throbber
6614
`
`J. Med, Chem. 2006, 49, 6614—6620
`
`Synthesis and Pharmacokinetics of Valopicitabine (NM283), an Efficient Prodrug of the Potent
`Anti-HCV Agent 2’-C-Methylcytidine
`
`Claire Pierra,t Agnés Amador,' Samira Benzaria,t Erika Cretton-Scott,! Mare D’Amours,’ John Mao,! Steven Mathieu,?
`Adel Moussa,! Edward G. Bridges,! David N. Standring,' Jean-Pierre Sommadossi,! Richard Storer,’ and Gilles Gosselin*+
`Laboratoire Cooperatif Idenix-CNRS— Université Montpellier Il, Case Courrier 008, Université Montpellier If, Place Eugene Bataillon, 34095
`Montpellier Cedex 5, France, and Idenix Pharmaceuticals Inc., 60 Hampshire Street, Cambridge, Massachusetts 02139, and Laboratoires
`Idenix SARL, Laboratoire de Chimie Médicinale, Cap Gamma, 1682 Rue de la Valsiere, BP 50001, 34189 Montpellier Cedex 4, France
`
`Received March 28, 2006
`
`In our search for new therapeutic agents against chronic hepatitis C, a ribonucleoside analogue,
`2’-C-methylcytidine, was discovered to be a potent and selective inhibitor in cell culture of a number of
`RNAviruses, including the pestivirus bovine viral diarrhea virus, a surrogate model for hepatitis C virus
`(HCV), and three flaviviruses, namely, yellow fever virus, West Nile virus, and dengue-2 virus. However,
`pharmacokinetic studies revealed that 2’-C-methylcytidine suffers from a low oral bioavailability. To overcome
`this limitation, we havesynthesized the 3’-O-L-valinyl ester derivative (dihydrochloride form, valopicitabine,
`NM283)of 2’-C-methylcytidine. Wedetail herein for the first time the chemical synthesis and physicochemical
`characteristics of this anti-HCV prodrug candidate, as well as a comparative study of its pharmacokinetic
`parameters with those of its parent nucleoside analogue, 2’-C-methylcytidine.
`
`®O
`O°
`NH,
`Cl
`q
`SN
`NH
`;
`C
`LA
`wo
`Homo. N°
`20=-/
`Hts
`“Fay
`HO
`OH
`Oxe3
`OH
`2'-C-methyluridine(2) ot=i
`
`HO
`
`3
`
`3-O-valinyi ester of
`2'-C-methylcytidine (3)
`(dihydrochloride salt)
`
`HOF
`
`NH
`>
`‘e
`a
`on
`L Hic?
`HO
`OH
`2'-C-methylcytidine(1)
`
`Introduction
`
`Hepatitis C virus (HCV) hasinfected an estimated 170 million
`individuals, 3% of the world’s population.’ The virus establishes
`a persistent infection in the majority of cases, leading to chronic
`hepatitis that often develops into cirrhosis and, in many cases,
`causes hepatocellular carcinoma.! There is no vaccine available
`against HCV, and current
`therapies, namely, pegylated or
`nonpegylated interferon-c (IFN-c) monotherapy and combina-
`tion of IFN-c with oral ribavirin, are expensive, often poorly
`tolerated, and effective only in half of the patient population.27
`Therefore, there is an urgent need to develop new and more
`effective therapies in response to this important unmet medical
`need,*
`
`In the course of our HCV program, we recently discovered
`that 2’-C-methylcytidine (1, Figure 1) is a potent and selective
`inhibitor of Flaviviridae virus replication in cell culture.More
`particularly, 1 inhibited the replication of the bovine viral
`diarrhea virus (BVDV,a pestivirus surrogate model for HCV),’
`eliminated persistent BVDV infection at nontoxic concentra-
`tions, and was synergistic in combination with interferon-o,
`but not with ribavirin.’ Compound 1 has no activity against
`human immunodeficiency virus (HIV)or against DNA viruses.
`In contrast to ribavirin, which is not effective alone in reducing
`vital RNAlevels but stimulates the immune boosting capacity
`of interferon-a when used in combination, 1 is the first example
`of an anti-RNA virus agent that is active via a nucleoside
`analogue mode of action. Thus, in primary human hepatocyte
`cultures, in a human hepatomacell line (HepG2), and in a bovine
`kidney cell
`line (MDBK), 1 is converted into its major
`metabolite, 2’-C-methylcytidine-5’-triphosphate, along with
`smaller amounts of 2’-C-methyluridine-5‘-triphosphate, resulting
`from deamination. The active metabolite 2’-C-methylcytidine
`
`
`* Corresponding author. Tel: + 33-4-67143855. Fax: + 33-4-67549610.
`E-mail: gosselin@univ-montp2.fr.
`¥ Université Montpellier I.
`1 Tdenix Pharmaceuticals Inc.
`§ Laboratoires Idenix SARL.
