Uracyl spirooxetane nucleosides

Information

  • Patent Grant
  • 10301347
  • Patent Number
    10,301,347
  • Date Filed
    Thursday, December 6, 2018
    5 years ago
  • Date Issued
    Tuesday, May 28, 2019
    5 years ago
Abstract
The present invention relates to compounds of the formula I:
Description
BACKGROUND OF THE INVENTION

This invention relates to spirooxetane nucleosides and nucleotides that are inhibitors of the hepatitis C virus (HCV).


HCV is a single stranded, positive-sense RNA virus belonging to the Flaviviridae family of viruses in the hepacivirus genus. The NS5B region of the RNA polygene encodes a RNA dependent RNA polymerase (RdRp), which is essential to viral replication. Following the initial acute infection, a majority of infected individuals develop chronic hepatitis because HCV replicates preferentially in hepatocytes but is not directly cytopathic. In particular, the lack of a vigorous T-lymphocyte response and the high propensity of the virus to mutate appear to promote a high rate of chronic infection. Chronic hepatitis can progress to liver fibrosis, leading to cirrhosis, end-stage liver disease, and HCC (hepatocellular carcinoma), making it the leading cause of liver transplantations. There are six major HCV genotypes and more than 50 subtypes, which are differently distributed geographically. HCV genotype 1 is the predominant genotype in Europe and in the US. The extensive genetic heterogeneity of HCV has important diagnostic and clinical implications, perhaps explaining difficulties in vaccine development and the lack of response to current therapy.


Transmission of HCV can occur through contact with contaminated blood or blood products, for example following blood transfusion or intravenous drug use. The introduction of diagnostic tests used in blood screening has led to a downward trend in post-transfusion HCV incidence. However, given the slow progression to the end-stage liver disease, the existing infections will continue to present a serious medical and economic burden for decades.


Current HCV therapy is based on (pegylated) interferon-alpha (IFN-α) in combination with ribavirin. This combination therapy yields a sustained virologic response in more than 40% of patients infected by genotype 1 HCV and about 80% of those infected by genotypes 2 and 3. Beside the limited efficacy against HCV genotype 1, this combination therapy has significant side effects and is poorly tolerated in many patients. Major side effects include influenza-like symptoms, hematologic abnormalities, and neuropsychiatric symptoms. Hence there is a need for more effective, convenient and better-tolerated treatments.


Recently, therapy possibilities have extended towards the combination of a HCV protease inhibitor (e.g. Telaprevir or boceprevir) and (pegylated) interferon-alpha (IFN-α)/ribavirin.


Experience with HIV drugs, in particular with HIV protease inhibitors, has taught that sub-optimal pharmacokinetics and complex dosing regimes quickly result in inadvertent compliance failures. This in turn means that the 24 hour trough concentration (minimum plasma concentration) for the respective drugs in an HIV regime frequently falls below the IC90 or ED90 threshold for large parts of the day. It is considered that a 24 hour trough level of at least the IC50, and more realistically, the IC90 or ED90, is essential to slow down the development of drug escape mutants. Achieving the necessary pharmacokinetics and drug metabolism to allow such trough levels provides a stringent challenge to drug design.


The NS5B RdRp is essential for replication of the single-stranded, positive sense, HCV RNA genome. This enzyme has elicited significant interest among medicinal chemists. Both nucleoside and non-nucleoside inhibitors of NS5B are known. Nucleoside inhibitors can act as a chain terminator or as a competitive inhibitor, or as both. In order to be active, nucleoside inhibitors have to be taken up by the cell and converted in vivo to a triphosphate. This conversion to the triphosphate is commonly mediated by cellular kinases, which imparts additional structural requirements on a potential nucleoside polymerase inhibitor. In addition this limits the direct evaluation of nucleosides as inhibitors of HCV replication to cell-based assays capable of in situ phosphorylation.


Several attempts have been made to develop nucleosides as inhibitors of HCV RdRp, but while a handful of compounds have progressed into clinical development, none have proceeded to registration. Amongst the problems which HCV-targeted nucleosides have encountered to date are toxicity, mutagenicity, lack of selectivity, poor efficacy, poor bioavailability, sub-optimal dosage regimes and ensuing high pill burden and cost of goods.


Spirooxetane nucleosides, in particular 1-(8-hydroxy-7-(hydroxy-methyl)-1,6-dioxaspiro[3.4]octan-5-yl)pyrimidine-2,4-dione derivatives and their use as HCV inhibitors are known from WO2010/130726, and WO2012/062869, including CAS-1375074-52-4.


There is a need for HCV inhibitors that may overcome at least one of the disadvantages of current HCV therapy such as side effects, limited efficacy, the emerging of resistance, and compliance failures, or improve the sustained viral response.


The present invention concerns a group of HCV-inhibiting uracyl spirooxetane derivatives with useful properties regarding one or more of the following parameters: antiviral efficacy towards at least one of the following genotypes 1a, 1b, 2a, 2b, 3, 4 and 6, favorable profile of resistance development, lack of toxicity and genotoxicity, favorable pharmacokinetics and pharmacodynamics and ease of formulation and administration.


DESCRIPTION OF THE INVENTION

In one aspect the present invention provides compounds that can be represented by the formula I:




embedded image



including any possible stereoisomer thereof, wherein:


R9 is C1-C6alkyl, phenyl, C3-C7cycloalkyl or C1-C3alkyl substituted with 1, 2 or 3 substituents each independently selected from phenyl, naphtyl, C3-C6cycloalkyl, hydroxy, or C1-C6alkoxy;


or a pharmaceutically acceptable salt or solvate thereof.


Of particular interest are compounds of formula I or subgroups thereof as defined herein that have a structure according to formula Ia:




embedded image


In one embodiment of the present invention, R9 is C1-C6alkyl, phenyl, C3-C7cycloalkyl or C1-C3alkyl substituted with 1 substituent selected from phenyl, C3-C6cycloalkyl, hydroxy, or C1-C6alkoxy. In another embodiment of the present invention, R9 in Formula I or Ia is C1-C6alkyl or C1-C2alkyl substituted with phenyl C1-C2alkoxy or C3-C6cycloalkyl. In a more preferred embodiment, R9 is C2-C4alkyl and in a most preferred embodiment, R9 is i-propyl.


A preferred embodiment according to the invention is a compound according to formula Ib:




embedded image



including any pharmaceutically acceptable salt or solvate thereof and the use of compound (V) in the synthesis of a compound according to Formula I, Ia or Ib.


