throbber
Infectious Disorders - Drug Targets 2006, 6. ”-29
`
`I?
`
`Nucleoside Analog Inhibitors of Hepatitis C Virus Replication
`
`S. S. Carroll and D. B. Oisen*
`
`Department ofAntiviml' Research, Merck Research Laboratories. West Point, PA.
`
`1' 9486
`
`infections, 15 are
`Abstract: 0f the 30 compounds currently marketed in the United States for treatment of viral
`nucleoside analogs. demonstrating the utility of this class of compound as a source of antiviral drugs. The success of
`nucleoside analogs in treating other viral
`infections provides a compelling rationale for the significant effort that is
`currently being devoted to the discovery and development of nucleosidc analogs to treat infection by hepatitis C virus
`{HCV) that may lead to improvements in response rates compared to currently available therapies. Several different
`approaches have been adopted to identify promising analogs, including the use of surrogate viruses in cell culture assays,
`screening in the cell-based bicistronic HCV rcplicon assay. and screening nucleoside triphosphates for the ability to
`inhibit the activity of the HCV RNA-dependent RNA polymerase in virro. Several classes of ribonucleoside analogs with
`modifications of the ribose inhibit HCV replication. Nucleoside analogs incorporating a 2‘-C-mcthyl modification are
`potent inhibitors in the rcplicon assay in the absence of cytotoxicity, and appear to exert their inhibition by acting as
`functional chain terminators of RNA synthesis. NM283, a prodrug of 2’-C-methylcytidinc. has entered clinical trials and
`demonstrated viral load reductions in subjects infected with genotype 1 HCV, a genotype known to be difficult to treat
`effectively with currently approved therapies. Overall, results to date offer encouragement that improved therapies to treat
`HCV infection including newly developed nucleoside analogs may become available within the next few years.
`
`Keywords: Nucleoside analog, replicon, structure-activity relationships, RNA polymerase, chain terminator.
`
`HCV BACKGROUND
`
`HCV1 was recognized as the infectious agent responsible
`for community-acquired non-A non-B hepatitis in 1989 [1].
`This discovery made possible the development of diagnostic
`tests for HCV that have reduced the risk of infection through
`blood transfusion. Yet, estimates of the total number of
`infected individuals are currently 17l0~200 million worldwide
`[2]. HCV infection
`is
`the
`leading
`cause of
`liver
`transplantation in the United States, Currently preferred
`therapies to treat HCV infection consist of six to twelve
`month courses of combinations of pegylated interferon o. and
`ribavirin, which result in sustained viral response (SVR. no
`detectable virus six months afier cessation of therapy) in 40—
`60% of treated patients [3, 4]. The SVR rates with patients
`infected with genotype 1 virus, the predominant genotype in
`most western countries, are lower than with genotype 2
`infections [3, 4]. The low SVR coupled with the frequency
`of
`side
`effects
`associated with
`interferon-ribavirin
`
`the development of
`combination therapies necessitates
`improved therapies to treat HCV infection.
`
`HCV is a positive-stranded RNA virus of the family
`Flaviridae, which also includes human pathogens yellow
`fever, West Nile, and dengue viruses. Replication of the
`HCV genome is catalyzed by a complex of virally-encodcd
`and potentially cellular [5] proteins. At
`the heart of the
`replication complex is
`the HCV RNA—dependent RNA
`polymerase (RdRp) which is
`responsible for catalyzing
`ribonucleotidc incorporation leading to the formation of both
`
`‘Addrcss correspondence to this author at
`Research Laboratories, West Point. PA 19486:
`E-mail: david_olsen@merck.com
`
`the WPZéA-SOUO. Merck
`
`the negative strand copy of the viral genome and subsequent
`positive strand copies that serve as the genomes of progeny
`virus. Since it is absolutely required for viral infectivity [6],
`HCV RdRp is a validated and attractive target
`for the
`development of new treatments for HCV infection based on
`administration of compounds
`that directly inhibit viral
`enzyme function.
`Extensive structural
`
`information is now available for
`
`HCV RdRp including co-crystals with some non-nuclcoside
`inhibitors [7-11]. As with other polymerases such as the
`Klenow fragment of E. coli DNA polymerase I and HIV
`reverse transcriptasc, the overall structure of HCV RdRp has
`been compared to a right hand with fingers. palm and thumb
`subdomains. Unique to the HCV RdRp, though, are extended
`finger domains that come into contact with the thumb
`leading to a completely encircled active site, which includes
`catalytic aspartic acids 318 and 319. The dynamics of the
`contacts between the fingertips and the thumb have been
`implicated in the mechanism of action of some non—
`nucleoside inhibitors [12]. The C-terminal 21 amino acids
`are highly hydrophobic and likely form a membrane anchor
`[13]. Deletion of the C-tcrrninal
`tail gives rise to a more
`soluble enzyme which is utilized in many in vitro studies
`[14].
