphosphates included in the reactions were as follows: 100
`␮M CTP, ATP, and UTP (lane 1); 20, 50, and 200 ␮M ATP (lanes 2–5); 100 ␮M
`UTP without ATP (lane 6) or with 2, 20, 50, and 200 ␮M ATP (lanes 7–10); 100
`␮M RO2433-TP without ATP (lane 11) or with 20, 50, and 200 ␮M ATP (lanes
`12–15); 100 ␮M 3⬘-dUTP without ATP (lane 16) or with 20, 50, and 200 ␮M
`ATP (lanes 17–20).
`
`To investigate the molecular mechanism of HCV polymerase
`inhibition by PSI-6130-TP and RO2433-TP, we measured their
`incorporation and chain -termination properties. The incorpo-
`ration of nucleotide and nucleotide analogs by HCV polymer-
`ase NS5B was determined in a gel-based assay using a short
`RNA template (Fig. 3A). Incorporation of CMP, PSI-6130-MP,
`and 3⬘-dCMP was initiated from a 33P-labeled GG primer (Fig.
`3A). Incorporated CMP (Fig. 3B, lane 6) could be further
`extended in the presence of the next nucleotide UTP (Fig. 3B,
`lanes 7–10). PSI-6130-TP and 3⬘-dCTP could also serve as sub-
`strates for HCV NS5B and were incorporated into the nascent
`RNA product (Fig. 3B, lane 11 and 16). After incorporation of
`PSI-6130-MP or 3⬘-dCMP, further extension in the presence of
`the next nucleotide UTP was completed blocked even when
`UTP was present at concentrations up to 1 mM (Fig. 3B, lane
`12–15 and lanes 16–20, respectively). Incorporation of UMP,
`RO2433-MP, and 3⬘-dUMP was initiated from a 33P-labeled
`GGC primer using the same RNA template (Fig. 3A). Incorpo-
`rated UMP (Fig. 3C, lane 6) could be further extended in the
`presence of the next nucleotide ATP (Fig. 3C, lanes 7–10). Full-
`length RNA product was observed in the presence of UTP and
`ATP but absence of CTP (Fig. 3C, lanes 7–10), possibly due to
`misincorporation by NS5B through G-U wobble base-pairing.
`HCV NS5B was also able to incorporate RO2433-MP (Fig. 3C,
`lane 11) and 3⬘-dUMP (Fig. 3C, lane 16) but was unable to
`further extend the incorporated RO2433-MP and 3⬘-dUMP in
`the presence of the next nucleotide ATP (Fig. 3C, lanes 12–15
`and lanes 17–20, respectively). The control samples with GG
`primer and UTP-only as well as GGC primer and ATP-only did
`not lead to further extension of the primers, suggesting the
`incorporation of PSI-6130-MP and RO2433-MP was base-spe-
`cific. Taken together, these results demonstrate that PSI-
`6130-TP and RO2433-TP serve as alternative substrates for
`HCV NS5B and act as functional chain terminators once incor-
`porated into nascent RNA. While this manuscript was under
`preparation it was reported that the incorporation of PSI-
`6130-MP into the nascent RNA by HCV polymerase led to
`chain termination, in agreement with our observations (17).
`Kinetics of Phosphorylation of PSI-6130 in Primary Human
`Hepatocytes—To determine the steady state level of the two
`triphosphates in hepatocytes after exposure to PSI-6130, pri-
`mary human hepatocytes from 4 different donors were incu-
`bated with 2 ␮M PSI-6130 for up to 72 h. The uptake of PSI-6130
`by human hepatocytes was fast, and total intracellular activity
`reached steady state levels at 1 h ofPSI-6130 incubation, the
`earliest time point in this study (Table 2). PSI-6130-TP was
`detectable in hepatocytes from all 4 donors at 6 h after PSI-6130
`incubation and increased with time to reach steady state levels
`
`29816 JOURNAL OF BIOLOGICAL CHEMISTRY
`
`VOLUME 282 • NUMBER 41 • OCTOBER 12, 2007
`
`IPR2018-00119
`
`Page 5 of 10
`
`I-MAK 1005
`
`