The invention further relates to a compound of formula V:




embedded image



including any pharmaceutically acceptable salt or solvate thereof and the use of compound (V) in the synthesis of a compound according to Formula I, Ia or Ib.


In addition, the invention relates to a compound of formula VI:




embedded image



including any stereochemical form and/or pharmaceutically acceptable salt or solvate thereof.


Additionally, the invention relates to a pharmaceutical composition comprising a compound according to Formula I, Ia or Ib, and a pharmaceutically acceptable carrier. The invention also relates to a product containing (a) a compound of formula I, Ia or Ib a, and (b) another HCV inhibitor, as a combined preparation for simultaneous, separate or sequential use in the treatment of HCV infections


Yet another aspect of the invention relates to a compound according to Formula I, Ia or Ib or a pharmaceutical composition according to the present invention for use as a medicament, preferably for use in the prevention or treatment of an HCV infection in a mammal.


In a further aspect, the invention provides a compound of formula I Ia or Ib or a pharmaceutically acceptable salt, hydrate, or solvate thereof, for use in the treatment or prophylaxis (or the manufacture of a medicament for the treatment or prophylaxis) of HCV infection. Representative HCV genotypes in the context of treatment or prophylaxis in accordance with the invention include genotype 1b (prevalent in Europe) or 1a (prevalent in North America). The invention also provides a method for the treatment or prophylaxis of HCV infection, in particular of the genotype 1a or 1b.


Of particular interest is compound 8a mentioned in the section “Examples” as well as the pharmaceutically acceptable acid addition salts of this compound.


The compounds of formula I have several centers of chirality, in particular at the carbon atoms 1′, 2′, 3′, and 4′. Although the stereochemistry at these carbon atoms is fixed, the compounds may display at least 75%, preferably at least 90%, such as in excess of 95%, or of 98%, enantiomeric purity at each of the chiral centers.


The phosphorus center can be present as RP or SP, or a mixture of such stereoisomers, including racemates. Diastereoisomers resulting from the chiral phosphorus center and a chiral carbon atom may exist as well.


The compounds of formula I are represented as a defined stereoisomer, except for the stereoisomerism at the phosphorous atom. The absolute configuration of such compounds can be determined using art-known methods such as, for example, X-ray diffraction or NMR and/or implication from starting materials of known stereochemistry. Pharmaceutical compositions in accordance with the invention will preferably comprise stereoisomerically pure forms of the indicated stereoisomer of the particular compound of formula I.


Pure stereoisomeric forms of the compounds and intermediates as mentioned herein are defined as isomers substantially free of other enantiomeric or diastereomeric forms of the same basic molecular structure of said compounds or intermediates. In particular, the term “stereoisomerically pure” concerns compounds or intermediates having a stereoisomeric excess of at least 80% (i.e. minimum 90% of one isomer and maximum 10% of the other possible isomers) up to a stereoisomeric excess of 100% (i.e. 100% of one isomer and none of the other), more in particular, compounds or intermediates having a stereoisomeric excess of 90% up to 100%, even more in particular having a stereoisomeric excess of 94% up to 100% and most in particular having a stereoisomeric excess of 97% up to 100%, or of 98% up to 100%. The terms “enantiomerically pure” and “diastereomerically pure” should be understood in a similar way, but then having regard to the enantiomeric excess, and the diastereomeric excess, respectively, of the mixture in question.


Pure stereoisomeric forms of the compounds and intermediates of this invention may be obtained by the application of art-known procedures. For instance, enantiomers may be separated from each other by the selective crystallization of their diastereomeric salts with optically active acids or bases. Examples thereof are tartaric acid, dibenzoyl-tartaric acid, ditoluoyltartaric acid and camphorsulfonic acid. Alternatively, enantiomers may be separated by chromatographic techniques using chiral stationary layers. Said pure stereochemically isomeric forms may also be derived from the corresponding pure stereochemically isomeric forms of the appropriate starting materials, provided that the reaction occurs stereospecifically. Preferably, if a specific stereoisomer is desired, said compound is synthesized by stereospecific methods of preparation. These methods will advantageously employ enantiomerically pure starting materials.


The diastereomeric racemates of the compounds of formula I can be obtained separately by conventional methods. Appropriate physical separation methods that may advantageously be employed are, for example, selective crystallization and chromatography, e.g. column chromatography.


The pharmaceutically acceptable addition salts comprise the therapeutically active non-toxic acid and base addition salt forms of the compounds of formula I. Of interest are the free, i.e. non-salt forms of the compounds of formula I, or of any subgroup of compounds of formula I specified herein.


The pharmaceutically acceptable acid addition salts can conveniently be obtained by treating the base form with such appropriate acid. Appropriate acids comprise, for example, inorganic acids such as hydrohalic acids, e.g. hydrochloric or hydrobromic acid, sulfuric, nitric, phosphoric and the like acids; or organic acids such as, for example, acetic, propionic, hydroxyacetic, lactic, pyruvic, oxalic (i.e. ethanedioic), malonic, succinic (i.e. butanedioic acid), maleic, fumaric, malic (i.e. hydroxyl-butanedioic acid), tartaric, citric, methanesulfonic, ethanesulfonic, benzenesulfonic, p-toluenesulfonic, cyclamic, salicylic, p-aminosalicylic, palmoic and the like acids. Conversely said salt forms can be converted by treatment with an appropriate base into the free base form.


The compounds of formula I containing an acidic proton may also be converted into their non-toxic metal or amine addition salt forms by treatment with appropriate organic and inorganic bases. Appropriate base salt forms comprise, for example, the ammonium salts, the alkali and earth alkaline metal salts, e.g. the lithium, sodium, potassium, magnesium, calcium salts and the like, salts with organic bases, e.g. the benzathine, N-methyl-D-glucamine, hydrabamine salts, and salts with amino acids such as, for example, arginine, lysine and the like.


The term “solvates” covers any pharmaceutically acceptable solvates that the compounds of formula I as well as the salts thereof, are able to form. Such solvates are for example hydrates, alcoholates, e.g. ethanolates, propanolates, and the like.


Some of the compounds of formula I may also exist in their tautomeric form. For example, tautomeric forms of amide (—C(═O)—NH—) groups are iminoalcohols (—C(OH)═N—), which can become stabilized in rings with aromatic character. The uridine base is an example of such a form. Such forms, although not explicitly indicated in the structural formulae represented herein, are intended to be included within the scope of the present invention.





SHORT DESCRIPTION OF THE FIGURE


FIG. 1: In vivo efficacy of compound 8a and CAS-1375074-52-4 as determined in a humanized hepatocyte mouse model.