`
`NUCLEOSIDES AS THE BASIS FOR ANTIVIRAL
`THERAPIES
`
`Nucleoside analogs are successfully employed to treat
`infections with HIV, hepatitis B virus. and herpes viruses, all
`of which encode a polymerase whose primary activity is
`DNA synthesis. All of the nucleoside drugs used to treat
`these infections can be considered deoxynucleosidc analogs.
`
`18? I -5265l06 $50.00+.00
`
`© 2006 Bentham Science Publishers Ltd.
`
`G|L2001
`l-MAK, INC. V GILEAD PHARMASSET LLC
`|PR201 8-00123
`
`1
`
`GIL2001
`I-MAK, INC. V GILEAD PHARMASSET LLC
`IPR2018-00123
`
`

`

`13
`
`Infectious Disorders - Drug Targets 2006, Vol. 6, No. I
`
`Carroll and (listen
`
`HCV RdRp as an RNA polymerase might be expected to
`exhibit different
`structural
`requirements
`for nuclcoside
`analogs that would function as inhibitors, particularly at the
`2’—position of the ribose where inclusion of a hydroxyl
`would likely be advantageous for recognition by RdRp. The
`presence of a 2‘-hydroxyl group on the nucleoside offers a
`chance for greater selectivity of inhibition of the HCV RdRp
`over cellular DNA polymerases. inhibition of the mitochona
`drial DNA polymerase gamma by deoxynucleoside analog
`triphosphates is thought to be the cause of mitochondrial
`toxicity associated with the administration of some nucleo—
`side analogs,
`for example, ddC‘
`[15, 16]. Mitochondrial
`toxicity is likely the basis for the hyperlactatemia,
`lactic
`acidosis,
`steatosis,
`peripheral
`neuropathy, myopathy,
`cytopenias, pancreatitis, and lipoatrophy that are aSSociated
`with long term use of nucleoside inhibitors of HIV reverse
`transeriptase [[7]. However the use of a ribonucleoside
`analog could also open the door to toxicities assoeiated with
`interfering with one or more of the many other roles that
`ribonucleosldes play in viva — in inter- or
`intracellular
`signaling, energy storage and use, protein modification, or
`cellular RNA synthesis.
`
`chain-
`effective
`an
`for
`requirements
`overall
`The
`terminating nucleoside analog inhibitor for oral treatment of
`HCV infection are numerous. First,
`the analog must be
`absorbed efficiently from the gastrointestinal tract. Nucleo-
`side transporters expressed in the intestinal epithelium that
`are responsible for the uptake of most nucleoside analogs
`
`B
`
`In contrast, passive diffusion
`have been described [18].
`appears to be important for acyclovir. Prodrug approaches to
`improving the oral bioavailability of nucleosides have made
`use of amino acid ester modifications,
`for example
`valacyclovir,
`that
`appear
`to
`allow transport
`by the
`oligopeptide transporter pepTl and others [18, l9].
`
`Additionally, the analog must have acceptable pharmaco-
`kinctic (PK) parameters, have a long half-tife sufficient
`preferably for once or twice a day dosing, and achieve
`significant concentrations in the target organ, liver.
`In this
`sense standard plasma PK analysis,
`though useful
`in
`determining the overall exposure to a compound, can
`actually be misleading. A compound that can achieve
`excellent uptake into the liver may appear to have poor
`exposure in plasma in a short-term analysis, since little
`remains in the circulation. In other words, a high first-pass
`uptake into liver is desirable in this case. Balancing the need
`for high concentrations of the nucleoside in liver is the
`general requirement
`that since drug exposure is typically
`monitored in plasma in the clinical setting, some plasma
`exposure is also required. Optimally, the ratio of compound
`concentration in liver to that
`in plasma will be constant
`across time and species, so that the level of compound in
`plasma will be predictive of liver concentrations when the
`compound is dosed in human subjects.
`
`Once it has been absorbed into blood the analog must
`enter hepatocytes,
`either
`through nucleoside or other
`transporters or by passive diffusion, as shown in Fig {I}. The
`
`3215!: Modifications
`
`HO
`
`O
`
`5
`i
`
`MFOHd B
`A HO
`0
`g
`$39-
`9‘?
