throbber
J. Med. Chem. 2006, 49, 7215-7226
`
`7215
`
`Application of Phosphoramidate ProTide Technology Significantly Improves Antiviral Potency
`of Carbocyclic Adenosine Derivatives
`
`Christopher McGuigan,*,† Alshaimaa Hassan-Abdallah,† Sheila Srinivasan,† Yikang Wang,† Adam Siddiqui,† Susan M. Daluge,‡
`Kristjan S. Gudmundsson,‡ Huiqiang Zhou,‡ Ed W. McLean,‡ Jennifer P. Peckham,‡ Thimysta C. Burnette,‡ Harry Marr,‡
`Richard Hazen,‡ Lynn D. Condreay,‡ Lance Johnson,‡ and Jan Balzarini§
`Welsh School of Pharmacy, Cardiff UniVersity, King Edward VII AVenue, Cardiff CF10 3XF, UK, DiVision of Chemistry MV CEDD; Drug
`Metabolism and Pharmacokinetics and Virology Departments, GlaxoSmithKline, Research Triangle Park, North Carolina 27709, and Rega
`Institute for Medical Research, Katholieke UniVersiteit LeuVen, B-3000 LeuVen, Belgium
`ReceiVed June 30, 2006
`
`We report the application of phosphoramidate pronucleotide (ProTide) technology to the antiviral agent
`carbocyclic L-d4A (L-Cd4A). The phenyl methyl alaninyl parent ProTide of L-Cd4A was prepared by
`Grignard-mediated phosphorochloridate reaction and resulted in a compound with significantly improved
`anti-HIV (2600-fold) and HBV activity. We describe modifications of the aryl, ester, and amino acid regions
`of the ProTide and how these changes affect antiviral activity and metabolic stability. Separate and distinct
`SARs were noted for HIV and HBV. Additionally, ProTides were prepared from the D-nucleoside D-Cd4A
`and the dideoxy analogues L-CddA and D-CddA. These compounds showed more modest potency
`improvements over the parent drug. In conclusion, the ProTide approach is highly successful when applied
`to L-Cd4A with potency improvements in vitro as high as 9000-fold against HIV. With a view to preclinical
`candidate selection we carried out metabolic stability studies using cynomolgus monkey liver and intestinal
`S9 fractions.
`
`Introduction
`Nucleoside analogues continue to dominate antiviral therapy
`and also make a significant contribution to the chemotherapy
`of cancer, particularly leukemia. Without exception, nucleoside
`analogues with such activity require phosphorylation in vivo
`to their active nucleotide forms. In the case of antiviral
`nucleosides this is almost always the 5¢-triphosphate. Poor
`phosphorylation can be a major cause of poor activity, with
`several examples now known where nucleoside analogues are
`inactive, despite the corresponding triphosphates being inhibitors
`at their enzyme (polymerase, reverse transcriptase) target.1,2 The
`triphosphates themselves cannot be considered to be useful drugs
`due to their inherent hydrolytic instability and poor membrane
`permeation. However, it appears that in most cases the first
`phosphorylation to the 5¢-monophosphate is the rate-limiting
`step,3 leading to the consideration of the monophosphates as
`chemotherapeutic agents. In fact, nucleoside monophosphates
`suffer from similar qualitative problems as triphosphates;
`instability (in this case to phosphatases and nucleotidases) and
`poor membrane permeation. Given these problems, and the
`perceived advantage of bypassing the nucleoside kinase depen-
`dence of nucleoside analogues, many groups have worked on
`phosphate prodrug (“ProTide”) strategies.4-6 Since 1990, we
`have developed a phosphoramidate strategy; initial work was
`on anti-retroviral AZTa derivatized with alkyl phosphates
`carrying an esterified amino acid.7 Alanine quickly emerged as
`a most effective amino acid. Subsequently, we discovered aryl
`
`* To whom correspondence should be addressed. Tel: +44 29 20874537.
`Fax: +44 29 20874537. E-mail: mcguigan@cardiff.ac.uk.
`† Cardiff University.
`‡ GlaxoSmithKline.
`§ Katholieke Universiteit Leuven.
`a Abbreviations: HIV, human immunodeficiency virus; HBV, hepatitis
`B virus; AZT, 3¢-azido-3¢-deoxythymidine; d4T/d4A, 2 ¢,3¢-dideoxy-2¢,3¢-
`didehydrothymidine/adenosine; ddU/ddA, 2¢,3¢-dideoxyuridine/adenosine;
`3TC, L-3¢ -thia-2¢ ,3¢ -dideoxycytidine; BVDU, E-5-(2-bromovinyl)-2¢ -deox-
`yuridine; L-Cd4A, (1R,cis)-4-(6-amino-9H-purin-9-yl)-2-cyclopentene-1-
`methanol.
