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,l Agnes Amador,i Samira Benzaria,’r Erika Cretton—Scott,I Marc D’Amours,I John Mao,t Steven Matthieu}
`Adel Moussa,I Edward G. Bridges} David N. Standring,I Jean-Pierre Sommadossi,1 Richard Storer,§'v and Gilles Gosselin‘hl
`Laboraloire Coopémtifldenix—CNRS— Université MontpeNiet‘ 11, Care Courrier 008, Université Mom‘pellier 11, Place Eugene Bataillon, 34095
`Montpellt'er Cedex 5, France, and 102er Pharmaceuticals Inc, 60 Hampshire Street, Cambridge, Massachusetts 02139, and Laboratoires
`Idem'x SARL, Labaratoire de Chimie Médicinale, Cap Gamma, 1682 Rue de la Valsiére, BP 50001, 34189 Montpellter 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
`RNA viruses, including the pestivirus bovine viral diarrhea virus, a surrogate mode] 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 have synthesized the 3’-O—L—valinyl ester derivative (dihydrochloride form, valopicitabinc,
`NM283) of 2’-C—methylcytidine. We detail herein for the first time the chemical synthesis and physicochcmical
`characteristics of this anti—HCV prodiug candidate, as well as a comparative study of its pharmacokinetic
`parameters with those of its parent nucleosidc analogue, 2’—C—methylcytidine.
`
`Introduction
`
`Hepatitis C virus (HCV) has infected 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.1 There is no vaccine available
`against HCV, and current
`therapies, namely, pegylated or
`nonpegylated interferon—(1 (IFN-o.) monotherapy and combina~
`tion of IFN-o. with oral ribavirin, are expensive, often poorly
`tolerated, and effective only in half of the patient population}3
`Therefore, there is an urgent need to develop new and more
`effective therapies in response to this important unmet medical
`need.4
`
`In the course of our HCV program, we recently discovered
`that 2’-C—methylcytidine (1, Figure l) is a potent and selective
`inhibitor of Flauiuiridae virus replication in cell culture.5-6 More
`particularly, 1 inhibited the replication of the bovine viral
`diarrhea virus (BVDV, a pestivirus surrogate model for HCV),7
`eliminated persistent BVDV infection at nontoxic concentra-
`tions, and was synergistic in combination with interferon—omJ
`but not with ribavirin.8 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
`viral RNA levels 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 nucleosidc
`analogue mode of action, Thus, in primary human hepatocyte
`cultures, in a human hepatoma cell line (HepGZ), 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'-tiiphosphate, resulting
`from deamination. The active metabolite 2’~C~methylcytidine
`
`‘ Corresponding author Tel: + 33-4-67143855. Fax: + 33-4—67549610.
`Email: gosselin@univ-montp2ifr.
`fUniversité Montpellicr ll.
`3 Idenix Pharmaceuticals Inc.
`§Laboratoires Idcnix SARL,
`V Present address: VASTox plc, 91 Milton Park, Abingdon, Oxfordshire,
`OX14 4RY, U.K,
`
`H0
`
`NH;
`\N
`l as
`"05
`N
`l" H,i:,v’
`OH
`HO
`2'-C-methyleylidine (1)
`
`0
`
`0
`[-
`
`HO
`
`'
`'NH
`l
`‘N’RO
`O
`l” chf
`OH
`HO
`'-C-mathyluridlno(2)
`
`@e
`NH,
`CI
`‘N
`(:K
`,0“ 54
`H3433
`H
`0
`
`0
`
`HO
`
`L;
`-
`o; 0
`OleGHSNJY'
`3'-O-vallnyl ester of
`2‘-C-methyleytidlna (3)
`(cf/hydrochloride salt)
`
`Figure 1. Structures of compounds 1-3.