`v
`si’
`Ql'lo
`
`;
`i
`
`0
`
`B
`
`...,o,
`
`i
`OH
`
`0
`
`;
`:-
`x
`
`o
`
`'
`
`8'
`
`most
`
`.mou
`
`B
`
`'.
`:-
`x
`
`Ho
`
`8
`
`""H
`
`‘HfOH
`4),},
`\460
`4a,,
`Run \ 0'52-
`‘
`a
`\ 01¢
`0“ [OP
`O 0’ \O.
`fa;
`0, o o-
`:P\
`o 0‘
`
`O
`
`Ct ,o
`o
`it
`\‘P.O
`0‘
`Ho’ ‘0.
`
`_
`5'.
`x
`
`Fig. (I). Intracellular metabolism of nucleoside analogs. A ribonucteoside analog with a modified ribose enters a cell via nucleoside
`transponers, passive diffilsion, or peptide tranSporters. 1n the intracellular environment, the nucleoside can either undergo S’-phospho_tylation
`to the monophosphate by the action of' a nucleoside kinase, conversion to a base modified form {8’} via nucleoside metabolizing enzymes
`such as adenosine deaminase. or “base-swapping” via purine nucleoside phosphorylase which reversibly catatyzcs the cleavage of the
`glycosidic bond. The monophosphatc is converted to the diphosphate via nucleotidyt kinase. The diphoSphate may be converted to a
`deoxyribose diphosphate by ribonucleotide reductase, which may then be incorporated into cellular DNA. Altemately the disphosphate may
`be converted to the triphosphate by nucleoside diphosphate kinase.
`
`2
`
`

`

`Nutteoside Arming Inhibitors offleporr‘ris C Virus Replication
`
`Infectious Disorders - Drug Targets 2006, V0}. 6, No. I
`
`19
`
`roles of equilibrative and concentrative transporters in the
`uptake and efflux of nucleoside analogs from cells have been
`recently reviewed [20]. The analog must
`then serve as a
`substrate for three intracellular kinases to be converted to the
`active S‘-triphosphate,
`in general without undergoing
`unwanted metabolism through
`the
`activity of other
`nucleos(t)ide metabolizing enzymes. Modification of an
`adenine base by adenosine deaminase, for example, would
`form the inosine analog, which may not be inhibitory to
`polymerase activity. Cleavage of
`the glycosidic bond
`through the activity of purine nucleoside phosphorylase
`(PNP) can result in scrambling of a modified ribose with
`other nucleobases. In particular, conversion of the diphos—
`phate to a deoxynucleoside diphosphate via ribonucleotide
`reductase is undesirable for nucleobase—modified analogs,
`since that would allow for possible incorporation into
`cellular nuclear DNA which may lead to mutagenesis and
`carcinogenesis. For nucleoside analogs intended to treat
`hepatitis, an important consideration is the expression pattern
`of transporters and the nucleoside metabolizing enzymes
`specific to hepatic tissue which is characterized by high
`catabolic rates [21].
`
`it
`Once the 5‘-triphosphate of the analog is generated,
`must serve specifically as a substrate for the HCV RdRp and
`not as a substrate for cellular RNA polymerases and be
`incorporated into HCV genomic RNA. Incorporation of the
`nucleotide analog must take place against a background of
`competing natural nucleoside triphosphates that are present
`at concentrations that range from hundreds of micromolar for
`CTP, UT? and GTP to millimolar concentrations for
`intracellular ATP [22].
`In contrast
`the concentrations of
`deoxynucleoside triphosphates are generally 10-fold lower.
`Thus, effective inhibition through incorporation of a chain
`terminator may be more difficult
`to
`achieve
`in
`the
`intracellular environment with an RdRp than with a DNA
`polymerase. Once incorporated, the analog should no longer
`support further RNA synthesis by prevcnting the addition of
`more nucleotides.
`In the absence of a mechanism for
`
`excision of the incorporated chain terminator. the truncated
`viral genome would be unable to support further rounds of
`RNA synthesis. Although
`it
`has
`not
`been
`directly
`demonstrated, since HCV RdRp catalyzes the incorporation
`of ribonucleotides with concomitant release of pyrophos~
`phate,
`it must
`also
`catalyze
`the
`reverse
`reaction,
`pyrophosphorolysis. It remains to be seen whether such an
`excision mechanism operates
`in cells and under what
`conditions. These multiple
`requirements
`for
`effective
`nucleoside analog inhibitors present a real challenge to drug
`development.