`¥ Present address: VASToxplc, 9} Milton Park, Abingdon, Oxfordshire,
`OX14 4RY, U.K.
`
`Figure 1. Structures of compounds 1—3.
`
`triphosphate is a competitive inhibitor of purified BVDV RNA
`polymerase in vitro (Kj = 160 nM).°
`Preliminary pharmacokinetic studies in animals revealed that
`further development of 1 would be hampered byits low oral
`bioavailability. To overcome this limitation, we devoted our
`efforts to the design of 2’-C-methylcytidine prodrugs with more
`favorable oral absorption profiles. From a literature survey, it
`was found that a broad variety of amino acid ester derivatives
`have been studied and successfully employed as nucleoside
`prodrug forms, Moreparticularly, efficacy of such derivatives
`has been proved in the case of valacyclovir, the L-valiny] ester
`of acyclovir (Figure 2).!%'! After active absorption via peptide
`transport mechanism, valacyclovir is rapidly and almost com-
`pletely converted into acyclovir by enzymatic hydrolysis,
`increasing considerably the oral bioavailability and celular
`uptake of the parent drug.’°" The efficacy of L-valinyl
`derivatives has been also demonstrated in the case of ganciclovir
`(GCV), since valganciclovir (L-valinyl ester of GCV, Figure 2)
`has an oral bioavailability 10-fold higher than the parent
`nucleoside.'23 Recently, as part of our hepatitis B program,
`we have synthesized and studied several L-valiny! ester prodrugs
`of 2’-deoxy-f-L-cytidine (1-dC) in order to improve the oral
`bioavailability of L-dC. Among them, the 3’-O-L-valinyl ester
`of L-dC (Val-L-dC,valtorcitabine, Figure 2) emerged as the most
`attractive L-dC prodrug! and is currently in phaseII clinical
`studies.'5
`Onthe basis of these considerations, we decided to synthesize
`and study the 3’-O-.-valinyl ester derivative (NM283, valop-
`
`10.102 1/jm0603623 CCC: $33.50
`© 2006 American Chemical Society”
`Published on Web 10/06/2006
`s
`
`CLARK EXHIBIT 2125
`Sommadossi v. Clark
`Contested Case 105,871
`
`GIL2018
`I-MAK, INC. V GILEAD PHARMASSETLLC
`IPR2018-00120
`
`1
`
`GIL2018
`I-MAK, INC. V GILEAD PHARMASSET LLC
`IPR2018-00120
`
`

`

`Synthesis and Pharmacokinetics of Valopicitabine

`
`qe®
`
`N
`e
`O
`N7
`HgN. tl
`L.~ loo
`:
`Valaciciovir
`
`| ae
`SN
`>NH2,
`
`o
`
`® 0
`NH,
`Ci
`>
`Rea ,
`=
`Nagy _OH
`j
`
`oO
`
`NH
`
`Ox
`
`fe)
`
`.
`
`N
`if
`
`HO
`
`eo,
`9
`ll
`
`
`Cl HNg5og ON v7 20 iy
`E
`~/
`crm
`H3N"
`Val-L-dc
`
`Journal of Medicinal Chemistry, 2006, Vol. 49, No. 22 6615
`
`fluoride in tetrahydrofuran (TBAF/THF), followed by acidic
`hydrolysis using a saturated solution of hydrogen chloride
`in ethyl acetate,”* led to the target prodrug 3 as a dihydro-
`chloride salt.
`
`Although this conventional synthetic route afforded the
`desired prodrug in a satisfactory overall yield of 52%, several
`chemical difficulties (instability of the formamidine intermediate
`4 and partial racemization of the L-valine during the coupling
`step), as well as cost issues (use of expensive feri-butyldiphe-
`nylsilyl group), made scale-up using this initial route unattrac-
`tive. In our search for a more direct and scalable synthetic
`process, we then developed a new synthetic strategy based on
`the direct condensation of 1 with L-Boc-valine, followed by
`removal of the N-zert-butyloxycarbony! group, thus reducing
`the number ofthe synthetic steps from 5 to 2 (Scheme 2). We
`decided to explore this direct coupling approach after examining
`a report by McCormick et al, who described the room-
`temperature reaction of unprotected guanosine with Boc-
`anhydride in the presenceoftriethylamine, DMAP, and DMSO.-
`This reaction selectively gave 3’-Boc-guanosinein 55% yield.23
`In our first attempt, we reacted compound 1 in DMSO with
`carbonyldiimidazole(CDD-activated L-Boc-valine in the presence
`of triethylamine and DMAP. Thereaction did not progress at
`all when kept at room temperature. The reaction produced the
`desired product and several byproducts when heated at 50 °C
`using 4 equiv of CDI-activated L-Boc-valine. Optimum selectiv-
`ity was achieved by using 1.1 equiv of CDI-activated L-Boc-
`valine and runningthe reaction for only 1 h at 80 °C. We used
`only 0.1 equiv of DMAPin order to avoid the racemization of
`the L-valine. Other coupling reagents such as EDC and N,N’-
`dicyclohexylcarbodiimide (DCC). did not produce desired
`product, and DMF was difficult to change due to the insolubility
`of compound 1 in most organic solvents, HPLC analysis showed
`68% of the desired compound, 11% of starting nucleoside 1,
`and the two 3’,5’/3’,N*-divalinyl ester byproducts. Pure product
`3 was obtained in 54% yield and 99% purity by using simple
`acid—baseextraction, eliminating the extensive chromatographic
`step. Thus, this tume- and labor-saving alternative process could
`be accomplished without
`involvement of chromatographic
`
`Valganciclovir
`
`~
`
`Figure 2, Structures of valacyclovir, valganciclovir, and val-L-dC.