`triphosphate is a competitive inhibitor of purified BVDV RNA
`polymerase in vitro (K, = 160 nM).g
`Preliminary pharmacokinetic studies in animals revealed that
`further development of 1 would be hampered by its low oral
`bioavailability. To overcome this limitation, we devoted our
`efforts to the design of 2’-C—rnethylcytidine 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 nucleosidc
`prodrug forms. More particularly, efficacy of such derivatives
`has been proved in the case of valacyclovir, the L—valinyl 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 cellular
`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
`nucleosidcfllu Recently, as part of our hepatitis B program,
`we have synthesized and studied several L—valinyl ester prodrugs
`of 2’—deoxy-,B-L-cytidine (L—dC)
`in order to improve the oral
`bioavailability of L—dC, Among them, the 3’—0—L—valinyl ester
`of L-dC (Val-L—dC, valtorcitabine, Figure 2) emerged as the most
`attractive L<dC prodrug14 and is currently in phase II clinical
`studies.”
`On the basis of these considerations, we decided to synthesize
`and study the 3’—O~L-valinyl ester derivative (NM283, valop—
`
`10.1021/jm0603623 CCC: $33.50 © 2006 American Chemical Society”
`Published on Web 10/06/2006
`‘
`
`CLARK EXHIBIT 2125
`Sommadossi v. Clark
`Contested Case 105,871
`
`1
`
`G|L2018
`I-MAK, INC. V GILEAD PHARMASSET LLC
`|PR2018—00122
`
`1
`
`GIL2018
`I-MAK, INC. V GILEAD PHARMASSET LLC
`IPR2018-00122
`
`

`

`Synthesis and Pharmacokinetics of Valopicitabine
`o
`
`ee
`
`0
`y
`H3N .JL.O
`
`c!
`
`I
`
`l”
`M” NH
`
`2
`
`0
`
`</N
`N
`\/
`VEIZCIMOVII'
`
`Cl
`
`as)
`
`‘
`
`o
`
`N
`/
`0
`<
`l
`_:L_
`HaNl" O‘KOVN N
`
`NH
`/L
`/
`
`””2
`
`(a e
`3
`c:
`NH
`N/J
`N-___.0.
`t.
`
`o
`
`_
`
`OH
`
`.
`
`.
`
`Or
`
`o
`
`.
`
`-[
`one ©H;N""|
`
`Val-L-dc
`
`Valgancicluvlr
`
`‘
`
`Figure 2. Structures of valacyclovir, valganciclovir, and val-L—dC.
`
`in order to improve the oral
`icitabine, 3; Figure l) of 1
`bioavailability of the parent nucleoside.
`Results and Discussion
`
`Synthesis. The 3’—O-valiny1 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—
`Dialkylformamidines and especially N,N-dimethylformamidine
`have been widely used as protective groups for exocyclic amino
`function of nucleosidesfléi17 In the present work, the synthesis
`ofN4—[(dimethylamino)methylene]—2’-C-methylcytidine (4) was
`carried out following a procedure described by Kerr et al for
`the protection of 1 using N,N—dimethylformamide dimethylac—
`eta].18 Selective silylation of the 5’ primary hydroxyl group using
`a reported method19 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),20 with 4-(dimethylamino)pyridine (DMAP) as a cata-
`lyst,“ provided the N-Boc-protected ester derivative 6. Finally,
`desilylation of 6 with excess ammonium fluoride (NH4F) in
`methanol,21 an economical alternative to tetrabutylammonimn
`
`Scheme 1. A Conventional Route for the Synthesis pf 3"
`
`Journal of Medicinal Chemistry, 2006, V01. 49, No. 22 6615
`
`fluoride in tetrahydrofuran (TBAF/TIIF), followed by acidic
`hydrolysis using a saturated solution of hydrogen chloride
`in ethyl acetate,22 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 raceinization of the L—valine during the coupling
`step), as well as cost issues (use of expensive tert-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—tert—butyloxycarbonyl group, thus reducing
`the number of the 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 presence of triethylamine, DMAl’, and DMSO.
`This reaction selectively gave 3’—Boc—guanosine in 55% yield.23
`In our first attempt, we reacted compound 1 in DMSO with
`carbonyldiimidazole(CDI)-activated L-Boc-valine in the presence
`of triethylamine and DMAP. The reaction 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 running die reaction for only 1 h at 80 °C. We used
`only 0.] equiv of DMAP in order to avoid the racemization of
`the L—valine. Other coupling reagents such as EDC and MN-
`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 l,
`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—base extraction, eliminating the extensive chromatographic
`step. Thus, this time- and labor-saving alternative process could
`be accomplished without
`involvement of chromatographic
`
`.
`
`H0
`
`NH2
`.