`
`Despite these challenges, several different nucleoside
`analogs that
`inhibit HCV replication as measured in the
`replicon assay have been discovered. The majority of the
`published infomiation is available for the class of nucleoside
`analogs
`containing a
`2’-C~methyl
`substituent. Recent
`reviews of progress in the discovery and development of
`other classes of inhibitors of HCV replication are available
`[23-26]. The proposed mechanism of ribavirin notwith~
`standing, this review will focus on novel nucleoside analogs
`that inhibit HCV RdRp.
`
`2’-C-METHYL
`OF
`DISCOVERY
`ADENOSINE ANALOGS
`
`MODIFIED
`
`In the past, the inability to propagate HCV robustly in
`cell culture necessitated the use of surrogate assays to screen
`for inhibitors of viral replication. Bovine viral diarrhea virus
`(BVDV) is a pestivirus within the family Flavivirr‘dae, with
`homology to HCV that has been used as a surrogate, since it
`is easily propagated in cultures of MDBK cells, and does not
`infect humans. The use of BVDV antiviral assays for
`investigating inhibitors has primarily been replaced with
`assays based on the HCV replicon [27]. Recently, cell
`culture systems capable of propagating HCV have been
`developed [28-30]
`that will
`likely replace the replicon
`system for routine compound screening.
`
`level of homology between the RNA
`The overall
`polymerases ofHCV and BVDV, even when ignoring an N-
`terminal domain of BVDV RdRp that is not found in the
`HCV enzyme, is quite low. However, within the active sites
`of the polymerases,
`the homology is much stronger,
`suggesting that BVDV might be a useful surrogate for HCV
`for discovery of activevsite directed inhibitors, such as
`nucleoside analogs. Confounding this approach,
`though,
`would be the use of MDBK cells to propagate BVDV which
`might have different capabilities for converting nucleoside
`analogs to the active S’rtriphosphates than human hepatic or
`hepatoma cells [31]. The low overall homology between
`HCV and BVDV RdRp suggests that searches for non-active
`site directed compounds analogous to the non-nucleoside
`reverse
`transcriptase inhibitors of HIV might not be
`successful. This lack of correspondence has been borne out
`experimentally by the
`identification of non—nucleoside
`inhibitors (Nle} that are BVDV- or HCV-specific [10. 32-
`37].
`
`identified as potent
`2’—C—Methyl nucleosides were
`inhibitors of BVDV replication in cell culture by screening
`libraries of nucleoside analogs
`(R. LaFemina, personal
`communication; [38]). Subsequently it was shown that the
`5’-triphosphates of 2’-C-Me~nucleosides can inhibit
`the
`catalytic activity of both BVDV and HCV RdRp in vitro. In
`parallel
`to screening nucleosides in BVDV antiviral cell
`culture assays, chemically synthesized nucleoside triphos-
`phates were screened for inhibition of purified HCV RdRp.
`The in virro screens for HCV RdRp inhibitors aided in the
`understanding of some aspects of the structure-activity
`relationships for inhibition of the target enzyme.
`
`STRUCTURE-ACTIVITY RELATIONSHIPS OF 2’-
`MODIFIED NUCLEOSIDE ANALOGS
`
`Structural modifications to the ribose and nucleobase
`
`for
`have delineated some aspects of the requirements
`efficient inhibition in both the RdRp and cell—based replicori
`assay. In general the results indicate that a very narrow range
`of substituents gives rise to potent inhibition, particularly in
`the replicon assay, owing in large part
`to the multiple
`structural requirements for efficient uptake of the nucleoside
`into the cell, conversion to the 5’-triphosphate, and the
`absence of unwanted metabolic conversion to inactive
`
`that are necessary in order to inhibit viral RNA
`analogs,
`replication in the cellular environment.
`
`3
`
`

`

`20
`
`Infectious Disorders - Drug Targets 2006, V0}. 6. No. I
`
`Modifications to the ribose have investigated the size,
`hydrophobicity,
`electronics,
`and
`regiospecificity
`of
`substituents‘ with the focus on modifications at the 2’ and 3’
`positions, as shown in Fig, (2), Conversion of the TC-
`methyl substituent of the adenosine analog (I) to 2’-C-ethyl
`(2} completely abolishes inhibition by the nucieoside in the
`replicon assay, and by the corresponding 5‘—triphosphate in
`the enzyme assay [39]. Modeling of the ethyl substituted
`analog into the enzyme active site in the position that
`corresponds to the substrate nucleoside triphosphate suggests
`that where
`the
`2’—C-methyl modification
`can
`be
`accommodated into the enzyme active site, the larger ethyl
`modification would clash stericaliy with the side chain of
`Scr282. analogously to the steric clash between the 2’—C-
`methyl substituent with the methyl group of the resistance
`mutation Ser282Thr, as discussed below.