DEFINITIONS

As used herein “C1-Cnalkyl” as a group or part of a group defines saturated straight or branched chain hydrocarbon radicals having from 1 to n carbon atoms. Accordingly, “C1-C4alkyl” as a group or part of a group defines saturated straight or branched chain hydrocarbon radicals having from 1 to 4 carbon atoms such as for example methyl, ethyl, 1-propyl, 2-propyl, 1-butyl, 2-butyl, 2-methyl-1-propyl, 2-methyl-2-propyl. “C1-C6alkyl” encompasses C1-C4alkyl radicals and the higher homologues thereof having 5 or 6 carbon atoms such as, for example, 1-pentyl, 2-pentyl, 3-pentyl, 1-hexyl, 2-hexyl, 2-methyl-1-butyl, 2-methyl-1-pentyl, 2-ethyl-1-butyl, 3-methyl-2-pentyl, and the like. Of interest amongst C1-C6alkyl is C1-C4alkyl.


‘C1-Cnalkoxy’ means a radical —O—C1-Cnalkyl wherein C1-Cnalkyl is as defined above. Accordingly, ‘C1-C6alkoxy’ means a radical —O—C1-C6alkyl wherein C1-C6alkyl is as defined above. Examples of C1-C6alkoxy are methoxy, ethoxy, n-propoxy, or isopropoxy. Of interest is ‘C1-C2alkoxy’, encompassing methoxy and ethoxy.


“C3-C6cycloalkyl” includes cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl.


In one embodiment, the term “phenyl-C1-C6alkyl” is benzyl.


As used herein, the term ‘(═O)’ or ‘oxo’ forms a carbonyl moiety when attached to a carbon atom. It should be noted that an atom can only be substituted with an oxo group when the valency of that atom so permits.


The term “monophosphate, diphosphate or triphosphate ester” refers to groups:




embedded image


Where the position of a radical on a molecular moiety is not specified (for example a substituent on phenyl) or is represented by a floating bond, such radical may be positioned on any atom of such a moiety, as long as the resulting structure is chemically stable. When any variable is present more than once in the molecule, each definition is independent.


Whenever used herein, the term ‘compounds of formula I’, or ‘the present compounds’ or similar terms, it is meant to include the compounds of Formula I, Ia and Ib, including the possible stereochemically isomeric forms, and their pharmaceutically acceptable salts and solvates.


The present invention also includes isotope-labeled compounds of formula I or any subgroup of formula I, wherein one or more of the atoms is replaced by an isotope that differs from the one(s) typically found in nature. Examples of such isotopes include isotopes of hydrogen, such as 2H and 3H; carbon, such as 11C, 13C and 14C; nitrogen, such as 13N and 15N; oxygen, such as 15O, 17O and 18O; phosphorus, such as 31P and 32P, sulphur, such as 35S; fluorine, such as 18F; chlorine, such as 36Cl; bromine such as 75Br, 76Br, 77Br and 82Br; and iodine, such as 123I, 124I, 125I and 131I. Isotope-labeled compounds of the invention can be prepared by processes analogous to those described herein by using the appropriate isotope-labeled reagents or starting materials, or by art-known techniques. The choice of the isotope included in an isotope-labeled compound depends on the specific application of that compound. For example, for tissue distribution assays, a radioactive isotope such as 3H or 14C is incorporated. For radio-imaging applications, a positron emitting isotope such as 11C, 18F, 13N or 15O will be useful. The incorporation of deuterium may provide greater metabolic stability, resulting in, e.g. an increased in vivo half life of the compound or reduced dosage requirements.


General Synthetic Procedures


The following schemes are just meant to be illustrative and are by no means limiting the scope.


The starting material 1-[(4R,5R,7R,8R)-8-hydroxy-7-(hydroxymethyl)-1,6-dioxaspiro[3.4]octan-5-yl]pyrimidine-2,4(1H,3H)-dione (1) can be prepared as exemplified in WO2010/130726. Compound (1) is converted into compounds of the present invention via a p-methoxybenzyl protected derivative (4) as exemplified in the following Scheme 1.




embedded image


In Scheme 1, R9 can be C1-C6alkyl, phenyl, naphtyl, C3-C7cycloalkyl or C1-C3alkyl substituted with 1, 2 or 3 substituents each independently selected from phenyl, C3-C6cycloalkyl, hydroxy, or C1-C6alkoxy, preferably R9 is C1-C6alkyl or C1-C2alkyl substituted with phenyl, C1-C2alkoxy or C3-C6cycloalkyl, even more preferably R9 is C2-C4alkyl and most preferably R9 is i-propyl.


In a further aspect, the present invention concerns a pharmaceutical composition comprising a therapeutically effective amount of a compound of formula I as specified herein, and a pharmaceutically acceptable carrier. Said composition may contain from 1% to 50%, or from 10% to 40% of a compound of formula I and the remainder of the composition is the said carrier. A therapeutically effective amount in this context is an amount sufficient to act in a prophylactic way against HCV infection, to inhibit HCV, to stabilize or to reduce HCV infection, in infected subjects or subjects being at risk of becoming infected. In still a further aspect, this invention relates to a process of preparing a pharmaceutical composition as specified herein, which comprises intimately mixing a pharmaceutically acceptable carrier with a therapeutically effective amount of a compound of formula I, as specified herein.


The compounds of formula I or of any subgroup thereof may be formulated into various pharmaceutical forms for administration purposes. As appropriate compositions there may be cited all compositions usually employed for systemically administering drugs. To prepare the pharmaceutical compositions of this invention, an effective amount of the particular compound, optionally in addition salt form or metal complex, as the active ingredient is combined in intimate admixture with a pharmaceutically acceptable carrier, which carrier may take a wide variety of forms depending on the form of preparation desired for administration. These pharmaceutical compositions are desirable in unitary dosage form suitable, particularly, for administration orally, rectally, percutaneously, or by parenteral injection. For example, in preparing the compositions in oral dosage form, any of the usual pharmaceutical media may be employed such as, for example, water, glycols, oils, alcohols and the like in the case of oral liquid preparations such as suspensions, syrups, elixirs, emulsions and solutions; or solid carriers such as starches, sugars, kaolin, lubricants, binders, disintegrating agents and the like in the case of powders, pills, capsules, and tablets. Because of their ease in administration, tablets and capsules represent the most advantageous oral dosage unit forms, in which case solid pharmaceutical carriers are obviously employed. For parenteral compositions, the carrier will usually comprise sterile water, at least in large part, though other ingredients, for example, to aid solubility, may be included. Injectable solutions, for example, may be prepared in which the carrier comprises saline solution, glucose solution or a mixture of saline and glucose solution. Injectable suspensions may also be prepared in which case appropriate liquid carriers, suspending agents and the like may be employed. Also included are solid form preparations intended to be converted, shortly before use, to liquid form preparations. In the compositions suitable for percutaneous administration, the carrier optionally comprises a penetration enhancing agent and/or a suitable wetting agent, optionally combined with suitable additives of any nature in minor proportions, which additives do not introduce a significant deleterious effect on the skin. The compounds of the present invention may also be administered via oral inhalation or insufflation in the form of a solution, a suspension or a dry powder using any art-known delivery system.