`
`in order to improve the oral
`icitabine, 3; Figure 1) of 1
`bioavailability of the parent nucleoside.
`:
`Results and Discussion
`
`Synthesis. The 3’-O-valinyl ester prodrug of 2’-C-methyl-
`cytidine (3) was first prepared by the route described in Scheme
`1. Our multistep sequence involved successive protections of
`the exocyclic amino function and of the 5’-hydroxyl group of
`1, followed by condensation with N-tert-butyloxycarbonyl-L-
`valine (L-Boc-valine) and,
`finally,
`total deprotection. N,N-
`Dialkylformamidinesand especially N,N-dimethylformamidine
`have been widely used as protective groups for exocyclic amino
`function of nucleosides.'®!? In the present work, the synthesis
`of N*-[(dimethylamino)methylene]-2’-C-methyleytidine (4) was
`carried out following a procedure described by Kerr et al for
`the protection of 1 using N,N-dimethylformamide dimethylac-
`etal.'® Selective-silylation of the 5’ primary hydroxy! group using
`a reported method! led to the protected key intermediate 5.
`Condensation of 5 with L-Boc-valine using the coupling agent
`N’-(3-dimethylaminopropyl)-N-ethylcarbodiimide hydrochloride
`(EDC),?° with 4-(dimethylamino)pyridine (DMAP) asa cata-
`lyst,!! provided the N-Boc-protected ester derivative 6. Finally,
`desilylation of 6 with excess ammonium fluoride (NH4F) in
`methanol,?! an economical alternative to tetrabutylammonium
`
`Scheme 1. A Conventional Route for the Synthesis of 37
`NH,
`(oy
`HO 0 Nv
`ars
`HO
`OH
`1
`
`Oo
`
`athe
`
`HO
`
`ae
`"
`
`o
`
`=e: ;
`Hat
`HO
`4
`(80% first crop
`
`crystatlization)
`
`WONG
`.
`oN
`CO
`ls, INTO
`pao:
`Co , AG
`|
`~" HO
`OH
`5
`
`ii
`
`| iii
`

`NH,
`‘en
`SN
`HOY 9. NO
`Hgts
`—/
`Oxy®
`OH
`oe, ’ i
`;
`3'-O-valinyl ester of 1
`(hydrachloride farm, 3, 81%)
`
`Ct
`
`NH3
`CY
`SN
`HO 9. NO
`
`c
`v
`7
`9 0x0
`oon i
`
`iv
`
`NANG
`t
`“Sy
`0. no
`-+si-o
`Hay
`(
`O
`6 O9 OH
`os my
`x NH
`
`7
`(78% for 3 steps)
`(i) Mez2NCH(OMe)2, DMF,rt, 1.5 h; (ii) TBDPSCI, imidazole, pyridine,rt, 6 h; (iii) N-Boc-L-valine, EDC, DMAP, CH3CN/
`@ Reagents and conditions:
`DMF,tt, 2 d; (iv) NHaF, McOH,reflux, 3 h; (v) HCI/EtOAc, EtOAc,rt.
`
`2
`
`€
`

`

`6616
`
`Journal of Medicinal Chemistry, 2006, Vol. 49, No. 22
`
`Pierra et al.
`
`Scheme 2. Alternative Route for the Synthesis of 3¢
`
`NH
`
`NH»
`
`be
`Ce
`Hoy gp NO Hoya NO
`ean
`—iil
`Tas
`HO
`OH
`9 On 0
`OH
`J. Aly
`x? NH}
`7
`(54%)
`
`1
`

`NH;
`
`Co
`INO
`
`it
`
`HOsuo.
`Age
`0.0 OH
`aoe
`i 4
`cl
`NH3”
`|
`3'-O-valinyl ester of4
`(dihydrochloride sait, 3,
`
`Cl
`
`JTable 1. Pharmacokinetic Parameters of Compounds 1 and 3,
`Following a Single Oral Administration of 3 to Sprague-Dawley Rats*
`prodrug3
`compd 1
`dose
`equiv dose
`AUC
`Tin
`Cmax
`Tmax
`SF
`
`(mg/kg)
`(mg/kg)
`drug
`(wgb/mL)
`(bh)
`(agin)
`(bh)
`(%)
`100
`72
`3
`8.95
`0.64
`3.62
`i100
`-
`30.0 7.10 6.12 2.0] 33.6
`
`
`
`
`
`
`# AUCy-, area under the plasma concentration~time curve from time
`0 to 4 Tro:
`terminal elimination half-life. Cyax: maximum plasma
`concentration. Tinax;
`time to maximum plasma concentration. F: apparent
`oral bioavailability calculated on the basis of a dose-normalized AUC value
`of 1.24 (ug b/mL) from a single iv dose of 1 to the rat.