`
`phosphate analogues as potent, nucleoside kinase-independent
`antiretrovirals.8,9 Thus, phenyl methyl alanine phosphoramidates
`have emerged as general nucleotide delivery forms, known as
`aryloxy phosphoramidate ProTides. We have applied this motif
`successfully to d4T,10 ddU,11 3TC,12 ddA,13 and d4A.14 In the
`case of d4A, a 100-4000-fold boost in vitro antiviral activity
`was noted on application of phosphoramidate ProTide technol-
`ogy. Other labs have also utilized this methodology, notably
`Franchetti and co-workers15 on isoddA and 8-azaisoddA and
`Zemlicka et al.16 on alkene and related nucleosides. Applying
`our methodology to anti-herpetic BVDU gives unusual results;
`we found a decrease in antiviral action,17 while the NewBiotics
`group reported promising anticancer action for the same
`compounds.18 We have recently reported the enhancement of
`the in vitro profile of these agents by modifications in the
`phosphoramidate structure.19
`A further issue surrounding nucleosides as drugs is the lability
`of the glycoside (base-sugar) bond toward phosphorylase-
`induced cleavage. This frequently leads to inactivation of
`nucleoside drugs. Moreover, as in the case of 5-fluorouracil and
`E-5-(2-bromovinyl)arabinofuranosyluracil, for example, coad-
`ministration can lead to serious toxic events.20 Efforts to address
`this problem have largely led to carbocyclic nucleosides. The
`first of these to enter clinical use is the carbocyclic purine
`analogue abacavir (ABC) (1, Figure 1).21-23 We have recently
`reported the application of phosphoramidate ProTide methods
`to 1 and noted a ca. 50-fold boost in anti-HIV potency and
`correlated this directly with a similar increase in the intracellular
`levels of the bioactive carbovir triphosphate.24 A 10-20-fold
`boost was also noted in antihepatitis B activity for ProTides of
`1.
`
`Given the very high ProTide potentiation noted for adenines
`such as d4A,14 we were interested to examine the effect on
`carbocyclic adenines and particularly Cd4A.
`In fact,
`the
`“natural” D-form D-Cd4A (2) is approximately 3-fold less potent
`than (1) versus HIV.25 The enantiomer, L-Cd4A (3), has modest
`activity versus HBV (ca. 1 (cid:237)M) but is poorly active versus HIV
`
`10.1021/jm060776w CCC: $33.50 © 2006 American Chemical Society
`Published on Web 11/09/2006
`
`IPR2018-00123
`
`Page 1 of 12
`
`I-MAK 1008
`
`

`

`7216 Journal of Medicinal Chemistry, 2006, Vol. 49, No. 24
`
`McGuigan et al.
`
`the effect of ester modification on the antiviral potency of
`phosphoramidate ProTides, with a clear preference for benzyl.31-2
`Indeed, our recent work on BVDU ProTides versus cancer
`indicated a >100-fold improvement of in vitro potency on
`replacement of the methyl ester present in NewBiotics’ lead
`thymectacin18 by a benzyl ester.19 Similarly, we have reported
`extensive SAR studies on the amino acid region, including
`natural amino acid variation,33 un-natural R,R-dialkyls,34 ster-
`eochemical variation,35 amino acid extensions,36 and replace-
`ments.37 In general, alanine and the un-natural amino acid R,R-
`dimethylglycine emerged as the amino acids of choice. Indeed,
`we recently noted that dimethylglycine was a particularly
`efficacious motif with regard to anti-HBV activity when applied
`to abacavir (1).24 Thus, using similar methods (Scheme 2) we
`prepared the glycine (4g), valine (4h), leucine (4i), isoleucine
`(4j), methionine (4k), methyl aspartate (4l), phenylalanine (4m),
`proline (4n), lysine (4o), tyrosine-O-tert-butyl ether (4p), and
`dimethylglycine (4r) analogues, each as the methyl ester. The
`tyrosine compound (4q) was prepared via TFA-mediated
`hydrolysis of (4p), it being notable that the phosphoramidate
`was stable to these conditions. As noted above, we have
`previously found D-alanine to be less effective than L-alanine.35
`However, this has not been extended to other amino acids and
`not on L-nucleosides. Thus, we prepared a small panel of
`D-amino acid analogues: D-alanine (4s), D-phenylalanine (4t),
`D-leucine (4u), D-valine (4v), D-tryptophan (4w), D-methyl
`aspartate (4x), D-proline (4y), and D-methionine (4z).
`As long ago as 1992 we noted the effects on in vitro potency
`of aryl substitution in phosphoramidate ProTides.8,9 We identi-
`fied p-halogen systems as particularly effective, including the
`p-chloro.38,39 Indeed, we subsequently published a rigorous
`QSAR analysis of this effect.40 The group of Uckun have very
`actively pursued the p-bromo derivative on d4T (“stampi-
`dine”).41
`Thus, by the above methodologies, and preparing the aryloxy
`phosphorochloridate from the appropriate phenol where it was
`not commercially available, we prepared methyl alanine ana-
`logues with aryl substitution as follows: p-chloro (4aa), p-nitro
`(4ab), p-CF3 (4ac), m-CF3 (4ad), 3,4-dichloro (4ae), p-CO2Me
`(4af), m-CO2Et (4ag), and o-CO2Et (4ah).