It is especially advantageous to formulate the aforementioned pharmaceutical compositions in unit dosage form for ease of administration and uniformity of dosage. Unit dosage form as used herein refers to physically discrete units suitable as unitary dosages, each unit containing a predetermined quantity of active ingredient calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. Examples of such unit dosage forms are tablets (including scored or coated tablets), capsules, pills, suppositories, powder packets, wafers, injectable solutions or suspensions and the like, and segregated multiples thereof.


The compounds of formula I show activity against HCV and can be used in the treatment and/or prophylaxis of HCV infection or diseases associated with HCV. The latter include progressive liver fibrosis, inflammation and necrosis leading to cirrhosis, end-stage liver disease, and HCC. The compounds of this invention moreover are believed to be active against mutated strains of HCV and show a favorable pharmacokinetic profile and have attractive properties in terms of bioavailability, including an acceptable half-life, AUC (area under the curve) and peak values and lacking unfavorable phenomena such as insufficient quick onset and tissue retention.


The in vitro antiviral activity against HCV of the compounds of formula I can be tested in a cellular HCV replicon system based on Lohmann et al. (1999) Science 285:110-113, with the further modifications described by Krieger et al. (2001) Journal of Virology 75: 4614-4624 (incorporated herein by reference), which is further exemplified in the examples section. This model, while not a complete infection model for HCV, is widely accepted as the most robust and efficient model of autonomous HCV RNA replication currently available. It will be appreciated that it is important to distinguish between compounds that specifically interfere with HCV functions from those that exert cytotoxic or cytostatic effects in the HCV replicon model, and as a consequence cause a decrease in HCV RNA or linked reporter enzyme concentration. Assays are known in the field for the evaluation of cellular cytotoxicity based for example on the activity of mitochondrial enzymes using fluorogenic redox dyes such as resazurin. Furthermore, cellular counter screens exist for the evaluation of non-selective inhibition of linked reporter gene activity, such as firefly luciferase. Appropriate cell types can be equipped by stable transfection with a luciferase reporter gene whose expression is dependent on a constitutively active gene promoter, and such cells can be used as a counter-screen to eliminate non-selective inhibitors.


Due to their anti-HCV properties, the compounds of formula I, including any possible stereoisomers, the pharmaceutically acceptable addition salts or solvates thereof, are useful in the treatment of warm-blooded animals, in particular humans, infected with HCV, and in the prophylaxis of HCV infections. The compounds of the present invention may therefore be used as a medicine, in particular as an anti-HCV or a HCV-inhibiting medicine. The present invention also relates to the use of the present compounds in the manufacture of a medicament for the treatment or the prevention of HCV infection. In a further aspect, the present invention relates to a method of treating a warm-blooded animal, in particular human, infected by HCV, or being at risk of becoming infected by HCV, said method comprising the administration of an anti-HCV effective amount of a compound of formula I, as specified herein. Said use as a medicine or method of treatment comprises the systemic administration to HCV-infected subjects or to subjects susceptible to HCV infection of an amount effective to combat the conditions associated with HCV infection.


In general it is contemplated that an antiviral effective daily amount would be from about 1 to about 30 mg/kg, or about 2 to about 25 mg/kg, or about 5 to about 15 mg/kg, or about 8 to about 12 mg/kg body weight. Average daily doses can be obtained by multiplying these daily amounts by about 70. It may be appropriate to administer the required dose as two, three, four or more sub-doses at appropriate intervals throughout the day. Said sub-doses may be formulated as unit dosage forms, for example, containing about 1 to about 2000 mg, or about 50 to about 1500 mg, or about 100 to about 1000 mg, or about 150 to about 600 mg, or about 100 to about 400 mg of active ingredient per unit dosage form.


As used herein the term “about” has the meaning known to the person skilled in the art. In certain embodiments the term “about” may be left out and the exact amount is meant. In other embodiments the term “about” means that the numerical following the term “about” is in the range of ±15%, or of ±10%, or of ±5%, or of ±1%, of said numerical value.


Examples



embedded image


Synthesis of Compound (2)


Compound (2) can be prepared by dissolving compound (1) in pyridine and adding 1,3-dichloro-1,1,3,3-tetraisopropyldisiloxane. The reaction is stirred at room temperature until complete. The solvent is removed and the product redissolved in CH2Cl2 and washed with saturated NaHCO3 solution. Drying on MgSO4 and removal of the solvent gives compound (2).


Synthesis of Compound (3)


Compound (3) is prepared by reacting compound (2) with p-methoxybenzylchloride in the presence of DBU as the base in CH3CN.


Synthesis of Compound (4)


Compound (4) is prepared by cleavage of the bis-silyl protecting group in compound (3) using TBAF as the fluoride source.


Synthesis of Compound (6a)


A solution of isopropyl alcohol (3.86 mL, 0.05 mol) and triethylamine (6.983 mL, 0.05 mol) in dichloromethane (50 mL) was added to a stirred solution of POCl3 (5) (5.0 mL, 0.0551 mol) in DCM (50 mL) dropwise over a period of 25 min at −5° C. After the mixture stirred for 1 h, the solvent was evaporated, and the residue was suspended in ether (100 mL). The triethylamine hydrochloride salt was filtered and washed with ether (20 mL). The filtrate was concentrated, and the residue was distilled to give the (6) as a colorless liquid (6.1 g, 69% yield).


Synthesis of Compound (7a)


To a stirred suspension of (4) (2.0 g, 5.13 mmol) in dichloromethane (50 mL) was added triethylamine (2.07 g, 20.46 mmol) at room temperature. The reaction mixture was cooled to −20° C., and then (6a) (1.2 g, 6.78 mmol) was added dropwise over a period of 10 min. The mixture was stirred at this temperature for 15 min and then NMI was added (0.84 g, 10.23 mmol), dropwise over a period of 15 min. The mixture was stirred at −15° C. for 1 h and then slowly warmed to room temperature in 20 h. The solvent was evaporated, the mixture was concentrated and purified by column chromatography using petroleum ether/EtOAc (10:1 to 5:1 as a gradient) to give (7a) as white solid (0.8 g, 32% yield).