`
`The importance of the stereoelectronic character of the
`2’-C-rnethyl to inhibition was investigated by conversion of
`the methyl group to either CHEF {3) or to CF; (4) [39]. [n the
`case of CHEF, a 16-fold reduction in inhibitory potency in
`the
`replicon assay was observed. whereas
`the CF;
`substitution led to abolishment of cell—based activity. The
`stereospecificity for inhibition was examined by inverting
`
`Carrot! and 0km
`
`the configuration at the 2’-position. The inverted analog (5)
`was not inhibitory either as the triphosphate in the enzyme
`assay or as the nucleoside in the replicon assay. suggesting
`the importance of maintaining the ribo configuration at the
`2’-carbon [39]. The critical
`importance of retaining the 3’-
`hydroxyl to inhibitory potency is demonstrated by the lack of
`activity of the 3‘—deoxy-2’—C-mcthyladenosine analog (6) in
`both the enzyme and replicon assays. Though HCV RdRp
`does not make use of the 3‘-hydroxyl of 2’-C-methyl
`nucleotides as a nucleophile during extension as discussed
`below, the 3’~hydroxyl likely serves as an important binding
`determinant for the initial incorporation event that leads to
`formation of the analog-terminated primer. Moving the
`methyl substituent from the 2‘-carbon to the 3’-carbon {7}
`also leads to complete loss of inhibitory potency [39].
`
`Exploration of the structure-activity relationships of the
`nucleobasc reveals modifications that enhance inhibitory
`potency. 2’-C-Methyicytidine (8) and 2’-C-methylguanosine
`(9) were both active in the rcplicon assay though with
`reduced
`potency
`relative
`to
`2’-C-methyladenosine,
`suggesting that
`the identity of the base is not critical
`to
`inhibition [40]. The reduced inhibitory potency relative to
`the adenosine analog is a consequence of deleterious
`
`H2
`
`/=”
`WW.'
`3
`--..
`-
`EH “\/
`H6
`1.replit:on E050 = 0.25 W
`enzyme IC50 = 1.9 “M
`
`FN
`
`H2
`
`”DA
`
`.-
`Hd
`
`.3
`OH
`
`”mete/l
`
`.4“
`HO
`
`L
`on
`
`2 -
`
`MHz
`
`2, replicon BLT-50> 5!) uM
`enzyme K350 3‘ 5i] iJM
`
`3. replicon ECSO = 4 uM
`enzyme ICSO = 4.5uM
`
`/
`
`HO.“3 N‘x/N
`
`4, replieon E050 > 50 [AM
`
`H0
`.'
`H3
`He
`at.
`7.redbOnEQ,g >50uM
`enzyme [059 > 50 WI
`
`a...
`
`It
`
`_
`
`m .... .
`'5 M
`OH
`
`HO
`
`WHO/3C1;
`
`HO'
`
`‘2
`
`F
`
`H
`
`”2 ”a”
`L CH3
`OH
`
`5. replicon EC60 > 50 em
`erevme I050 > 50 W
`
`6, replioonEcso > 50 pM
`enzyme ICW > 50 14M
`
`0
`
`H2
`
`H
`
`CH3
`a
`50H
`H5
`8. replcon Eng =5 pM
`enzymelCMJ = 0.2 pM
`1
`
`NH2
`
`. can5
`I.
`HO
`”OH
`
`"h.
`
`2
`
`Ho"
`
`5
`
`9.
`
`NH2
`replioon E650 = 2.7ul'vt
`enzyme I050 = 0.15 pM
`
`
`
`10.
`
`replicon E050 = 0.25 an
`enzyme It?“ =0.12 nM
`
`repicon E055 =29 1.1M
`11. R1 = Me
`12. R1 = (3(0le cytotoxic
`13. R1 = CN cytotoxic
`
`14. R2 = Cl cytotoxic
`15. R2 = Br
`cytotoxic
`16. R2 = F
`replicon E050 = 0.07;.LM
`
`Fig. (2). Structure activity relationships for inhibition of ] lCV" RdRp by nucleoside analogs.
`
`4
`
`

`

`Nucleosr'o‘e Analog Inhibitors of Hepatitis C Virus Repficarion
`
`Infectious Disorders - Drug Targets 2906, Vol. 6, No.