`Finally, for purposes of comparison, the parent phenyl methyl
`alanine derivatives were prepared from enantiomeric D-Cd4A-
`(2) and the corresponding L-CddA (6) and D-CddA(7) (com-
`pounds 5, 8, and 9, respectively).
`Antiviral Activity. All of the phosphoramidates described
`above (4a-ah), 5, 8, and 9 were tested in vitro against HIV-1,
`HIV-2, and HBV, with nucleosides 2, 3, 6, and 7 as controls.
`Cytotoxicity was also evaluated in MT4 and CEM cells. All of
`the data are presented in Tables 1 and 2 (in (cid:237)M). Thus, the
`parent phenyl methylalaninyl ProTide of L-Cd4A (4a) displayed
`a ca. 2700-fold boost in anti-HIV potency, being active at 30
`nM, vs 80 (cid:237)M for the parent. The ProTide was ca. >15 times
`more cytotoxic than the parent but still displays a selectivity
`index (SI ) CC50/EC50) of >200. As expected, no significant
`differences in potency were noted for HIV-2 vs HIV-1 and for
`MT4 vs CEM cells. Versus hepatitis-B virus (HBV), where 3
`is already quite active (EC50 ca. 1 (cid:237)M), 4a is ca. 60-times more
`potent at 17 nM and shows little toxicity (CC50 1280 (cid:237)M; SI
`ca. 75 000).
`As the ester was lengthened from methyl to ethyl (4b), there
`was no significant change in antiviral potency, while the pattern
`was variable for the secondary, ispropyl ester (4c) and tertiary
`tert-butyl ester (4d). In general, the tert-butyl ester was less
`active; this correlates with our previous conclusions29 and has
`
`Figure 1. Structures of some antiviral carbocyclic nucleoside ana-
`logues.
`
`(ca. 80 (cid:237)M).26 We wondered to what extent this difference might
`reflect the relative efficiency of phosphorylation, which might
`be bypassed by ProTide methodologies. This paper describes
`our initial attempts in this regard.
`
`Results and Discussion
`Chemistry. D-Cd4A (2) and L-Cd4A (3) were prepared as
`outlined in Scheme 1 following published procedures. Briefly,
`4-amino-2-cyclopentene-1-methanol (D) was prepared from
`commercially available azabicyclo[2.2.2]hept-5-en-3-one (A) as
`peviously described in the literature.27 Condensation of this
`(D) and 5-amino-4,6-
`4-amino-2-cyclopentene-1-methanol
`dichloropyrimidine (E) in butanol at elevated temperature
`resulted in the formation of 4-[(5-amino-6-chloro-4-pyrimidi-
`nyl)amino]-2-cyclopentene-1-methanol (F).28-29
`The carbocyclic chloropurine (G) was formed by treatment
`of F with triethylorthoformate in the presence of acid. Finally,
`treatment with liquid ammonia in a Parr bomb gave the desired
`carbocyclic L-Cd4A (3). Carbobocyclic L-ddA (6) was synthe-
`sized by reducing the cyclopentene using 5% Pd/C under 40
`psi of hydrogen.
`The D-analogues 2 and 7 were prepared in a similar manner
`as described for the L-analogues.
`We followed the standard phosphorochloridate approach to
`the synthesis of ProTides that we developed in the 1990s.9 This
`involved the preparation of an aryloxy phosphorodichloridate
`by reaction of an appropriate phenol with phosphoryl chloride,
`followed by condensation with an esterfied amino acid hydro-
`chloride to give the key phosphorochloridate reagent. Reaction
`of these phosphorochloridates with nucleosides such as Cd4A
`has two challenges. The first is poor solubility, and the second
`is regiochemistry. It is important to restrict the phosphorylation
`to the 5¢-hydroxyl group and eliminate any base (amino)
`phosphorylation. This was addressed very successfully by
`Uchiyama30 using Grignard reagents of strong bases to generate
`the 5¢-alkoxide, which gives preferential reaction with electro-
`philes. We have noted the efficacy of the Uchiyama method on
`abacavir.24 Thus, we employed the same general method here
`(Scheme 2).
`In the first instance, L-Cd4A (3) was converted into its phenyl
`methylalaninyl phosphoramidate (4a) in 81% yield. As noted
`for almost every nucleoside phosphoramidate ProTide, this was
`isolated as a roughly 1:1 mixture of phosphate diastereomers,
`as evidenced by two closely spaced 31P NMR signals ((cid:228)P 3.8,
`4.1). The isomeric mixture was also evident in the 1H NMR
`(e.g., OMe (cid:228)H 3.70, 3.72) and the 13C NMR (e.g., CH3-Ala, (cid:228)C
`19.8, 20.0). Similarly prepared were the alanine analogues with
`ester modification: ethyl (4b), isopropyl (4c), tert-butyl (4d),
`tert-butyl-CH2 (4e), and benzyl (4f). We have previously noted
`
`IPR2018-00123
`
`Page 2 of 12
`
`I-MAK 1008
`
`

`

`Phosphoramidate ProTide Technology
`
`Journal of Medicinal Chemistry, 2006, Vol. 49, No. 24 7217
`
`Scheme 1. The Synthetic Route to L-Cd4A (3) and L-CddA (6)
`
`Scheme 2. The Synthetic Route to ProTides of Carbocylic
`Nucleoside Analogues 2, 3, 6, and 7a
`
`a For details of the structures, see Table 1.