`N
`,1,
`
`l
`0
`N
`[K'Hfig'
`HO
`OH
`1
`
`0
`
`_',_
`
`Ho
`
`O
`
`7
`(78% for 3 steps)
`(i) MexNCl-I(0Me)2, DMF, 11, 1.5 11; (ii) TBDPSCl, imidazolc, pyridine, it, 6 h; (iii) N—Boc-L-valinc, EDC, DMAP, CH3CN/
`" Reagents and conditions:
`DMF, rt, 2 d; (iv) NHqF, McOH, reflux, 3 11; (v) HCl/EtOAc, EtOAc, rt.
`
`5
`
`NflN’
`J:N
`i
`.. A
`N
`..o..
`" H3?
`HO
`OH
`4
`(60% first crop
`crystallization)
`NH2
`‘N
`' t
`,N
`
`,
`
`N4\N:
`.t
`7]
`N
`II
`t A
`"
`-
`N 0'
`.0.
`-TSi-Ct'
`" Hit?“
`rial
`|
`"H“ HO
`OH
`5
`
`_
`___II _
`
`J iii
`
`0
`
`.
`
`IV
`
`E
`
`MN:
`“N
`'P/KO
`
`o.
`—|~SI-O
`'ng'C
`I”
`'3
`‘5 DVD
`OH
`0* "'f'
`NH
`
`><
`
`HO-
`
`_o__
`
`’ ch
`v
`0 0:.0
`0H
`XOJKNH '[J
`
`(96
`NHa
`c1
`‘N
`(a
`N
`
`0
`
`HO-
`
`.0.
`' Hat:
`i—’
`cc 0
`OH
`CIGGDNH't'l/
`s
`3'-O-valinyl ester of 1
`(hydrochloride form, 3, 81%)
`
`2
`
`

`

`Pierm et a].
`
`Table 1. Pharmacokinctic Parameters of Compounds 1 and 3,
`Following a Single Oral Administration of 3 to Sprague—Dawlcy Rats“
`prong 3
`oompd 1
`dose
`equiv dose
`AUC
`Ti/2
`Cmax
`Tum
`F
`
`(mg/kg)
`(mykg)
`diug
`(ug li/mL)
`(h)
`(fig/mL)
`(h)
`(%)
`100
`72
`3
`8.95
`0.64
`3.62
`1.0
`-
`1
`30.0
`7.10
`6.12
`2.0
`33.6
`
`
`" AUClH: area under the plasma concentration—time curve from time
`0 to r. Tm:
`terminal elimination half-life. Cm“: maximum plasma
`concentration. Tm“:
`time to maximum plasma concentration. F: apparent
`oral bioavailability calculated on the basis of a dose—normalized AUC value
`of 1.24 (fig li/mL) from a single iv dose of] to the rat.
`
`+ 3 in = 3)
`
`«O~
`
`'1 (n '-" 3)
`
`
`
`Ti
`“a.E
`
`E iE
`
`(J
`
`.i
`
`1
`
`0
`
`
`.
`.
`__.
`.
`x
`lo
`M
`.32
`to
`Timeth}
`
`__.
`as
`
`I:
`1
`.9
`::
`g Q3.
`
`g 0,6-
`.
`8 0.4
`2 0,2
`g 0
`a:
`
`D
`
`500 1000 1500 2000
`Incubation tlme (min)
`G @
`NHa
`Cl
`‘N
`6
`’0
`NAG
`H3?
`OH
`
`H
`
`Ho
`
`6616
`
`Journal of Medicinal Chemistry, 2006, Val. 49, No. 22
`
`Scheme 2. Alternative Route for the Synthesis of 3“
`
`NH2
`b.
`..n.. N
`H0:
`
`“0'
`
`O
`
`.
`_I,
`
`NH;
`er
`“0- .u.
`l“
`l" Hp;
`
`O
`
`HO'
`
`ii
`
`696
`NH;
`Cl
`0“.
`0.
`N
`, Hat
`
`O
`
`HO
`
`oH
`
`1
`
`OH
`
`..
`
`00__o
`Jk __
`X0 NH Ii
`7
`(54%)
`
`0.