Synthesis of Compound (8a)


To a solution of (7a) in CH3CN (30 mL) and H2O (7 mL) was add CAN portion wise below 20° C. The mixture was stirred at 15-20° C. for 5 h under N2. Na2SO3 (370 mL) was added dropwise into the reaction mixture below 15° C., and then Na2CO3 (370 mL) was added. The mixture was filtered and the filtrate was extracted with CH2Cl2 (100 mL*3). The organic layer was dried and concentrated to give the residue. The residue was purified by column chromatography to give the target compound (8a) as white solid. (Yield: 55%)



1H NMR (400 MHz, CHLOROFORM-d) δ ppm 1.45 (dd, J=7.53, 6.27 Hz, 6H), 2.65-2.84 (m, 2H), 3.98 (td, J=10.29, 4.77 Hz, 1H), 4.27 (t, J=9.66 Hz, 1H), 4.43 (ddd, J=8.91, 5.77, 5.65 Hz, 1H), 4.49-4.61 (m, 1H), 4.65 (td, J=7.78, 5.77 Hz, 1H), 4.73 (d, J=7.78 Hz, 1H), 4.87 (dq, J=12.74, 6.30 Hz, 1H), 5.55 (br. s., 1H), 5.82 (d, J=8.03 Hz, 1H), 7.20 (d, J=8.03 Hz, 1H), 8.78 (br. s., 1H); 31P NMR (CHLOROFORM-d) δ ppm −7.13; LC-MS: 375 (M+1)+




embedded image


Step 1: Synthesis of Compound (9)


Compound (1), CAS 1255860-33-3 (1200 mg, 4.33 mmol) and 1,8-bis(dimethyl-amino)naphthalene (3707 mg, 17.3 mmol) were dissolved in 24.3 mL of trimethylphosphate. The solution was cooled to 0° C. Compound (5) (1.21 mL, 12.98 mmol) was added, and the mixture was stirred well maintaining the temperature at 0° C. for 5 hours. The reaction was quenched by addition of 120 mL of tetraethyl-ammonium bromide solution (1M) and extracted with CH2Cl2 (2×80 mL). Purification was done by preparative HPLC (Stationary phase: RP XBridge Prep C18 OBD-10 μm, 30×150 mm, mobile phase: 0.25% NH4HCO3 solution in water, CH3CN), yielding two fractions. The purest fraction was dissolved in water (15 mL) and passed through a manually packed Dowex (H+) column by elution with water. The end of the elution was determined by checking UV absorbance of eluting fractions. Combined fractions were frozen at −78° C. and lyophilized. Compound (9) was obtained as a white fluffy solid (303 mg, (0.86 mmol, 20% yield), which was used immediately in the following reaction.


Step 2: Preparation of Compound (VI)


Compound (9) (303 mg, 0.86 mmol) was dissolved in 8 mL water and to this solution was added N,N′-Dicyclohexyl-4-morpholine carboxamidine (253.8 mg, 0.86 mmol) dissolved in pyridine (8.4 mL). The mixture was kept for 5 minutes and then evaporated to dryness, dried overnight in vacuo overnight at 37° C. The residue was dissolved in pyridine (80 mL). This solution was added dropwise to vigorously stirred DCC (892.6 mg, 4.326 mmol) in pyridine (80 mL) at reflux temperature. The solution was kept refluxing for 1.5 h during which some turbidity was observed in the solution. The reaction mixture was cooled and evaporated to dryness. Diethylether (50 mL) and water (50 mL) were added to the solid residue. N′N-dicyclohexylurea was filtered off, and the aqueous fraction was purified by preparative HPLC (Stationary phase: RP XBridge Prep C18 OBD-10 μm, 30×150 mm, mobile phase: 0.25% NH4HCO3 solution in water, CH3CN), yielding a white solid which was dried overnight in vacuo at 38° C. (185 mg, 0.56 mmol, 65% yield). LC-MS: (M+H)+: 333.



1H NMR (400 MHz, DMSO-d6) δ ppm 2.44-2.59 (m, 2H) signal falls under DMSO signal, 3.51 (td, J=9.90, 5.50 Hz, 1H), 3.95-4.11 (m, 2H), 4.16 (d, J=10.34 Hz, 1H), 4.25-4.40 (m, 2H), 5.65 (d, J=8.14 Hz, 1H), 5.93 (br. s., 1H), 7.46 (d, J=7.92 Hz, 1H), 2H's not observed


Biological Examples

Replicon Assays


The compounds of formula I were examined for activity in the inhibition of HCV-RNA replication in a cellular assay. The assay was used to demonstrate that the compounds of formula I inhibited a HCV functional cellular replicating cell line, also known as HCV replicons. The cellular assay was based on a bicistronic expression construct, as described by Lohmann et al. (1999) Science vol. 285 pp. 110-113 with modifications described by Krieger et al. (2001) Journal of Virology 75: 4614-4624, in a multi-target screening strategy.


Replicon Assay (A)


In essence, the method was as follows. The assay utilized the stably transfected cell line Huh-7 luc/neo (hereafter referred to as Huh-Luc). This cell line harbors an RNA encoding a bicistronic expression construct comprising the wild type NS3-NS5B regions of HCV type 1b translated from an internal ribosome entry site (IRES) from encephalomyocarditis virus (EMCV), preceded by a reporter portion (FfL-luciferase), and a selectable marker portion (neoR, neomycine phosphotransferase). The construct is bordered by 5′ and 3′ NTRs (non-translated regions) from HCV genotype 1b.


Continued culture of the replicon cells in the presence of G418 (neoR) is dependent on the replication of the HCV-RNA. The stably transfected replicon cells that express HCV-RNA, which replicates autonomously and to high levels, encoding inter alia luciferase, were used for screening the antiviral compounds.


The replicon cells were plated in 384-well plates in the presence of the test and control compounds which were added in various concentrations. Following an incubation of three days, HCV replication was measured by assaying luciferase activity (using standard luciferase assay substrates and reagents and a Perkin Elmer ViewLux™ ultraHTS microplate imager). Replicon cells in the control cultures have high luciferase expression in the absence of any inhibitor. The inhibitory activity of the compound on luciferase activity was monitored on the Huh-Luc cells, enabling a dose-response curve for each test compound. EC50 values were then calculated, which value represents the amount of the compound required to decrease the level of detected luciferase activity by 50%, or more specifically, the ability of the genetically linked HCV replicon RNA to replicate.