`
`@ Reagents and conditions:
`THF; (Gi) HCVEtOH.
`
`(i) N-Boc-L-valine, CDI, DMAP, TEA, DMF/
`
`At pH 4.5
`
`
`
` Relativeconcentration
`
`0
`
`at
`“O-~
`
`3in=3)
`4(n=3)
`
`
`
`
`Z
`
`2s
`
`s 5a Ga
`
`|
`|
`S
`“160
`gO]
`
`{
`
`4
`
`
`+
`' ag
`8
`is
`ee
`32
`40
`48
`Time (h)
`
`Figure 4, Mean plasma concentration of compounds 1 and3, following
`a single oral administration of 3 to Sprague-Dawleyrats.
`
`human liver cytosol, and 89 fractions are not surprising and
`depend on the activity/quantity of enzyme(s) present in these
`samples. In all cases, no further metabolism of 1, such as
`deamination to its corresponding uridine metabolite 2, has been
`observed.
`Protein Binding of Compounds 1 and 3 to Rat Plasma.
`Thein vitro binding of compounds 1 and 3 to rat plasmaproteins
`was investigated bytheultrafiltration method using radiolabeled
`test articles ((3H]-2’-C-methylcytidine and [°H]-3’-O-valinyl
`ester of 2’-C-methyleytidine) at a concentration of 20 “M for
`each compound.As expected for nucleosides, the protein binding
`for the two compounds was low, with 7% and 5% boundfor 3
`and 1, respectively.
`Oral Bioavailability and Pharmacokinetic Studies of
`Compounds 1 and 3. The pharmacokinetics (PK) of prodrug
`3 were evaluated in Sprague—Dawley rats in a mass balance
`using radiolabeled 3. Both intact and bile duct cannulated (BDC)
`rats were used in this study. PK parameters of 1 and its 3’-O-
`valinyl ester 3 are presented in Table 1, Plasma concentration—
`time profiles of 1 and 3 are shown in Figure 4.
`Following a single oral dose of ['4C]-3’-O-valinyl ester of
`2’-C-methylcytidine (3) at 100 mg/kg (as free base), urine, feces,
`and cage rinses were collected for a period of 72—168 h
`postdoseto evaluate the excretion of radioactivity and the overall
`mass balance. Plasma samples were collected over a 48-h period
`postdose to study the PK of1 and3.In intactrats, fecal excretion
`was also the predominant route of elimination of total radio-
`activity, accounting for 63.5% of the administered dose.
`Excretion of radioactivity occurred largely within the first 24 h
`(averaging 60% of the dose). Urinary excretion accounted for
`31.9% of the administered dose. The mean total recovery of
`radioactivity, in urine, feces, and cage rinses, was 96.6%. Bilary
`excretion oftotal radioactivity in BDC rats accounted for a mean
`
`At pH 7.2
`
`10
`
`
`
`Relativeconcentration o >
`
`0
`
`§00 1000 1600 2000
`
`(
`
`Incubation time (min)
`®o
`s
`NH,
`Cl
`N
`
`HOq of
`Hats
`Hi
`OH
`A= 2-C-methylcytidine (1)
`Kinax = 273.6 am
`
`5000 10000 15000 20000
`tncubation time (min)
`ee
`NH,
`Cl
`Shy
`
`aa ae
`Test
`OH
`
`:
`
`HOS
`on?
`ee
`cl
`HGNT TT
`O = 3-0-valinyl ester of 2'-C-methylcytidine (3)
`A max 5 273.5 1M
`
`Tage 6.1 days at pH 4.5
`Tae 3.9 hours atpH7.2
`
`Figure 3. Kinetic curves ofhydrolysis of prodrug 3 at pH 4.5 and 7.2.
`
`purifications, and compound 3 was obtained in high purity using
`simple crystallization.
`AqueousSolubility, Lipophilicity, and Chemical Stability
`of Prodrug 3. To be considered as a suitable prodrug for oral
`administration, the 3’-O-valinyl ester of 2’-C-methylcytidine (3)
`should possess adequate solubility in aqueous media in order
`to dissolve in the small
`intestine,
`thus being available for
`absorption. The aqueous solubility of 3 has been determined in
`comparison with the solubility of the parent nucleoside 1. Thé
`concentration of saturated solutions of 1 and 3 in water were
`32 and 423 g/L, respectively. Both compounds were found to
`be highly soluble in aqueous media, which has been cor-
`roborated by their low distribution coefficient (log P) values:
`—0.965 and —1.34 for 1 and 3, respectively.