`
`OH
`
`to
`,_ .-
`GE)
`'l
`Cl
`NH,
`3'-O-va|iny| ester of 1
`(dihydrochloride salt, 3,
`96.5%l
`
`" Reagents and conditions:
`THF; (ii) HCl/EtOH.
`
`(i) N—Boc-L-valine, CDI, DMAP, TEA, DMF/
`
`At H45
`
`Atpl-l 7.2
`
`=
`l
`g
`E
`E 0.9
`
`§ 0.6
`m
`3 0.4
`E 0.2
`%
`0
`m
`
`0
`
`
`
`_.I
`—--i
`5000 10000 15000 20000
`Incubation time (min)
`6) G
`NH3
`CI
`\N
`(x
`N’KO
`'
`0H
`
`5
`H'
`
`_
`
`
`Tm=6.1daysatpH4.5
`Tm: 3.9 hours at pH 7.2
`
`H5.
`0- .0
`eo 'l
`CI
`H,N’ ‘1’
`O = 3‘-0vallnyl ester 0! 2'»C—melhylcytidlne (3)
`1 ,m = 273.5 nm
`
`A = 2'-C-meihylcytidlne (1)
`A w = 273.5 nm
`
`Figure 4. Mean plasma concentration of compounds 1 and 3, following
`a single oral administration of 3 to Sprague-Dawley rats.
`
`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.
`Aqueous Solubility, Lipophilicity, and Chemical Stability
`of Prodrug 3. To be considered as a suitable prodrug for oral
`administration, the 3'—0—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. The
`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:
`#0965 and —1.34 for 1 and 3, respectively.
`The main aim of stability 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 appeared to 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 at all 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 presence of esterases in blood and plasma. Conversion of 3
`into 1 was also observed in human liver cytosol and S9 fractions,
`with approximately 30% of 3 converted into 1 within 1 h.
`Differences in half-life for prodrug 3 in plasma, whole blood,
`
`human liver cytosol, and S9 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.
`The in vitro binding of compounds 1 and 3 to rat plasma proteins
`was investigated by the ultrafiltration method using radiolabeled
`test articles ([3H]-2’-C—methylcytidine and [3H]-3’—0-valinyl
`ester of 2’-C—methylcytidine) 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% bound for 3
`and 1, respectively.
`Oral Bioavailability and Pharmacokinetic Studies of
`Compounds 1 and 3. The pharmacokinetics (PK) of prodt‘ug
`3 were evaluated in Sprague—Dawley rats in a mass balance
`using radiclabeled 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), Lu‘ine, feces,
`and cage rinses were collected for a period of 72—168 h
`postdose to evaluate the excretion of radioactivity and the overall
`mass balance. Plasma samples were collected over a 48-h period
`postdose to study the PK of 1 and 3. 1n intact rats, 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%. Biliary
`excretion of total radioactivity in BDC rats accounted for a mean
`
`3
`
`

`

`Synthesis and Pharmacokinetics of Valopicitabine
`
`Journal of Medicinal Chemistry, 2006, V01. 49, No. 22 6617
`
`of 0.3% of the dose over the 72—h collection period. In this
`group, fecal excretion was the predominant route 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 apparent that at least 30.0% of the [14C]-3’-0-valinyl ester
`of 2’—C-methylcytidiue (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 M03
`at 100 mg/kg (as free base), maximum plasma concentrations
`of total
`radioactivity (averaging 11.9 ng equiv/mL) were
`achieved 1—2 h after the oral dose. Plasma concentrations of
`3, determined by LC/MS/MS, were greatest 0.5~l h after
`dosing, and Cmax averaged 3.62 yg/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 Cmax
`averaged 6.12 rig/mL. Plasma concentrations of1 decayed with
`a half-life of 7.1 h. The AUC of 1 was more than 3—fold greater
`than the AUC of 3. 2’—C—Methyluridine (2) was not detected in
`rat plasma (<0.1 ,ug/rnL).
`The pharmacokinetics of 1 were also studied in Sprague—
`Dawley rats 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 1 following the first
`dose and on day 15 after the last dose. For the scope of this
`paper, only the TK parameters of the 60 mg/kg/day dose group
`on day 1 are presented. Following a single iv dose of compound
`1 to rats at 60 mg/kg, the mean plasma Cmax of l was 75.6
`,ug/mL and the mean AUC was 74.4 pg 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 apparent oral
`bioavailability was calculated for 1. Thus, the apparent oral
`bioavailability of 1 following oral administration of the valinyl
`ester derivative 3 at 100 mg/kg (as free base) was calculated to
`be 33.6%.