`
`been ascribed to the relative stability of tertiary esters to enzyme-
`mediated hydrolysis. However, the isopropyl showed a slight
`increase in potency versus HBV and variable results versus HIV
`dependent on the cell line and assay. The extended system with
`a tBuCH2 ester (4e) showed high activity versus both HIV and
`HBV, being ca. 6-12-fold more potent
`than 4a. Finally,
`regarding esters, the benzyl analogue 4f emerged as the most
`potent ester versus HIV, being active at 9 nM and thus ca. 9000
`times more active than 3. It was also rather nontoxic and
`displayed an SI of >28 000. It was also highly active versus
`HBV, with an EC50 of ca. 7 nM, although some cytotoxicity
`was noted in this assay (at 5 (cid:237)M).
`Turning now to amino acid variations, leaving the ester as
`methyl, we have previously noted a 60-70-fold reduction in
`anti-HIV potency for d4T ProTides on alanine to glycine
`replacement29 and a 20-40-fold reduction for the corresponding
`abacavir ProTides.24 In this study with L-Cd4A we again note
`a significant drop in anti-HIV potency on this substitution (4g),
`giving, depending on the cell line, a 10-200-fold reduction.
`However, HBV activity displays a different trend, showing only
`a modest 5-fold drop in potency, thus retaining a log more
`potency than the parent 3. Similarly, the valine compound 4h
`showed a 10-20-fold reduction in anti-HIV potency as com-
`pared to 4a but was equipotent to 4a against HBV, at 20 nM.
`The data for the isoleucine analogue 4j parallel that of the valine
`compound, as might be expected from their similar structure,
`while the leucine analogue (4i), with the amino acid branch
`one bond further out from the asymmetric center,
`is ap-
`
`proximately a log more active in each assay and thus rather
`similar to alanine. The methionine (4k) and methyl aspartate
`(4l) analogues were rather similar to the leucine compound,
`while the phenylalanine analogue 4m was slightly more active,
`particularly versus HBV, where it was the most potent amino
`acid to date at 4.5 nM. The proline compound 4n was the least
`active of the amino acids to date, an observation that we have
`made previously of this rather unique amino acid.29 We report
`in this paper our first successful ProTide example with lysine
`as the amino acid. This was isolated and tested as its TFA salt
`(4o) and found to be rather poorly active; in fact, it is rather
`similar to parent 3 in several assays and 2-10-fold less active
`than the proline analogue 4n. It is interesting to compare the
`methionine (4k) and lysine (4o) cases, as they have side chains
`with similar geometries. The lysine case is ca. 50-100 fold
`less potent versus both HIV and HBV. Partly,
`this may
`correspond with the higher polarity of the lysine compound
`(particularly when protonated) and diminished membrane
`permeability. The calculated ClogP values of 4k and 4o are
`0.9 and 0.46 (Chemdraw Ultra 9.0), but these figures may not
`fully reflect the likely protonation of the lysine side chain at
`physiological pH, further diminishing its lipophilicity.
`The tyrosine analogue 4q was prepared via its protected tert-
`butyl ether (4p), so we evaluated both the free and protected
`versions in vitro. In fact, they were both rather similar in
`antiviral profile and similar to valine. The boost in anti-HBV
`activity seen for the Phe analogue (4m) was not seen for the
`Tyr compounds.
`The dimethylglycine compound (4r) was not available for
`evaluation versus the whole panel of assays, but initial data
`indicate a slight reduction in potency vs HIV and slight increase
`in potency against HBV. Thus, 4r emerged as the most potent
`member of this series against HBV with activity at 2.5 nM,
`thus being almost 400 times more potent
`than the parent
`nucleoside.
`We have previously found D-alanine to be significantly less
`effective than natural L-alanine in phosphoramidate ProTides
`of d4T.35 We recently noted the same trend for activity of
`abacavir ProTides against HIV.24 In the present case the data
`are clear and marked; the D-alanine system (4s) is ca. 100-fold
`less potent than the L-alanine parent (4a) against HIV, but D
`and L are equipotent against HBV. Indeed, comparison of the
`D-alanine (4s) and dimethylglycine (4r) systems is instructive.
`
`IPR2018-00123
`
`Page 3 of 12
`
`I-MAK 1008
`
`

`

`7218 Journal of Medicinal Chemistry, 2006, Vol. 49, No. 24
`
`McGuigan et al.