`The main aim ofstability studies was to determine whether
`the prodrug of 2’-C-methylcytidine would be sufficiently
`chemically stable in the gastrointestinal tract before its absorp-
`tion. Prodrug 3 appearedto be fully stable at pH 1.2 but was
`hydrolyzed at pH 4.5 and 7.2 into the parent drug 1 following
`first-order kinetics. The half-lives of 3 at pH 4.5 and 7.2 were
`6.1 days and 3.9 h,respectively (Figure 3). It is noteworthy
`that 1 was fully stable atall the acidic and neutral studied pHs.
`Stability in Human Blood, Plasma, and Liver Cytosol.
`Prodrug 3 was rapidly converted into 1 in both human plasma
`and whole blood, exhibiting in vitro half-life values of 130 and
`40 min, respectively. This rapid conversion is primarily due to
`the presenceof esterases in blood and plasma. Conversion of 3
`into 1 was also observed in humanliver cytosol and $9fractions,
`with approximately 30% of 3 converted into 1 within | h.
`Differences in half-life for prodrug 3 in plasma, whole blood,
`
`3
`
`

`

`Synthesis and Pharmacokinetics of Valopicitabine
`
`Journal of Medicinal Chemistry, 2006, Vol. 49, No. 22 6617
`
`of 0.3% of the dose over the 72-h collection period. In this
`group, fecal excretion was the predominantroute of elimination,
`accounting for a mean 65.9% of the dose. A mean 29.8% of
`the dose was recovered in urine. Excretion of radioactivity was
`largely completed by 24 h in BDC rats. On the basis of the
`combined mean recovery of radioactivity in bile and urine, it
`was apparentthat at least 30.0% ofthe [!4C]-3’-O-valiny] ester
`of 2’-C-methylcytidine (3) oral dose was absorbed. The mean
`recovery of total radioactivity in this group was 97.0% of the
`administered dose, In the group following a single dose of '4C-3
`at 100 mg/kg (as free base), maximum plasma concentrations
`of total
`radioactivity (averaging 11.9 wg equiv/mL) were
`achieved 1—2 h after the oral dose. Plasma concentrations of
`3, determined by LC/MS/MS, were greatest 0.5—1 h after
`dosing, and Cmax averaged 3.62 ug/mL. Plasma 3 concentrations
`decayed with a half-life of 0.64 h, and no prodrug 3 was detected
`in plasma by 12 h, Plasma concentrations of 1, determined by
`LC/MS/MS, were greatest 1-2 h after dosing, and Cinax
`averaged 6.12 zg/mL. Plasma concentrations of 1 decayed with
`a half-life of 7.1 h. The AUC of 1 was morethan 3-fold greater
`than the AUC of 3, 2’-C-Methyluridine (2) was not detected in
`rat plasma (<0.1 wg/mL).
`The pharmacokinetics of 1 were also studied in Sprague—
`Dawleyrats in a toxicokinetic (TK) study in which compound
`1 was administered intravenously daily for 15 days. There were
`three dose groups (60, 120, and 300 mg/kg/day) in the study,
`and TK. sampling was conducted on day | following the first
`dose and on day 15 after the last dose. For the scope ofthis
`paper, only the TK. parameters of the 60 mg/kg/day dose group
`on day | are presented. Followinga single iv dose of compound
`1 to rats at 60 mg/kg, the mean plasma Cmax of 1 was 75.6
`#e/mL and the mean AUC was 74.4 ug h/mL. The mean AUC
`value of 1 from this dose group was compared to the AUC value
`of 1 from the prodrug 3 rat study above, and an apparentoral
`bioavailability was calculated for 1. Thus, the apparent oral
`bioavailability of 1 following oral administrationof the valiny]
`ester derivative 3 at 100 mg/kg (as free base) was calculated to
`be 33.6%.
`
`Conclusion
`
`3’-O-Valinyl ester of 2’-C-methylcytidine (dihydrochloride
`salt, NM283, valopicitabine, 3) has been synthesized in order
`to improve the oral bioavailability of the parent compound 2’-
`C-methylcytidine (1). For that purpose, two different strategies
`have been developed, both starting from 1. The first one is a
`conventional route that involves successive protection steps, and
`the second one is more appropriate for large-scale synthesis and
`is based on selective 3’-O-esterification. Physicochemical,
`pharmacokinetic, and toxicokinetic studies have shown that
`compound 3 is an acid-stable prodrug of 1 with excellent
`pharmacokinetic and toxicokinetic profiles, Prodrug 3 is rapidly
`converted into compound 1 in both human plasma and whole
`blood, probably due to the presence of esterases. The apparent
`oral bioavailability of the parent drug 1 following oral admin-
`istration of the prodrug 3 is 34%.