`
`Conclusion
`
`3’-0-Valinyl ester of 2’-C—methy1cytidinc (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’-0-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 the basis of its ease of synthesis, its physicochernical
`properties, and phamracokinetic profile, 3’-0-valinyl ester 3 has
`emerged as a promising prodrug of 2’—C—methylcytidine (1). The
`. 3’—0~va1inyl ester of 2’-C—methylcytidine (3, NM283, valopic—
`itabine) is currently undergoing phase Ilb clinical trials for the
`treatment of chronic HCV infection.24
`
`Experimental Section
`General Methods for Chemistry. 1H NMR spectra were
`recorded at ambient temperature on a Bruker AC 200 MHz, 250,
`
`300, or 400 MHz spectrometer. 1H NMR chemical shifts (d) are
`quoted in parts per million (ppm) referenced to the residual solvent
`peak [dimethyl sulfoxide (DMSO-dg] set at 2.49 ppm. The accepted
`abbreviations are as follows: 5, 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 (l/l, v/v, G—T) as the matrix. Melting
`points were determined in open capillary tubes with a Bijchi 13-545
`apparatus and are uncorrected. UV spectra were recorded on an
`Uvikon XS spectrophotometer. Elemental analyses were carried out
`by the Service de Microanalyscs du CNRS, Division de Vernaison
`(France). Thin-layer chromatography (TLC) was performed on
`precoated aluminum sheets of silica gel 60 F254 (Merck, Art. 5554),
`visualization of products being accomplished by UV absorbance
`and by charring with 10% ethanolic sulfirric acid with heating or
`with a 0.2% ethanolic ninhydrin solution for compounds bearing
`an amide function.
`_
`Column chromatography was carried out on 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 P105 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 am,
`C13, 150 x. 3.9 mm). The compound to be analyzed was eluted
`using a linear gradient of 0—50% acetonitrile in 20 mM trierhy—
`lammonium acetate buffer (TEAC, pH 7) programmed over a 30—
`min period with a flow rate of 1 mL/min.
`General Procedure for Aqueous Solubility Studies. Aqueous
`solubilin'es were determined in distilled water at room temperature.”
`An excess of studied compounds was added to aqueous solution,
`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
`saturated solution was determined according to a calibration curve.
`The experiment was repeated three times.
`General Procedure for Distribution Coefficient Studies.
`Distribution coefficients between l-octanol and an aqueous phase
`(phosphate buffer solution 0.02 M) were determined at room
`temperature using a shake-flask procedure.25 An aliquot of a 10‘2
`M aqueous solution of studied compounds was diluted to 1 mL
`with the aqueous phase previously 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 the ratio of the area of the signal detected in the oetanol and
`aqueous phases. Each experiment was repeated twice.
`General’Procedure for Chemical Stability Studies. Chemical
`hydrolysis rates were determined in KCl—HCl buffer 0.135 M
`solution (pH 1.2, 37 °C), acetate buffer 0.02 M solution (pl-l 4.5,
`37 °C), and phosphate buffer 0.02 M solution (pH 7.2, 37 °C).26
`Solutions (10‘4 M) of 3 have been incubated at 37 °C in buffer
`solutions. Aliquot samples were collected at different time intervals
`and analyzed by HPLC (the same conditions as described in General
`Methods for Chemistry). Rates of decomposition have been easily
`dctcrmincd using a method developed in our laboratory and based
`on pseudo—first—order kinetic models.27
`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 [Ag/mL with
`either human plasma or whole blood (Bioreclamation lnc., Hicks»
`ville, NY) preheated to 37 °C. Aliquot samples were collected at
`
`4
`
`

`

`6618
`
`Journal QfMedieinal Chemistry, 2006, V01. 49, Na. 22
`
`different time intervals 0, 2, 5, 10, 20, 40, and 60 min for blood
`studies and 0, 30, 70, 100, and 140 min for plasma studies. Samples
`were quenched in 5 volumes of ice-cold solvent consisting of 50%
`acetonitiile/50% methanol and centrifuged at 16 000 rcf for 3 min.