`
`i'
`
`2]
`
`metabolism to the inactive uridine analog in the case of 2’—C—
`merhylcytidine, and a reduction in cellular uptake andfor
`efficiency of kinasing in the case of the guanosine analog.
`One of the most useful modifications of the base that has
`been identified is the 7—deaza—purine substitution. The 7-
`deaza modification was
`first
`identified as
`a potencyw
`enhancing substitution within the context of the enzyme
`assay, By comparison of [C59 values for inhibition in the
`enzyme assay for pairs of nucleoside triphosphates with
`either nitrogen or carbon at the 7-position of either adenine
`or guanine-containing analogs and with several different
`ribose modifications, a consistent 10-20-fold increase in
`inhibitory potency was observed for the 7-deaza version,
`demonstrating that the 7—deaza modification is beneficial to
`inhibitory potency independent of the nature of the ribose
`modification [4]},
`in fact, the substrate efficiency of ATP
`compared to that of T—deaza—ATP (tubercidin iriphosphate)
`also demonstrated a 10-fold improvement for the 7-deaza—
`modified
`triphosphate,
`suggesting
`that
`the
`improved
`recognition by the enzyme for the 7-deaza substitution does
`not require any ribose modification (Carroll, 8., unpublished
`observation).
`
`The physical basis for the improved recognition of the 7—
`deaza substitution by HCV RdRp is not clear but could be a
`result of changes in glycosidic bond angle or length. or
`changes in the electronic character of the purine base, or
`possibly suggestive of a direct
`interaction between a
`hydrophobic region of the RdRp active site and the 7-
`position of the purine base. A number of substituents at the
`7-deaza position are tolerated by HCV RdRp, including 7—
`methyl (ll), T-carboxamidc (sangivamycin analog, 12},
`7—
`cyano {toyocamycin analog, 13) with little change in
`inhibitory potency in the enzyme assay. However. in general,
`substitutions at the 7-position give rise to cytotoxicity in the
`cell-based assay and are therefore not useful [40, 42]. A
`trend towards decreasing cytotoxicity was noted for the 7—
`halogen substituted series, ”ii-Br > ”ii-Cl > 7-1: (compounds
`14—16). The i-F-Tdeaza—Z’-C—methyladenosine analog was
`found to be essentially non—cytotoxic by MTS assay in the
`replicon cells at 100 paid, but
`to have the most potent
`repiicon EC50 measured for a nucleoside analog (EC 50 = 0.0?
`pM) [40]. Further investigations of 16 are underway.
`
`The 7—deaza-adenosine modification is beneficial not just
`in terms of target enzyme potency but also in terms of
`stabilization of the analog to unwanted modification by
`adenosine-metabolizing enzymes. Most notably, the 7—deaza—
`modification essentially eliminates the ability of adenosine
`deaminase (ADA) to convert the 2’—C-methyladenosine to
`the corresponding inosine analog which is inactive in the
`either the replicon or enzyme assays. Under conditions
`where 2’—C-methyl adenosine is quantitatively converted to
`2’—C-rnethylinosine in in virro ADA-catalyzed reactions. 2’-
`C—methyl-7—deazaadenosine is left
`intact [40]. The lack of
`conversion of the 7~dcaza—adenosine analog is related to the
`mechanism of catalysis by ADA, wherein the 7—nitrogen is
`protonated to aid in the delocalization of charge that occurs
`during formation of a tetrahedral intermediate that results in
`disptacement of the 67amino group by hydroxyl anion [43].
`
`purine
`the
`phosphorolysis of
`the
`PNP catalyzes
`giycosidic bond, resulting in removal of the modified ribose,
`
`and allowing for subsequent metabolism of the ribose. The
`2'~C-methyl substitution itself reduces the ability of PNP to
`catalyze glycosidic bond breaking [39]. However,
`the F’-
`deaza modification further stabilizes the glycosidic bond to
`PNP-cataiyzed breakdown, further increasing the metabolic
`stabiiity of 7~deazaesubstituted nucleoside analogs
`[40].
`These results demonstrating increased stability to adenosine-
`metabolizing enzymes in vin'o correlate well with increased
`pharmacokinetic stability in vivo as discussed below.