Results (A)


Table 1 shows the replicon results (EC50, replicon) and cytotoxicity results (CC50 (μM) (Huh-7)) obtained for the compound of the examples given above.











TABLE 1





Compound

CC50 (μM)


Number
EC50 (μM) (HCV)
(Huh-7)







8a
0.13 (n = 4)
>100









Replicon Assays (B)


Further replicon assays were performed with compound 8a of which the protocols and results are disclosed below.


Assay 1


The anti-HCV activity of compound 8a was tested in cell culture with replicon cells generated using reagents from the Bartenschlager laboratory (the HCV 1b bicistronic subgenomic luciferase reporter replicon clone ET). The protocol included a 3-day incubation of 2500 replicon cells in a 384-well format in a nine-point 1:4 dilution series of the compound. Dose response curves were generated based on the firefly luciferase read-out. In a variation of this assay, a 3 day incubation of 3000 cells in a 96-well format in a nine-point dilution series was followed by qRT-PCR Taqman detection of HCV genome, and normalized to the cellular transcript, RPL13 (of the ribosomal subunit RPL13 gene) as a control for compound inhibition of cellular transcription.


Assay 2


The anti-HCV activity of compound 8a was tested in cell culture with replicon cells generated using reagents from the Bartenschlager laboratory (the HCV 1b bicistronic subgenomic luciferase reporter replicon clone ET or Huh-Luc-Neo). The protocol included a 3-day incubation of 2×104 replicon cells in a 96-well format in a six-point 1:5 dilution series of the compound. Dose response curves were generated based on the luciferase read-out.


Assay 3


The anti-HCV activity of compound 8a was tested in cell culture with replicon cells generated using reagents from the Bartenschlager laboratory (the HCV 1b bicistronic subgenomic luciferase reporter replicon clone ET or Huh-Luc-Neo). The protocol included either a 3-day incubation of 8×103 cells or 2×104 cells in a 96-well format in an eight-point 1:5 dilution series of the compound. Dose response curves were generated based on the luciferase read-out.


Results


Table 2 shows the average replicon results (EC50, replicon) obtained for compound 8a following assays as given above.












TABLE 2







Assay
Average EC50 value (8a):




















1
57
μM (n = 8)



2
17.5
μM (n = 4)



3
>100
μM (n = 1)










Primary Human Hepatocyte In Vitro Assay


The anti-HCV activity of compound 8a was determined in an in vitro primary human hepatocyte assay. Protocols and results are disclosed below.


Protocol


Hepatocyte Isolation and Culture


Primary human hepatocytes (PHH) were prepared from patients undergoing partial hepatectomy for metastases or benign tumors. Fresh human hepatocytes were isolated from encapsulated liver fragments using a modification of the two-step collagenase digestion method. Briefly, encapsulated liver tissue was placed in a custom-made perfusion apparatus and hepatic vessels were cannulated with tubing attached multichannel manifold. The liver fragment was initially perfused for 20 min with a prewarmed (37° C.) calcium-free buffer supplemented with ethylene glycol tetraacetic acid (EGTA) followed by perfusion with a prewarmed (37° C.) buffer containing calcium (CaCl2), H2O2) and collagenase 0.05% for 10 min. Then, liver fragment was gently shaken to free liver cells in Hepatocyte Wash Medium. Cellular suspension was filtered through a gauze-lined funnel. Cells were centrifuged at low speed centrifugation. The supernatant, containing damaged or dead hepatocytes, non parenchymal cells and debris was removed and pelleted hepatocytes were re-suspended in Hepatocyte Wash Medium. Viability and cell concentration were determined by trypan blue exclusion test.


Cells were resuspended in complete hepatocyte medium consisting of William's medium (Invitrogen) supplemented with 100 IU/L insulin (Novo Nordisk, France), and 10% heat inactivated fetal calf serum (Biowest, France), and seeded at a density 1.8×106 viable cells onto 6 well plates that had been precoated with a type I collagen from calf skin (Sigma-Aldrich, France) The medium was replaced 16-20 hours later with fresh complete hepatocyte medium supplemented with hydrocortisone hemisuccinate (SERB, Paris, France), and cells were left in this medium until HCV inoculation. The cultures were maintained at 37° C. in a humidified 5% CO2 atmosphere.


The PHHs were inoculated 3 days after seeding. JFH1-HCVcc stocks were used to inoculate PHHs for 12 hours, at a multiplicity of infection (MOI) of 0.1 ffu per cell. After a 12-hours incubation at 37° C., the inoculum was removed, and monolayers were washed 3 times with phosphate-buffered saline and incubated in complete hepatocyte medium containing 0.1% dimethylsulfoxide as carrier control, 100 IU/ml of IFNalpha as negative control or else increasing concentrations of compound 8a. The cultures then were maintained during 3 days.


Quantitation of HCV RNA


Total RNA was prepared from cultured cells or from filtered culture supernatants using the RNeasy or Qiamp viral RNA minikit respectively (Qiagen SA, Courtaboeuf, France) according to the manufacturer's recommendations. HCV RNA was quantified in cells and culture supernatants using a strand-specific reverse real-time PCR technique described previously (Carrière M and al 2007):


Reverse transcription was performed using primers described previously located in the 50 NCR region of HCV genome, tag-RC1 (5′-GGCCGTCATGGTGGCGAATAAGTCTAGCCATGGCGTTAGTA-3′) and RC21 (5′-CTCCCGGGGCACTCGCAAGC-3′) for the negative and positive strands, respectively. After a denaturation step performed at 70° C. for 8 min, the RNA template was incubated at 4° C. for 5 min in the presence of 200 ng of tag-RC1 primer and 1.25 mM of each deoxynucleoside triphosphate (dNTP) (Promega, Charbonnieres, France) in a total volume of 12 μl.


Reverse transcription was carried out for 60 min at 60° C. in the presence of 20 U RNaseOut™ (Invitrogen, Cergy Pontoise, France) and 7.5 U Thermoscript™ reverse transcriptase (Invitrogen), in the buffer recommended by the manufacturer. An additional treatment was applied by adding 1 μl (2 U) RNaseH (Invitrogen) for 20 min at 37° C.