`
`Table 1. Anti-HIV Activity and Cytotoxicity Data for Nucleoside and Nucleotide Analogues
`MT4/(cid:237)M Rega
`HIV-1
`HIV-2
`
`Ar
`
`ester
`
`AA
`
`Ph
`Ph
`Ph
`Ph
`Ph
`Ph
`Ph
`Ph
`Ph
`Ph
`Ph
`Ph
`Ph
`Ph
`Ph
`Ph
`Ph
`Ph
`Ph
`Ph
`Ph
`Ph
`Ph
`Ph
`Ph
`Ph
`p-ClPh
`p-NO2Ph
`p-CF3Ph
`m-CF3Ph
`m,p-Cl2Ph
`p-CO2MePh
`m-CO2EtPh
`o-CO2EtPh
`-
`-
`Ph
`-
`Ph
`-
`Ph
`
`Me
`Et
`iPr
`tBu
`tBuCH2
`Bn
`Me
`Me
`Me
`Me
`Me
`Me
`Me
`Me
`Me
`Me
`Me
`Me
`Me
`Me
`Me
`Me
`Me
`Me
`Me
`Me
`Me
`Me
`Me
`Me
`Me
`Me
`Me
`Me
`-
`-
`Me
`-
`Me
`-
`Me
`
`Ala
`Ala
`Ala
`Ala
`Ala
`Ala
`Gly
`Val
`Leu
`Ile
`Met
`MeAsp
`Phe
`Pro
`Lys(TFA)
`Tyr(OtBu)
`Tyr
`Me2Gly
`D-Ala
`D-Phe
`D-Leu
`D-Val
`D-Trp
`D-Asp(OMe)
`D-Pro
`D-Met
`Ala
`Ala
`Ala
`Ala
`Ala
`Ala
`Ala
`Ala
`-
`-
`Ala
`-
`Ala
`-
`Ala
`
`0.045
`ND
`0.15
`8.19
`0.043
`ND
`4.35
`1.09
`0.19
`0.96
`0.16
`0.32
`ND
`5.28
`15.2
`0.11
`0.58
`ND
`ND
`13.3
`1.24
`6.93
`13.3
`3.9
`117
`4.07
`0.009
`0.12
`0.035
`0.044
`0.12
`0.14
`0.053
`ND
`ND
`ND
`ND
`ND
`ND
`ND
`ND
`
`0.043
`ND
`0.22
`3.08
`0.03
`ND
`5.73
`1.05
`0.13
`1.13
`0.21
`0.26
`ND
`21
`27.2
`0.13
`0.72
`ND
`ND
`5.93
`0.93
`14.2
`21.9
`3.71
`105
`3.48
`0.018
`0.17
`0.046
`0.04
`0.076
`0.056
`0.033
`ND
`ND
`ND
`ND
`ND
`ND
`ND
`ND
`
`CEM/(cid:237)M Rega
`HIV-1
`HIV-2
`
`0.13
`ND
`0.3
`2.93
`0.067
`ND
`13.5
`1
`0.42
`2.1
`0.53
`1.1
`ND
`3.5
`25
`3
`5.33
`ND
`ND
`9
`1.6
`7.5
`20
`5
`110
`7.67
`0.015
`0.1
`0.06
`0.15
`0.09
`0.13
`0.065
`0.053
`ND
`ND
`ND
`ND
`ND
`ND
`ND
`
`0.09
`ND
`0.25
`3.67
`0.083
`ND
`20
`4.5
`0.56
`3.5
`1
`3.5
`ND
`7.5
`150
`5.5
`7
`ND
`ND
`8.67
`3.5
`10
`25
`15
`105
`7.87
`0.047
`0.1
`0.06
`0.1
`0.11
`0.13
`0.08
`0.057
`ND
`ND
`ND
`ND
`ND
`ND
`ND
`
`MT4/(cid:237)M GSK
`HIV-1
`CC50
`6.4
`>25
`100
`<25
`>10
`255
`.100
`<25
`>5
`>5
`>5
`>16
`16
`.10
`.25
`<40
`>32
`20
`>100
`>40
`<16
`>16
`>40
`.10
`ND
`>25
`2
`10
`ND
`4
`>4
`>4
`-
`<4
`>100
`>500
`3.7
`>10
`>12.5
`>125
`6.5
`
`0.03
`0.017
`0.425
`<0.26
`>4
`0.009
`7.3
`0.64
`0.64
`1.3
`1.4
`>1
`0.1
`10
`25
`,1
`0.95
`0.14
`2.05
`2.8
`<1
`2.3
`.16
`2.1
`ND
`2.5
`0.013
`0.18
`ND
`0.06
`0.077
`0.12
`-
`<0.26
`80
`15
`0.3
`>10
`1.8
`50
`0.20
`
`CC50
`18.1
`ND
`84
`130
`19.5
`ND
`g250
`167
`70
`118
`85.8
`>50
`ND
`92.1
`>250
`79.7
`110
`ND
`ND
`108
`88.5
`230
`93.4
`g250
`g250
`211
`3.11
`17.4
`6.4
`16.3
`13
`12.9
`8.39
`4.17
`ND
`ND
`ND
`ND
`ND
`ND
`ND
`
`CC50
`16.4
`ND
`91.4
`84.9
`20.2
`ND
`>250
`152
`23.2
`99.6
`34.7
`53.5
`ND
`106
`>250
`19.8
`26.9
`ND
`ND
`71
`77.1
`101
`66.9
`g250
`227
`133
`3.43
`21.6
`5.87
`22.4
`15.6
`11.5
`4.39
`ND
`ND
`ND
`ND
`ND
`ND
`ND
`ND
`
`compd
`4a
`4b
`4c
`4d
`4e
`4f
`4g
`4h
`4i
`4j
`4k
`4l
`4m
`4n
`4o
`4p
`4q
`4r
`4s
`4t
`4u
`4v
`4w
`4x
`4y
`4z
`4aa
`4ab
`4ac
`4ad
`4ae
`4af
`4ag
`4ah
`3
`2
`5
`6
`8
`7
`9
`
`The D-alanine analogue is ca. 10-fold less potent versus both
`HIV and HBV. However, it is also less cytotoxic (>5 to>20
`fold), leaving its selectivity index similar or slightly better.