`
`MECHANISM OF CHAIN TERMINATION BY 2'-C-
`METHYL NUCLEOTIDES
`
`The physical mechanism of the inhibition of RdRp
`activity by 2’—C—methyl—modified nucleotides appears to
`involve disruption of the growth of the RNA strand after
`incorporation of
`the modified
`nucleotide. Typically
`nucleoside analogs used as antiviral therapeutics act as chain
`terminators due to the absence of the 3‘-hydroxyl group that
`functions as the nucleophile during extension of the DNA
`chain by incorporation of a nucleotide. Thus. analogs such as
`AZT, 3TC, ddC, d4T and the active forms of didanosine, and
`abacavir are incorporated onto the 3'-end of a growing
`primer strand but are unable to support further elongation of
`the growing strand. The family of 2’-C-methyl modified
`nucieotides appears to act via a similar mechanism despite
`the fact
`that
`the 3’—hydroxyl
`is still present
`in the 2‘—C-
`methyl
`nucleotides. The 2‘-C-methyl nucleotides
`are
`therefore non-obligate chain ten-ninators.
`
`the
`supporting chain termination as
`evidence
`The
`mechanism of inhibition by 2’-C-methy| nucleotides comes
`from analysis of polyacrylamide gel electrophoresis — based
`nucleotide incorporation experiments. Hairpin RNAs formed
`from synthetic oligoribonucleosides have been found to
`serve as efficient templates for RNA synthesis catalyzed by
`HCV RdRp [44]. An example of the designed structure of an
`RNA hairpin template is shown in Fig. (3). which has been
`labeled with ”P at its 5’—end via polynucleotide kinase. The
`RNA template and products resulting from the incorporation
`of nucleotides or nucleotide analogs are then separated via
`gel electrophoresis and visualized via Phosphorlmaging. In
`the case of this RNA template sequence, the first nucleotide
`to be incorporated would be an adenosine or adenosine
`analog, which would be followed by incorporation of a
`uridine or uridine analog, as shown in the control reactions
`on the left side of the gel. HCV RdRp is capable of
`incorporating 3’-deoxy-adenosine monophosphate, but
`is
`incapable of extending the 3‘—deoxy-adenosine monophos-
`phate once it has been incorporated, as expected for this
`obligate chain terminating analog. as shown in lanes 6 and 7.
`HCV RdRp is aiso capable of incorporating 7—deaza—
`adenosine monophosphate (tubercidin monophosphate) into
`this RNA hairpin, and is also capable of extending the
`incorporated T—deaza—adenosinc by addition of the next
`nucleotide, as shown in lanes 8 and 9. However even though
`HCV RdRp can incorporate either 2’-C-methyladencsine or
`2’-C-methyl-‘i—deaza-adenosine into the RNA template, the
`enzyme is incapable of adding the next nucleotide onto the
`end of the analog-tenninated template. Thus the 2’~C—methyl
`nucleoside analogs act as chain terminators.
`
`5
`
`

`

`22
`
`Infectious Disorders - Drug Targets 2006. Vol 6. No. I
`
`Carrot! and Olsen
`
`U
`
`u
`
`00 CCC GGU AGA 5’
`
`U CC GGG CC 3‘
`
`E
`
`++++
`
`ATP (pM)
`UTP (HM)
`3‘dATl’ (HM)
`T-deaz.a~ATP (HM)
`2"-('- lVie-ATP (HM)
`7-dcaza—2'-C Mc-ATP (pM)
`Fig. (3). Gel based incorporationfextension assays. The hairpin RNA oligoribonucleotide with the sequence shown was radiolabeled at the
`5‘~end with 32P-phosphate via polynucleotide kinase. Reactions were catalyzed by purified HCV RdRp in the presence of the indicated
`concentrations of nucleoside triphosphates. Reactions were quenched by addition of loading buffer and reaction products were separated on
`20% polyacrylamide-S M urea sequencing gels. Radiolabeled products were visualized with Phosphorlmaging. The sequence was chosen so
`that the first nucleotide to be incorporated onto the 3‘-end of the hairpin is AMP (third lane of the gel from the left}, followed by
`incorporation of UMP (4"' lane). 3’-deoxy~AMP is incorporated in place of AMP [5‘h lane), but the product of incorporation of 3‘»dAMP
`cannot be extended by addition of UTP. Tubercidin triphosphate serves as a substrate for HCV RdRp, and can be incorporated in place of
`AMP and the product of the incorporation can be extended by addition of UMP (lanes 7 and 8). Both 2’-C-Me-ATP and 2‘-C-Me-7-deaza~
`ATP can be incorporated onto the end of the RNA, but neither is efficiently extended by addition of UTP. A trace of extended product is
`evident with 2’-C~Me—ATP that is absent with the T-deaza analog.
`
`ll) 10
`
`l0 [0
`
`IO 10
`
`2 0
`
`10!