The first round of nested PCR was performed with 2 μl of the cDNA obtained in a total volume of 50 μl, containing 3 U Taq polymerase (Promega), 0.5 mM dNTP, and 0.5 μM RC1 (5′-GTCTAGCCATGGCGTTAGTA-3′) and RC21 primers for positive-strand amplification, or Tag (5′-GGCCGTCATGGTGGCGAATAA-3′) and RC21 primers for negative strand amplification. The PCR protocol consisted of 18 cycles of denaturation (94° C. for 1 min), annealing (55° C. for 45 sec), and extension (72° C. for 2 min). The cDNA obtained was purified using the kit from Qiagen, according to the manufacturer's instructions.


The purified product was then subjected to real-time PCR. The reaction was carried out using the LightCycler 480 SYBR Green I Master (2× con) Kit (Roche, Grenoble, France), with LC480 instruments and technology (Roche Diagnostics). PCR amplifications were performed in a total volume of 10 μl, containing 5 μl of Sybrgreen I Master Mix (2×), and 25 ng of the 197R (5′-CTTTCGCGACCCAACACTAC-3′) and 104 (5′-AGAGCCATAGTGGTCTGCGG-3′) primers. The PCR protocol consisted of one step of initial denaturation for 10 min at 94° C., followed by 40 cycles of denaturation (95° C. for 15 sec), annealing (57° C. for 5 sec), and extension (72° C. for 8 sec).


The quantitation of 28Sr RNA by specific RT-PCR was used as an internal standard to express the results of HCV positive or negative strands per g of total hepatocyte RNA. Specific primers for 28 S rRNA were designed using the Oligo6 software 5′-TTGAAAATCCGGGGGAGAG-3′ (nt2717-2735) and 50-ACATTGTTCCAACATGCCAG-30 (nt 2816-2797). Reverse transcription was performed using AMV reverse transcriptase (Promega), and the PCR protocol consisted of one step of initial denaturation for 8 min at 95° C., followed by 40 cycles of denaturation (95° C. for 15 sec), annealing (54° C. for 5 sec), and extension (72° C. for 5 sec).


Results


Table 3 shows the anti-HCV activity of compound 8a as determined in the in vitro primary human hepatocyte assay described above. The numbers are expressed as 106 HCV RNA copies/μg of total RNA. Results of two independent experiments (Exp 1 and Exp 2) are given. The data per experiment is the average of two measurements.


Table 3: Effect of compound 8a on positive strand HCV-RNA levels in primary human hepatocytes (expressed as 106 HCV RNA copies/μg of total RNA).











TABLE 3






Exp. 1
Exp. 2

















No HCV
0
0


HCV control
3.56
5.53










IFNα
(100 IU/mL)
1.48
1.59


8a
(0.195 μM)
2.18
1.12


8a
(0.78 μM)
2.25
1.3


8a
(3.12 μM)
1.09
0.94


8a
(12.5 μM)
2.17
1.3


8a
(50 μM)
0.94
1.33









In Vivo Efficacy Assay


The in vivo efficacy of compound 8a and CAS-1375074-52-4 was determined in a humanized hepatocyte mouse model (PBX-mouse) as previously described in Inoue et. al (Hepatology. 2007 April; 45(4):921-8) and Tenato et. al. (Am J Pathol 2004; 165-901-912) with the following specification: Test animals: HCV G1a-infected PXB-mice, male or female, >70% replacement index of human hepatocytes. Dosing was performed p.o for 7 days at doses indicated below wherein QD represents a single dose per day, BID represents two doses per day.


Efficacy of compound 8a was compared to CAS-1375074-52-4. Results are indicated in FIG. 1. The FIGURE shows the log drop HCV viral RNA after dosing for a period of 7 days.



FIG. 1 clearly shows that a dosing of 100 mg/kg QD for CAS 1375074-52-4 (indicated as *, n=4) does not result in a significant log drop in HCV viral RNA. This in strong contrast to each of the indicated dose regimens for compound 8a, were a clear log drop is observed for 100 mg/kg QD (indicated as ♦, n=3), 200 mg/kg QD (indicated as ●, n=4), 50 mg/kg BID (indicated as ▪, n=4). The most pronounced log drop effect in viral RNA is observed after a 7 day dosing of compound 8a at 100 mg/kg BID (indicated as ▴, n=4).

Claims
  • 1. A method of preparing a compound of formula (7),
  • 2. A method as claimed in claim 1, wherein said reacting a compound of formula (6) with a compound of formula (4) takes place in the presence of triethylamine.
  • 3. A method as claimed in claim 1, wherein said reacting a compound of formula (6) with a compound of formula (4) takes place in the presence of N-methylimidazole.
  • 4. A method as claimed in claim 1, wherein said reacting a compound of formula (6) with a compound of formula (4) takes place in dichloromethane.
  • 5. A method as claimed in claim 1, wherein said reacting a compound of formula (6) with a compound of formula (4) takes place in the presence of triethylamine and N-methylimidazole in dichloromethane.
  • 6. A method as claimed in claim 1, wherein R9 is C1-C6alkyl.
  • 7. A method as claimed in claim 1, wherein R9 is C3-C7cycloalkyl.
  • 8. A method as claimed in claim 1, wherein R9 is isopropyl.
  • 9. A method as claimed in claim 1, wherein R9 is C2-C4alkyl.
  • 10. A method as claimed in claim 1, wherein R9 is C1-C2alkyl substituted with phenyl.
Priority Claims (1)
Number Date Country Kind
12169425 May 2012 EP regional
Parent Case Info

This application is a continuation of U.S. application Ser. No. 16/044,262 filed on Jul. 24, 2018, pending, which is a continuation of U.S. application Ser. No. 15/809,730 filed on Nov. 10, 2017, now U.S. Pat. No. 10,040,814 issued on Aug. 7, 2018, which is a continuation of U.S. application Ser. No. 15/423,985 filed on Feb. 3, 2017, now U.S. Pat. No. 9,845,336 issued on Dec. 19, 2017, which is a continuation of U.S. application Ser. No. 15/238,553 filed on Aug. 16, 2016, abandoned, which is a continuation application of U.S. application Ser. No. 14/403,587 filed on Nov. 25, 2014, now U.S. Pat. No. 9,422,323 issued on Aug. 23, 2016, which is a 35 U.S.C. § 371 nationalization of PCT application PCT/EP2013/060704 filed on May 24, 2013, which claims priority to European patent application EP 12169425.1 filed on May 25, 2012, all of which are incorporated herein by reference.