`Comparing the potency of the alanine (4a), D-alanine (4s),
`and dimethylglycine (4r) systems indicates that substitution on
`the “D-face” of the amino acid is beneficial for HBV and
`detrimental for HIV. This may reflect cell to cell differences in
`processing of the different phosphoramidates. Similar L to D
`trends were observed for D-Phe (4t), D-Leu (4u), D-Val (4v),
`D-Trp (4w), D-MeOAsp (4x), D-Pro (4y), and D-Met (4z), giving
`reductions in anti-HIV potency for D-systems of 5-50-fold and
`retention or only slight reduction for HBV. Exceptions were
`the Val (4v) and Met (4z) cases, which did show significant
`reductions in anti-HBV potency. Thus, versus HIV, in conclu-
`sion alanine remained the most effective amino acid, although
`dimethylglycine was of similar potency, as were Met and Leu
`in some assays, and Val, Ile, and Tyr were also reasonably
`effective. Glycine, proline, and lysine were poorly effective, as
`were the D-amino acids in most cases. The amino acid could
`be varied considerably with little reduction in potency against
`HBV, and several amino acids were either equipotent or more
`potent than alanine, notably dimethylglycine and D-alanine but
`also phenylalanine.
`Finally, on the SAR of the L-Cd4A ProTides, we probed
`several phenyl modifications. In 1995, we had highlighted
`p-halogen substitution as a key area where activity could be
`
`boosted in d4T phosphoramidates39 and this has later been
`picked up by the group of Uckun with their development of
`stampidine, the p-bromo species.41 Thus, in the present case
`we first prepared the p-chloro analogue, which we favor over
`the p-bromo for toxicological reasons, given the mole for mole
`release of p-halophenol on ProTide activation. Indeed, the
`p-chlorophenyl methyl alanine compound (4aa) emerged as the
`most potent methyl alanine to date with a ca. 3-10-fold potency
`improvement against HIV as compared to 4a, with some cell
`to cell variation, and a 5-fold greater potency against HBV than
`4a. We have previously noted the poor anti-HIV efficacy of
`ProTides with phenyl groups containing strongly electron
`withdrawing substitution,40 and to some extent we found the
`same to be the case here, with the p-nitro analogue (4ab) being
`10-20 fold less active than the p-chloro lead (4aa). However,
`it was not significantly less active than the unsubstituted phenyl
`parent (4a) versus HIV. Moreover, 4ab was rather potent vs
`HBV, being equipotent with 4aa and thus slightly more active
`than 4a. This is in contrast to previous experience with d4T
`ProTides and HIV.40 The CF3 group is not as electron
`withdrawing as nitro and is more lipophilic; we previously noted
`it to be more effective in the case of d4T ProTides vs HIV,40
`and we noted the same trend here. Thus, 4ac was more active
`than 4ab, and in general than 4a also, versus HIV and HBV.
`Thus, 4ac is active versus HBV below 3 nM and is the most
`active of the methyl alanine ProTides herein reported. However,
`
`IPR2018-00123
`
`Page 4 of 12
`
`I-MAK 1008
`
`

`

`Phosphoramidate ProTide Technology
`
`Journal of Medicinal Chemistry, 2006, Vol. 49, No. 24 7219
`
`Table 2. Anti-HBV Activity, Cytotoxicity, and Stability Data for
`Nucleoside and Nucleotide Analogues
`
`HepG2-2.2.15
`GSK
`
`HBV
`
`0.017
`0.02
`0.006
`0.25
`0.003
`0.0075
`0.086
`0.02
`0.01
`0.06
`0.03
`0.05
`0.0045
`0.51
`2.0
`0.026
`0.05
`0.0025
`0.0265
`0.7
`0.13
`0.6
`0.2
`0.02
`>2
`0.22
`0.003
`0.007
`<0.0032
`<2
`0.005
`0.004
`0.004
`0.004
`0.98
`52
`4
`61
`0.60
`33
`5.0
`
`CC50
`1280
`22
`>2
`170
`13
`5
`>2
`120
`20
`76
`16
`>2
`9
`>2
`.100
`>2
`>2
`10
`>200
`200
`>2
`>200
`>2
`>2
`>2
`>2
`>2
`>2
`0.72
`>2
`>2
`>2
`>2
`12
`>200
`>200
`5.4
`>200
`>2
`>200
`38
`
`compd
`4a
`4b
`4c
`4d
`4e
`4f
`4g
`4h
`4i
`4j
`4k
`4l
`4m
`4n
`4o
`4p
`4q
`4r
`4s
`4t
`4u
`4v
`4w
`4x
`4y
`4z
`4aa
`4ab
`4ac
`4ad
`4ae
`4af
`4ag
`4ah
`3
`2
`5
`6
`8
`7
`9
`
`int
`
`74
`39
`90
`96
`57
`0
`85
`91
`75
`69
`66
`8
`-
`-
`17
`-
`0
`70
`94
`-
`-
`-
`-
`-
`-
`-
`-
`-
`-
`92
`-
`48
`42
`80
`-
`-
`-
`-
`-
`-
`-
`
`% S9
`remaining
`
`liver
`
`24
`15
`8
`1
`0
`0
`46
`2
`0
`0
`0
`4
`-
`-
`95
`-
`0
`16
`41
`-
`-
`-
`-
`-
`-
`-
`
`45
`-
`7
`19
`25
`-
`-
`-
`-
`-
`-
`-
`
`it is also notably toxic in the HBV assay, being the only ProTide
`in the family that is toxic at submicromolar concentrations.