`
`The physical basis for the chain termination mechanism
`is unknown but a potential explanation is illustrated in the
`model shown in Fig. (4). This model was constructed using
`the initiation complex from a co-crystal of the bacteriophage
`phi-6 RdRp [45], consisting of two nucleoside triphosphates
`hydrogen bonded to the 3’-end of a single stranded RNA
`template bound to the phi-6 RdRp. One of the nucleoside
`triphosPhates corresponds to the nucleophile or primer 3’~
`terminal nucleotide {the one on the left in Figure (4)), and
`the
`other
`corresponds
`to
`a
`“substrate"
`nucleoside
`triphosphate, the pyrophosphate of which is displaced during
`catalysis. The initiation complex was copied into the HCV
`RdRp active site [8]. For
`illustrative purposes both the
`nucleophilic nucleoside triphosphate and the
`substrate
`nucleoside triphosphate are shown as 2’-C-rnethyl-adenosine
`triphosphatcs in the Figure. In the case of chain termination
`the RdRp is capable of incorporating the 2‘-C-n1ethyl
`nucleotide analog. We suppose that the enzyme can then
`translocate forward on the template strand such that
`the
`nucleotide
`analog now occupies
`the primer-terminus
`position, corresponding to the nucleophile during catalysis.
`When the substrate nucleoside triphosphate then binds (as
`shown in Fig.
`(4)
`the substrate is another 2‘-C-methyl
`adenosine triphosphate, but
`it could be any nucleoside
`triphosphatc)
`the 2’-C-mcthyl group of the incorporated
`analog is now in proximity to both the ribose ring oxygen,
`and the 8-position of the nucleobase of the incoming
`
`nucleoside triphosphatc, as represented by the yellow and
`red crosshatching. Thus the methyl group may provide a
`stel‘ic clash that either prevents binding or prevents optima]
`alignment of the substrate nucleoside triphosphate to inhibit
`efficient nucleophilic attack of the 3’-hydroxyl on the alpha-
`phosphorous, thus preventing chain extension of the 2‘-C-
`methyl analog-terminated primer.
`
`RESISTANCE T0 INHIBITION BY 2’-C-METHYL
`NUCLEOSIDES
`
`One of the most convincing lines of evidence to establish
`the intracellular target of inhibition by a given antiviral
`compound is the generation of resistance to inhibition in a
`cell culture setting. Mapping the mutation that creates
`resistance to a target gene, and correlating changes in the
`inhibitory potency of the compound in biochemical assays of
`the mutated enzyme demonstrates the validity of the target.
`Additionally,
`in vitro resistance
`studies
`can provide
`infotmation regarding the likelihood of the appearance of
`resiStance
`during
`clinical
`use of
`an
`investigational
`compound,
`as well
`as
`information on
`the potential
`deleterious effects of the mutation on the replicative capacity
`of the mutated virus.
`
`Resistance to 2’-C-methyl nucleosides in the bicistronic
`replicon assay is engendered by a single amino acid change,
`Ser282Thr, within the active site ofthe RdRp [46]. A 38-fold
`
`U i
`
`f}
`10
`
`it)
`
`if]
`ja.
`
`6
`
`

`

`Nrreteoi'ide Analog Inhibitors afHeparr‘riis C Virus Replication
`
`Infectious Disorders - Drug Targets 2006. Vol. 6, No. i'
`
`23
`
`
`
`Fig. (4). A model of the active site of HCV RdRp. The model was constructed using the structure of a de novo initiation complex structure of
`the phi~6 bacteriophage RdRp [45] containing a bound template RNA coordinated to two nucleoside triphosphates. The protein structure was
`replaced with the crystal structure of the HCVr RdRp [8]. In place of the nuclcosidc triphosphatcs were modeled two molecules of 2‘~C~
`methyl—T—dcazaadenosine triphosphate. The crosshatching represents a van der Waals radius of the Z'methyl group of one nucleoside analog
`and the ribosc ring oxygen and 8-position of the other. The blue numbers represent distances in Angstroms from the methyl carbon to the
`ring oxygen and 8-positions of the other nucleoside analog. The purple spheres represent Mg++ ions required for catalysis.
`
`loss of inhibitory potency was determined for 2‘—C—mcthyl
`adenosine in the resistant replicon. The mutation is quite
`specific for
`the 2’-C—methyl modification as shown by
`essentially no change in inhibitory potency for 2’-O-
`methylcytidine. where the methyl group is now present as an
`ether
`linkage. Likewise a 10-fold loss of potency was
`observed for inhibition by 2'-Cwmethyl~guanosine wit

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