`Thus,on thebasis ofits ease of synthesis, its physicochemical
`properties, and pharmacokineticprofile, 3’-O-valinyl ester 3 has
`emerged as a promising prodrug of 2’-C-methyleytidine (1). The
`. 3’-O-valinyl ester of 2’-C-methyleytidine (3, NM283, valopic-
`itabine) is currently undergoing phase IIb clinicaltrials for the
`treatment of chronic HCV infection.”
`
`Experimental Section
`'H NMR spectra were
`General Methods for Chemistry.
`recorded at ambient temperature on a Bruker AC 200 MHz, 250,
`
`'H NMR chemical shifts (4) are
`300, or 400 MHz spectrometer.
`quoted in parts per million (ppm) referencedto the residual solvent
`peak [dimethyl sulfoxide (DMSO-d,)] set at 2.49 ppm. The accepied
`abbreviations are as follows: s, singlet; d, doublet; t, triplet; m,
`multiplet. FAB mass spectra were recorded in the positive-ion or
`negative-ion mode on a JEOL DX 300 mass spectrometer operating
`with a JMA-DA 5000 mass data system and using a mixture of
`glycerol and thioglycerol (1/1, v/v, G--T) as the matrix. Melting
`points were determined in open capillary tubes with a Biicht B-545
`apparatus and are uncorrected, UV spectra were recorded on an
`Uvikon XS spectrophotometer. Elemental analyses werecarried out
`by the Service de Microanalyses du CNRS, Division de Vernaison
`(France). Thin-layer chromatography (TLC) was performed on
`precoated aluminum sheets of silica gel 60 Fos, (Merck, Art. 5554),
`visualization of products being accomplished by UV absorbance
`and by charring with 10% ethanolic sulfuric acid with heating or
`with a 0.2% cthanolic ninhydrin solution for compounds bearing
`an amide function.
`i
`Column chromatography was carried outon silica gel 60 (Merck,
`Art. 9385). Evaporation of solvents was carried out in a rotary
`evaporator under reduced pressure. All moisture-sensitive reactions
`were carried out under rigorous anhydrous conditions under an
`argon atmosphere using oven-dried glassware. Solvents were dried
`and distilled prior to use and solids were dried over POs under
`reduced pressure. Analytical high-performance liquid chromatog-
`raphy (HPLC) studies were carried out on a Waters Associates unit
`(multisolvent delivery system, 717 autosampler injector, 996
`photodiode array detector and a Millenium data workstation) using
`a reverse-phase analytical column (Nova-Pak Silica 60 A, 4 ym,
`Cis, 150. x3.9 mm). The compound to be analyzed was eluted
`using a linear gradient of 0-50% acetonitrile in 20 mM triethy-
`lammonium acetate buffer (TEAC, pH 7) programmed over a 30-
`min period with a flow rate of 1 mL/min.
`General Precedure for Aqueous Solubility Studies. Aqueous
`solubilities were determinedin distilled water at room temperature.25
`An excess of studied compounds was added to aqueoussolution,
`and the suspension was shaken and centrifugated. Samples of
`supernatant were analyzed by HPLC (same conditions as described
`in General Methods for Chemistry), and the concentration of
`saturatedsolution was determined accordingto a calibration curve.
`The experiment was repeated three times.
`General Procedure for Distribution Coefficient Studies.
`Distribution coefficients between 1-octanol and an aqueous phase
`(phosphate buffer solution 0.02 M) were determined at room
`temperature using a shake-flask procedure.An aliquot of a 107?
`M aqueous solution of studied compounds was diluted to 1 mL
`with the aqueous phasepreviously saturated with octanol. An equal
`volume of octanol, previously saturated with the aqueous phase,
`was added to give a total volume of 2 mL. The mixture was shaken
`vigorously. The two phases were centrifugated and separated.
`Samples of each phase were collected and analyzed by HPLC (same
`conditions as described in General Methods for Chemistry). UV
`absorbances for both phases were measured at the respective
`maximum wavelength. The distribution coefficient was calculated
`from theratio of the area of the signal detected in the octanol and
`aqueous phases. Each experiment was repeated twice.
`GeneralProcedure for Chemical Stability Studies. Chemical
`hydrolysis rates were determined in KCI—-HCI buffer 0.135 M
`solution (pH 1.2, 37 °C), acetate buffer 0.02 M solution (pH 4.5,
`37 °C), and phosphate buffer 0.02 M solution (pH 7.2, 37 °C).
`Solutions (10-4 M) of 3 have been incubated at 37 °C in buffer
`solutions. Aliquot samples were collected at different timeintervals
`and analyzed by HPLC(the sameconditions as described in General
`Methods for Chemistry), Rates of decomposition have beeneasily
`determined using a method developed in our laboratory and based
`on pseudo-first-order kinetic models.??
`General Procedure for in Vitro Blood and Plasma Stability
`Studies. A stock solution of 3 was aliquoted (1 mg/mL stored in
`MeOH at —20 °C), dried down, and resuspended to 20 zg/mL with
`either human plasma or whole blood (Bioreclamation Inc., Hicks-
`ville, NY) preheated to 37 °C. Aliquot samples were collected at
`
`4
`
`

`

`6618
`
`Journal of Medicinal Chemistry, 2006, Vol. 49, No, 22
`
`Pierra et al.