`Interestingly, the meta analogue 4ad is rather less cytotoxic but
`also apparently slightly less active. Several other aryl substituted
`compounds (4ae-4ah) are also noted in Table 2. The meta-
`substituted ester is the most active against HIV, while several
`compounds are highly active vs HBV. As noted above, the anti-
`HBV activity appears less sensitive to aryl substitution than the
`anti-HIV activity.
`The D-enantiomer of 3, D-Cd4A (2), is slightly more active
`than 3 against HIV and rather poorly active vs HBV. It was
`interesting to see whether ProTides would have a similar impact
`here and we report in Table 1 data on the parent phenyl methyl
`alanine parent (5). Thus, a 50-fold boost in anti-HIV potency
`is noted. This is much less of an improvement (ca. 2600-fold)
`than noted above for the analogous compounds in the L-series
`(3 and 4a), and thus the D-ProTide (5) is about a log less potent
`than the L-ProTide analogue (4a). Similarly, versus HBV, the
`D-ProTide (5) is only 13-fold more potent than the nucleoside,
`whereas in the L-family the boost was 60-fold. More dramatic,
`the D-ProTide (5) is cytotoxic at its effective concentration, with
`a SI barely above unity, while the L-compound (4a) has an anti-
`HBV SI of >75 000.
`Finally, we briefly pursued the application of the technology
`to the dideoxy analogues L-CddA (6) and D-CddA (7) with the
`
`preparation of the ProTides 8 and 9. Thus, unlike several other
`dideoxynucleosides, the carbocyclic analogues 6 and 7 are both
`rather poorly active. Application of ProTides did give interesting
`boosts in potency in both cases. Indeed, while the L-system (6)
`was primarily enhanced (100-fold) versus HBV, the D-com-
`pound (7) was only slightly enhanced vs HBV but significantly
`so versus HIV (250-fold).
`In conclusion, ProTide methods have been shown to signifi-
`cantly enhance the antiviral profile of a series of carbocyclic
`nucleosides, primarily L-Cd4A but also its D-analogue, and the
`dd analogues in both L- and D-series. Very significant differences
`were noted for HIV and HBV, with separate leads emerging
`for each virus, with quite separate and distinct SARs noted for
`each. Effects were noted for variations in the ester, amino acid,
`and aryl regions. In general, the HBV system was more tolerant
`of structural modifications.
`Several nanomolar compounds emerged, representing an
`almost 4-log improvement in potency of several ProTides
`versus the parent L-Cd4A nucleoside. With this background
`we were keen to seek to perform some preclinical analysis
`of the most promising compounds that would allow rational
`choices regarding further evaluation. In particular, the stability
`of the ProTides to metabolic deactivation prior to reaching their
`target site was of particular interest, given our experience with
`abacavir ProTides.24 Thus, we employed a cynomolgus monkey
`liver and intestinal S9 stability assay to gain an initial
`understanding of the stability issues in this family and to probe
`any stability-structure correlations. Data are reported for
`selected compounds in Table 2. Under the conditions of the
`assay 4a underwent some decomposition (ca. 25% over 1 h) in
`the intestinal fraction, but the majority of ProTide remained.
`The position was reversed in the liver fraction, where only 30%
`remained. The most striking of the ester variations is the benzyl
`(4f), which showed complete disappearance in both assays over
`1 h. Thus, although 4f was rather potent in the in vitro antiviral
`assays, the S9 data may be predictive of a limited in vivo
`exposure. Interestingly, the tyrosine compound (4q) revealed a
`similar instability.
`In terms of ester variation; the ethyl ester (4b) was less stable
`than the methyl parent (4a) in both media, whereas branching
`to isopropyl (4c) and tBu (4d) stabilized it in intestine but
`destabilized it in liver. This would suggest that such esters may
`be beneficial to consider for delivery to the liver, in hepatitis
`or liver cancer.