`Supernatant was removed, dried down in a centrifugal concentrator,
`resuspended in mobile phase (5% MeOH—95% potassium phos-
`phate, monobasic, pH 5.0), filtered (0.2 ,um nylon Spin X tubes),
`and analyzed by reverse-phase HPLC using an Agilent model 1100
`instrument with automatic injection and a diode~array spectropho-
`tometric detector. The mobile phase consisted of buffer A (25 mM
`potassium phosphate, pH 5) and buffer B (methanol). A linear
`gradient from 5% to 100% buffer B was run over 15 min. The
`HPLC system used a Cu Columbus column (5 pm, 250 x 4.6 mm,
`Phenomenex, Torrance, CA). Authentic standards were used to
`identify the peaks in HPLC. This assay was done one time (blood
`and plasma from one donor) in duplicate.
`General Procedure for Metabolic Stability Studies. The ability
`of derivative 3 to function as substrate for relevant metabolic
`enzymes was examined using human liver subcellular fractions
`obtained from BD_ Gentest (Woburn, MA). The prong was
`incubated for 1 h at 50 ug/mL with 1 mg/mL liver S9 or liver
`cytosol. Prior to HPLC analysis, samples were quenched in 5
`volumes of ice-cold solvent (50% acetonitrile/50% methanol) and
`centrifuged (16 000 ref for 3 min). Supernatant was removed, dried
`down in a centrifugal concentrator, resuspended in mobile phase
`(5% MeOH—95% potassium phosphate, monebasic, pH 5.0),
`filtered (0.2 ,um nylon Spin X tubes), and analyzed via HPLC (same
`conditions as described in General Procedure for in vitro Blood
`and Plasma Stability Studies).
`General Procedure for Protein Binding Studies. The protein
`binding of prodrug 3 and parent nucleoside 1 in rat plasma was
`investigated using the ultrafiltration centrifugation method at a
`concentration of 20 [2M for each. The studies used both nomadic—
`labeled and radiolabeled test articles. The [3H]-3’-0-valinyl ester
`of 2’-C—methylcytidine and [3H]-2’-C-methylcytidine, provided by
`Moravek Biochemicals, Inc. (Brea, CA), were prepared by tritium
`exchange (catalyst and labile tritium were removed during purifica—
`tion). To achieve the desired concentrations, concentrated stock
`solutions for each test article containing the appropriate amounts
`of the radiola‘belcd and nonradiolabeled articles were prepared in.
`HPLC grade water. Rat plasma (1 mL) was then added to each
`tube and vortexed. The sample tubes were incubated at 37 0'C for
`1 h.
`Centrifee micropartition units (l-mL capacity, Amicon Inc.) with
`a molecular weight cutoff of 30 000 Da were used to separate the
`unbound from the bound test article by membrane filtration.
`Fortified plasma (0.6 mL) was added to the sample reservoir portion
`of the ultratiltration device. Plasma samples were centrifuge for
`10 min at 37 °C and 1800g so that the amount of sample filtered
`ranged between 20 and 50% of the total volume. After centrifuga-
`tion, the ultrafiltrate of each sample was collected, and 10 pL
`aliquots were analyzed for radioactivity using liquid scintillation
`counting (LSC) on a Beckman Coulter LSéSOO. Protein binding
`determinations were done in duplicate for each test article.
`General Procedure for Pharmacokinetic Studies. The phar-
`macokinetic and mass balance study of the [14C]—3’-O-valinyl ester
`of 2’-C—methylcytidinc (14C label at C-2, Moravek Biochemicals,
`Inc.) was conducted in three groups of male Sprague—Dawley rats.
`A single oral dose of the [14C]-3’-0-valinyl ester of l was
`administered as a solution in 0.01 N HCl at a target dose of 100
`mg/kg (as free base) to the three groups of’male rats afier fasting
`overnight. Group 1 was used to characterize the excretion of the
`radioactivity derived from the [‘4C]—3’—0-valinyl ester of 2’-C—
`methylcytidine in urine and feces. Urine and feces were collected
`up to 168 h postdose from group 1 rats. Group 2 comprised bile
`duct cannulatcd (BDC) male rats and was used to characterize the
`excretion of the radioactivity derived from the [14C]-3’-0-valinyl
`ester of 2'-C-methylcytidine in bile, urine, and feces. Urine, bile,
`and feces were collected up to 72 h postdose in group 2 rats. Group
`3 consisted of jugular vein cannulated (JVC) male rats in which
`the pharmacokinetics of plasma radioactivity derived from the [14C]-
`
`Pierre er al.