`
`3’-O-valinyl ester of 2’-C-methylcytidine, and 1 and 3 were
`different time intervals 0, 2, 5, 10, 20, 40, and 60 min for blood
`evaluated. Blood samples were collected from group 3 male rats
`studies and 0, 30, 70, 100, and 140 minfor plasmastudies. Samples
`predose and 0.25, 0.5, 1, 2, 3, 4, 8, 12, 16, 24, and 48 h postdose
`were quenched.in 5 volumes ofice-cold solvent consisting of 50%
`(3 rats/time point), and the separated plasma was analyzed by LSC
`acetonitrile/50% methanol and centrifuged at 16 000 ref for 3 min.
`and by LC/MS/MS. The LC/MS/MS method was validated ac-
`Supernatant was removed, dried downina centrifugal concentrator,
`cordingto ICH guidelinesforbioanalytical method validation prior
`resuspended in mobile phase (5% MeOH~95% potassium phos-
`to sample analysis. A daily cage rinse, a cage wash, and a cage
`phate, monobasic, pH 5.0), filtered (0.2 4m nylon Spin X tubes),
`wipe at termination were also performed for groups 1 and 2. All
`and analyzed by reverse-phase HPLC using an Agilent model 1100
`postdose samplesofbile, urine, feces, plasma, and cage residues
`instrament with automatic injection and a diode-array spectropho-
`(cage rinse, cage wash, and cage wipe) were analyzed for
`tometric detector. The mobile phase consisted of buffer A (25 mM
`radioactivity by LSC. Mean plasma concentrations of1 andits 3’-
`potassium phosphate, pH 5) and buffer B (methanol), A linear
`O-valinyl ester were uscdto calculate descriptive pharmacokinctic
`gradient from 5% to 100% buffer B was run over 15 min. The
`parameters by noncompartmental analysis using WinNonlin 4.1
`HPLC system used a Cjg Columbus column (5 zm, 250 x 4,6 mm,
`(Pharsight).
`Phenomenex, Torrance, CA). Authentic standards were used to
`The 15-day toxicokinetic study of nucleoside analogue 1 was
`identify the peaks in HPLC. This assay was done onetime (blood
`conducted to evaluate the pharmacokinetics of 1
`following iv
`and plasma from one donor) in duplicate.
`administration to Sprague— Dawley rats. Compound 1 was admin-
`General Procedure for Metabolic Stability Studies. Theability
`of derivative 3 to function as substrate for relevant metabolic
`istered once daily to rats intravenously for 15 days at dose levels
`of 60, 120, and 300 mg/kg/day. Each dose group consisted of eight
`enzymes was examined using human liver subcellular fractions
`animals/gender. On days 1 and 15, blood samples were collected
`obtained from BD Gentest (Wobum, MA). The prodrug was
`from thefirst set of four rats/sex/group at predose and approximately
`incubated for 1 h at 50 wg/mL with | mg/mL liver 89 orliver
`20 min and 2 h postdose and from the secondset of four rats/sex/
`cytosol. Prior to HPLC analysis, samples were quenched in 5
`group at approximately 5 min and 1 and 4 h postdose, Plasma
`volumes of ice-cold solvent (50% acetonitrile/50% methanol) and
`samples were analyzed for 2’-C-methylcytidine and 2’-C-methy-
`centrifuged (16 000 ref for 3 min). Supernatant was removed,dried
`luridine by a validated bioanalytical method (LC/MS/MS), and
`down in a centrifugal concentrator, resuspended in mobile phase
`composite PK parameters were calculated by noncompartmental
`(5% McOH—95% potassium phosphate, monobasic, pH 5.0),
`analysis using WinNonlin 4.1 (Pharsight).
`filtered (0.2 4m nylon Spin X tubes), and analyzed via HPLC (same
`conditions as described in General Procedure for in vitro Blood
`For LC/MS/MSanalysis, compound 1,its 3’-O-valinyl ester, and
`added internal standards were extracted from plasma (50 #L) using
`and Plasma Stability Studies).