`Several amino acid substitutions for alanine gave a similar
`profile of increased or maintained intestinal stability but
`diminished liver stability, e.g., Val (4h), Leu (4i), Ile (4j), and
`dimethylglycine (4r). The only compounds with enhanced liver
`stability over the parent 4a were the glycine (4g), Lys (4o) and
`D-alanine (4s) amino acid variants and the m-CF3 aryl substitu-
`tion (4ad). Interestingly, the lysine stabilization in liver was
`uniquely accompanied by a significant labilization in intestine.
`In terms of overall maximal stability, a property which might
`be anticipated to be useful with regard to systemic drug delivery,
`the D-alanine and glycine compounds emerge as the most
`notable. Both retained good antiviral potency in the HBV assays
`but were only moderate in the HIV assay. The m-CF3 compound
`(4ad) also looked rather stable in both S9 assays and showed
`good anti-HIV potency. This indicates the potential merit of
`extensive aryl modification to tune stability and improve
`pharmacokinetic properties; however, the relevance of the S9
`stability data to the disposition of these ProTides in monkey
`and/or across species has not been demonstrated.
`
`IPR2018-00123
`
`Page 5 of 12
`
`I-MAK 1008
`
`

`

`7220 Journal of Medicinal Chemistry, 2006, Vol. 49, No. 24
`
`McGuigan et al.
`
`Conclusion
`
`A range of phosphoramidate ProTides of L-Cd4A were
`prepared and evaluated against HIV and HBV in vitro. Boosts
`in anti-HIV potency as high as 9000-fold were noted, with ca.
`250-fold enhancements for anti-HBV activity. Differing SARs
`emerged for each virus. The parent ProTide for enantiomeric
`D-Cd4A showed only modest potency boosts (10-50 fold) with
`parallel increases in cytotoxicity. Similar conclusions were
`reached with regard to the dideoxy analogues L-CddA and
`D-CddA. In conclusion, the ProTide approach is highly suc-
`cessful when applied to L-Cd4A with potency boosts in vitro as
`high as 9000-fold vs HIV. With a view to preclinical candidate
`selection, we carried out metabolic stability studies using
`cynomolgus monkey liver and intestinal S9 fractions;
`this
`revealed interesting differences in stability between the two
`enzyme systems, suggestive of a complex metabolic SAR.
`
`Experimental Section
`
`Chemistry. General Procedures. All experiments involving
`water-sensitive compounds were conducted under scrupulously dry
`conditions. Triethylamine was dried by refluxing over calcium
`hydride. Anhydrous tetrahydrofuran and dichloromethane were
`purchased from Aldrich. Nucleosides were dried by storage at
`elevated temperature over P2O5 in vacuo. Proton, carbon, and
`phosphorus nuclear magnetic resonance (1H, 13C, 31P NMR) spectra
`were recorded on a Bruker Avance DPX spectrometer operating at
`300, 75.5, and 121.5 MHz, respectively. All 13C and 31P spectra
`were recorded proton-decoupled. All NMR spectra were recorded
`in CDCl3 at room temperature (20 ( 3 (cid:176)C). Chemical shifts for 1H
`and 13C spectra are quoted in parts per million downfield from
`tetramethylsilane. Coupling constants are referred to as J values.
`Signal splitting patterns are described as singlet (s), doublet (d),
`triplet (t), quartet (q), or multiplet (m). Chemical shifts for 31P
`spectra are quoted in parts per million relative to an external
`phosphoric acid standard. Many proton and carbon NMR signals
`were split due to the presence of (phosphate) diastereoisomers in
`the samples. The mode of ionization for mass spectroscopy was
`fast atom bombardment (FAB) using MNOBA as matrix. Column
`chromatography refers to flash column chromatography carried out
`using Merck silica gel 60 (40-60 (cid:237)M) as stationary phase. HPLC
`(Shimadzu) was conducted on an SSODS2 reverse phase column
`using a water (containing 0.1% TFA)/acetonitrile (Fisher: HPLC
`grade) eluent. Method 1: 0% CH3CN (0 min), 80% CH3CN (35
`min), 80% CH3CN (45 min), 0% CH3CN (55 min); flow rate, 1
`mL/min; UV detection at 254 nm. Method 2: 0% CH3CN (0 min),
`80% CH3CN (15 min), 80% CH3CN (25 min), 0% CH3CN (35
`min); flow rate, 1 mL/min; UV detection at 254 nm. Final products
`showed purities exceeding 99% with undetectable levels (<0.02)
`of parent nucleosides in every case. UV absorptions were deter-
`mined using a Kontron Uvikon 860 UV spectrometer.
`Standard Procedure: Preparation of Amino Acid Ester Salts.
`Thionyl chloride (2.0 mol equiv) was added dropwise to a stirred
`solution of the appropriate alcohol (15.0 mol equiv) at 0 (cid:176)C under
`nitrogen. The mixture was stirred at 0 (cid:176) C for 30 min and then slowly
`allowed to warm to room temperature. The appropriate amino acid
`(1.0 mol equiv) was added and the mixture was heated at reflux
`

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