`

`Metabolism and Mechanism of Action of ␤-D-2ⴕ-Deoxy-2ⴕ-fluoro-2ⴕ-C-methylcytidine
`
`Downloaded from
`
`http://www.jbc.org/
`
` by guest on January 13, 2015
`
`FIGURE 4. Time course of uptake and metabolism of PSI-6130. A, time
`course of uptake of PSI-6130 and formation of PSI-6130-TP and RO2433-TP.
`B, time course of formation of PSI-6130-MP ⫹ RO2433-TP, PSI-6130-DP, and
`RO2433-TP. The data show mean values from four experiments performed
`with primary hepatocytes from four donors.
`
`PSI-6130-TP and RO2433-TP were quantified at different time
`points up to 72 h (see “Experimental Procedures”).
`The PSI-6130-TP level remained constant up to 1 h after the
`removal of extracellular PSI-6130. Thereafter, PSI-6130-TP
`decreased after a single-phase exponential decay kinetics pat-
`tern with a mean half-life of 4.7 ⫾ 0.6 h (Fig. 6, Table 4). The
`PSI-6130-TP level was below quantification limit at 48 h after
`the removal of extracellular PSI-6130. The RO2433-TP level
`increased slightly to reach steady state level at 1–2 h after extra-
`cellular PSI-6130 removal and remained at steady state level for
`up to 6 h before decreasing toward base-line levels. RO2433-TP
`decreased with a mean half-life of 38.1 ⫾ 16.1 h (Fig. 6, Table 4).
`The levels of unphosphorylated species PSI-6130 and RO2433
`decreased rapidly after removal of extracellular RO1656 (data
`
`TABLE 2
`Time course of uptake and phosphorylation of PSI-6130
`Levels of total intracellular species, PSI-6130-TP, and RO2433-TP were determined
`in hepatocytes incubated with 2 ␮M PSI-6130 for the indicated lengths of time. Data
`shown are mean ⫾ S.D. derived from four independent experiments using hepato-
`cytes from four donors. BQL, below the quantification limit.
`RO2433-TP
`Time
`Total intracellular species
`PSI-6130-TP
`pmol/million cells
`h
`pmol/million cells
`pmol/million cells
`BQL
`1
`30.4 ⫾ 18.8
`BQL
`0.3 ⫾ 0.01a
`6
`32.9 ⫾ 16.8
`0.6 ⫾ 0.5
`0.6 ⫾ 0.3
`16
`34.4 ⫾ 20.8
`0.7 ⫾ 0.6
`1.3 ⫾ 1.1
`24
`36.4 ⫾ 21.3
`1.1 ⫾ 0.7
`2.0 ⫾ 1.1
`48
`31.9 ⫾ 17.1
`1.3 ⫾ 0.6
`2.0 ⫾ 1.3
`72
`34.6 ⫾ 15.4
`1.0 ⫾ 0.4
`a Mean ⫾ S.D. derived from 2 donors as RO2433-TP was only detectable in 2 out of
`4 donors at the 6-h time point.
`
`at 24–48 h. The formation of the triphosphate of the uridine
`metabolite, RO2433-TP, demonstrated a delayed time course
`relative to that of PSI-6130-TP. RO2433-TP was detectable in
`hepatocytes from only 2 of 4 donors at 6 h and in hepatocytes of
`all 4 donors at 16 h. RO2433-TP formation reached steady
`state levels at 48–72 h. RO2433-TP concentrations were
`lower than those of PSI-6130-TP at time points earlier than
`16 h but surpassed those of PSI-6130-TP at 24 h and beyond.
`The mean steady state level concentrations of PSI-6130-TP
`and RO2433-TP after incubation with 2 ␮M PSI-6130 were
`1.3 ⫾ 0.6 and 2.0 ⫾ 1.1 pmol/106 cells at 48 h, respectively.
`Unchanged PSI-6130 was the major intracellular species at
`all time points tested after incubation of human hepatocytes
`with radiolabeled PSI-6130 (Fig. 4).
`To determine whether increasing the exposure of hepato-
`cytes to PSI-6130 will lead to increased uptake of PSI-6130 and
`formation of PSI-6130-TP and RO2433-TP, primary human
`hepatocytes from 3 different donors were incubated with PSI-
`6130 at different concentrations up to a maximal concentration
`of 250 ␮M for 24 h. Total intracellular species, determined by
`total intracellular radioactivity in the cell extracts, increased
`linearly with the extracellular PSI-6130 up to 250 ␮M (Table 3).
`The mean total intracellular species at 250 ␮M extracellular
`PSI-6130 reached 3591 pmol/106 cells, with the unphosphoryl-
`ated PSI-6130 being the predominant species (data not shown).
`The formation of both PSI-6130-TP and RO2433-TP increased
`linearly with the e

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