`protein precipitation with acetonitrile (0.5 mL). The supernatant
`General Procedure for Protein Binding Studies. The protem
`wasdried, reconstituted, and analyzed by LC/MS/MS using HPLC
`binding of prodrug 3 and parent nucleoside 1 in rat plasma was
`coupled to a PE Sciex API3000 tandem mass spectrometer with a
`investigated using the ultrafiltration centrifugation method at a
`Turbo IonSpray interface. The HPLC system used an Alltech
`concentration of 20 uM for each. The studies used both nonradio-
`Platinum Cyg column (@ ym, 53 x 7 mm, Alltech Associates,
`labeled and radiolabeled test articles. The [*H]-3’-O-valinyl ester
`Deerfield, IL), HPLC elution was carried out using a gradient of
`of 2’-C-methylcytidine and [?H]-2’-C-methylcytidine, provided by
`ammonium acetate (10 mM,pH 4) andacetonitrile. The flow rate
`Moravek Biochemicals, Inc. (Brea, CA), were prepared by tritium
`was 2.5. mL/min with a split of 0.5 mL/min into the mass
`exchange (catalyst and labile tritium were removed during purifica-
`spectrometer. Each analyte was detected by multiple reaction
`tion). To achieve the desired concentrations, concentrated stock
`solutions for each test article containing the appropriate amounts
`monitoring (MRM)underthe positive ion mode using the precursor
`to product ion pair of m/z 357 to 112 (collision energy 27 eV) and
`of the radiolabeled and nonradiolabeledarticles were prepared in
`m/z 258 to 112 (collision energy 20 eV), for 3 and 1, respectively.
`HPLC grade water. Rat plasma (1 mL) was then added to each’
`Quantitation was achieved by constructing a calibration curve by
`tube and vortexed. The sample tubes were incubated at 37 °C for
`Lh.
`weighted linear regression of the ratio of the analyte peak area to
`that of the addedinternal standard. Calibration standards were in
`Centrifee micropartition units (1-mL capacity, Amicon Inc.) with
`blank rat plasma. The calibration range was 0.01-5 yg/mL in
`a molecular weight cutoff of 30 000 Da were used to separate the
`plasma for 3 and 0.05~25 yg/mL in plasma for the parent
`unbound from the bound test article by membrane filtration.
`nucleoside 1.
`,
`Fortified plasma (0.6 mL) was added to the sample reservoir portion
`N‘.[(imethylamino)methylene]-2-C-methyl-f-p-cytidine (4).
`of the ultrafiltration device. Plasma samples were centrifuge for
`A solution of 15628 (1.65 g, 6.43 mmol) in V,N-dimethylformamide
`10 min at 37 °C and. 1800g so that the amount of sample filtered
`(DMF, 32 mL) wastreated with dimethylformamide dimethylacetal
`ranged between 20 and 50% ofthe total volume. After centrifuga-
`(8.2 mL, 61.73 mmol) andstirred for 1.5 h at room temperature."
`tion, the ultrafiltrate of each sample was collected, and 10 wL
`The solution was evaporated under reduced pressure and coevapo-
`aliquots were analyzed for radioactivity using liquid scintillation
`rated with ethanol. Crystallization from ethanol/ether yielded the
`counting (LSC) on a Beckman Coulter LS6500. Protein binding
`hitherto unknown title compound4(first crop = 1.21 g, 60%,
`determinations were done in duplicate for cach test article.
`second crop slightly impure on TLC = 0.46 g, 23%) as crystals.
`General Procedure for Pharmacokinetic Studies. The phar-
`All
`the following physicochemical characteristics have been
`macokinetic and massbalance study of the [!C]-3’-O-valinylester
`determined on the crystals issued from the first crop crystalliza-
`of 2’-C-methylcytidine ('4C label at C-2, Moravek Biochemicals,
`tion: mp 169-172 °C; 'H NMR6 8.62 (s, 1H, N=CH), 8.17 (d,
`Inc.) was conducted in three groups of male Sprague—Dawleyrats.
`1H, H-6, Js—s = 7.3 Hz), 5.91 (m, 2H, A-1’, H-5), 5.16 (m, 1H,
`A single oral dose of the [!4C]-3’-O-valinyl ester of 1 was
`OH-5’, D2O exchangeable), 5.06 (s, 1H, OH-2’, D,O exchangeable),
`administered as a solution in 0.01 N HCIat a target dose of 100
`3.8—3.5 (m, 4H, H-3’, H-4’, H-5’, and H-5”), 3.15 and 3.02 (2s,
`mg/kg (as free base) to the three groups ofmale rats after fasting
`6H, N(CHs)2), 0.92 (s, 3H, CH3); FAB>0 m/z 625 (2M + Hyt,
`overnight. Group | was used to characterize the excretion of the
`313 (M + H)*, 167 (B + 2H)'; FAB<0 m/z 419 (M+ T — H),
`radioactivity derived from the [!4C]-3’-O-valinyl ester of 2’-C-
`403 (M + G — H)-, 311 (M — H)-, 165 (B); HPLC tp = 5.96
`methylcytidine in urine and feces. Urine and feces were collected
`min; Amex = 316.1 nm; UV (H20): Amax = 313 nm (€ 30 200), Amin
`up to 168 h postdose from group 1 rats. Group 2 comprised bile
`= 242 nm (€ 3600);
`fa]*®p = +134.7 (c = 0.95, H20). Anal.
`duct cannulated (BDC) male rats and was used to characterize the
`(Cy3H29N405°0.1H20) Gs H, N.
`excretion of the radioactivity derived from the [‘4C]-3’-O-valinyl
`3’-O-[N-(tert-Butoxycarbonyl)-L-valinyl]-2’-C-methyl-

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