`
`3'-0-valinyl ester of .2'-C-methylcytidinc, and 1 and 3 were
`evaluated. Blood samples were collected from group 3 male rats
`predose and 0.25, 0.5, 1, 2, 3, 4, 8, 12, 16, 24, and 48 h postdose
`(3 rats/time point), and the separated plasma was analyzed by LSC
`and by LC/MS/MS. The LC/MS/MS method was validated ac-
`cording to ICH guidelines for bioanalytical method validation prior
`to sample analysis. A daily cage rinse, a cage wash, and a cage
`Wipe at termination were also performed for groups 1 and 2, All
`postdose samples of bile, urine, feces, plasma, and cage residues
`(cage rinse, cage wash, and cage wipe) were analyzed for
`radioactivity by LSC. Mean plasma concentrations of 1 and its 3’-
`0—valinyl ester were used to calculate descriptive phannacokinctic
`parameters by noncompartmental analysis using WinNonlin 4.1
`(Pharsight).
`The 15-day toxicokinetic study of nucleoside analogue 1 was
`conducted to evaluate the pharmacokinetics of 1
`following iv
`administration to Sprague~Dawlcy rats. Compound 1 was admin»
`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
`animals/gender. On days 1 and 15, blood samples were collected
`from the first set of four rats/sex/group at predose and approximately
`20 min and 2 h postdose and from the second set of four rats/sex/
`group at approximately 5 min and 1 and 4 h postdose. Plasma
`samples were analyzed for 2’-C—methylcytidine and 2'-C~methy-
`Iuridine by a validated bioanalytical method (LC/MS/MS), and
`composite PK parameters were calculated by noncompartmental
`analysis using WinNonlin 4.1 (Pharsight).
`For LC/MS/MS analysis, compound 1, its 3’-0-valiny1 ester, and
`added internal standards were extracted from plasma (50 ML) using
`protein precipitation with acetonitrile (0.5 mL). The supernatant
`was dried, reconstituted, and analyzed by LC/MS/MS using HPLC
`coupled to a PE Sciex API3000 tandem mass spectrometer with a
`Turbo IonSpray interface. The HPLC system used an Alltceh
`Platinum C13 column (3 ,um, 53 x 7 mm, Alltceh Associates,
`Deerfield, IL). HPLC elution was carried out using a gradient of
`ammonium acetate (10 mM, pH 4) and acetonitrile. The flow rate
`was 2.5, mL/min with a split of 0.5 mL/min into the mass
`spectrometer. Each, analyte was detected by multiple reaction
`monitoring (MRM) under the positive ion mode using the precursor
`to product ion pair of m/z 357 to 112 (collision energy 27 eV) and
`m/z 258 to 112 (collision energy 20 eV), for 3 and 1, respectively.
`Quantitation was achieved by constructing a calibration curve by
`weighted linear regression of the ratio of the analyte peak area to
`that of the added internal standard. Calibration standards were in
`blank rat plasma. The calibration range was 0.01~5 ,ug/mL in
`plasma for 3 and 0.05~25 ,ug/mL in plasma for the parent
`nucleoside I.
`.
`N“-[(Dimethylamino)methylene]—2’-C-methyI-fi-D-cytidine (4).
`A solution of 15,633 (1.65 g, 6.43 mmol) in NN—dimethylformamide
`(DMF, 32 mL) was treated with dimethylforrnamide dimethylacetal
`(8.2 mL, 61.73 mmol) and stirred for 1.5 h at room temperature.18
`The solution was evaporated under reduced pressure and coevapo-
`rated with ethanol. Crystallization from ethanol/ether yielded the
`hitherto unknown title compound 4 (first crop = 1.21 g, 60%;
`second crop slightly impure on TLC = 0.46 g, 23%) as crystals.
`All
`the following physicochemical characteristics have been
`determined on the

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