US Referenced Citations (323)
Number Name Date Kind
3480613 Walton et al. Nov 1969 A
3817978 Jenkins et al. Jun 1974 A
3852267 Meyer, Jr. et al. Dec 1974 A
4713383 Francis et al. Dec 1987 A
5049551 Koda et al. Sep 1991 A
5679342 Houghton et al. Oct 1997 A
5712088 Houghton et al. Jan 1998 A
5714596 Houghton et al. Feb 1998 A
5863719 Houghton et al. Jan 1999 A
6027729 Houghton et al. Feb 2000 A
6074816 Houghton et al. Jun 2000 A
6096541 Houghton et al. Aug 2000 A
6171782 Houghton et al. Jan 2001 B1
6174868 Anderson et al. Jan 2001 B1
6284458 Anderson et al. Sep 2001 B1
6348587 Schinazi et al. Feb 2002 B1
6391542 Anderson et al. May 2002 B1
6423489 Anderson et al. Jul 2002 B1
6433159 Anderson Aug 2002 B1
6455513 McGuigan et al. Sep 2002 B1
6495677 Ramasamy et al. Dec 2002 B1
6566365 Storer May 2003 B1
6573247 McGuigan et al. Jun 2003 B1
6608191 Anderson et al. Aug 2003 B1
6638919 McGuigan et al. Oct 2003 B2
6660721 Devos et al. Dec 2003 B2
6777395 Bhat et al. Aug 2004 B2
6784161 Ismaili et al. Aug 2004 B2
6784166 Devos et al. Aug 2004 B2
6787525 Schott et al. Sep 2004 B1
6800751 Sanghvi et al. Oct 2004 B2
6812219 LaColla et al. Nov 2004 B2
6833361 Hong et al. Dec 2004 B2
6846810 Martin et al. Jan 2005 B2
6897302 Kowalczyk et al. May 2005 B2
6911424 Schinazi et al. Jun 2005 B2
6914054 Sommadossi et al. Jul 2005 B2
6927291 Jin et al. Aug 2005 B2
6995146 Anderson et al. Feb 2006 B2
7018989 McGuigan et al. Mar 2006 B2
7019135 McGuigan et al. Mar 2006 B2
7094770 Watanabe et al. Aug 2006 B2
7105493 Sommadossi et al. Sep 2006 B2
7105499 Carroll et al. Sep 2006 B2
7115590 Daluge et al. Oct 2006 B1
7125855 Bhat et al. Oct 2006 B2
7138376 Gosselin et al. Nov 2006 B2
7148206 Sommadossi et al. Dec 2006 B2
7157441 Sommadossi et al. Jan 2007 B2
7163929 Sommadossi et al. Jan 2007 B2
7169766 Sommadossi et al. Jan 2007 B2
7192936 LaColla et al. Mar 2007 B2
7202224 Eldrup et al. Apr 2007 B2
7300924 Boojamra et al. Nov 2007 B2
7307065 Schinazi et al. Dec 2007 B2
7323449 Olsen et al. Jan 2008 B2
7323453 Olsen et al. Jan 2008 B2
7339054 Xu et al. Mar 2008 B2
7365057 LaColla et al. Apr 2008 B2
7378402 Martin et al. May 2008 B2
7384924 LaColla et al. Jun 2008 B2
7405204 Roberts et al. Jul 2008 B2
7429572 Clark Sep 2008 B2
7452901 Boojamra et al. Nov 2008 B2
7456155 Sommadossi et al. Nov 2008 B2
7524825 Keicher et al. Apr 2009 B2
7534767 Butora et al. May 2009 B2
7547704 LaColla et al. Jun 2009 B2
7582618 Sommadossi et al. Sep 2009 B2
7598373 Storer et al. Oct 2009 B2
7608597 Sommadossi et al. Oct 2009 B2
7608599 Klumpp et al. Oct 2009 B2
7608600 Storer et al. Oct 2009 B2
7608601 Devos et al. Oct 2009 B2
7625875 Gosselin et al. Dec 2009 B2
7632821 Butora et al. Dec 2009 B2
7632940 Harrington et al. Dec 2009 B2
7635689 LaColla et al. Dec 2009 B2
7645745 Sarma Jan 2010 B2
7645747 Boojamra et al. Jan 2010 B2
7662798 LaColla et al. Feb 2010 B2
7666856 Johansson et al. Feb 2010 B2
7741334 Pottage Jun 2010 B2
7754699 Chun et al. Jul 2010 B2
7772208 Schinazi et al. Aug 2010 B2
7781576 Mayes et al. Aug 2010 B2
7790366 Houghton et al. Sep 2010 B1
7820631 McGuigan et al. Oct 2010 B2
7824851 Sommadossi et al. Nov 2010 B2
7842672 Boojamra et al. Nov 2010 B2
7871991 Boojamra et al. Jan 2011 B2
7879815 MacCoss et al. Feb 2011 B2
7902202 Sommadossi et al. Mar 2011 B2
7915232 Martin et al. Mar 2011 B2
7951787 McGuigan May 2011 B2
7951789 Sommadossi et al. May 2011 B2
7964580 Sofia et al. Jun 2011 B2
7973013 Cho et al. Jul 2011 B2
8008264 Butler et al. Aug 2011 B2
8012941 Cho et al. Sep 2011 B2
8012942 Butler et al. Sep 2011 B2
8022083 Boojamra et al. Sep 2011 B2
8071567 Devos et al. Dec 2011 B2
8119779 McGuigan et al. Feb 2012 B2
8148349 Meppen et al. Apr 2012 B2
8168583 Schinazi et al. May 2012 B2
8173621 Du et al. May 2012 B2
8183216 Di Francesco et al. May 2012 B2
8236779 Ma et al. Aug 2012 B2
8299038 Sommadossi et al. Oct 2012 B2
8318682 Butler et al. Nov 2012 B2
8318701 Boojamra et al. Nov 2012 B2
8324179 Chen et al. Dec 2012 B2
8329926 Boojamra et al. Dec 2012 B2
8334270 Sofia et al. Dec 2012 B2
8399428 Wagner Mar 2013 B2
8399429 Jonckers et al. Mar 2013 B2
8404651 Iyer et al. Mar 2013 B2
8415308 Cho et al. Apr 2013 B2
8415321 Schinazi et al. Apr 2013 B2
8415322 Clark Apr 2013 B2
8455451 Cho et al. Jun 2013 B2
8481510 Jonckers et al. Jul 2013 B2
8481713 Wang et al. Jul 2013 B2
8507460 Surleraux et al. Aug 2013 B2
8551973 Baa et al. Oct 2013 B2
8552021 Jonckers et al. Oct 2013 B2
8563530 Chang et al. Oct 2013 B2
8569478 Du et al. Oct 2013 B2
8575119 Wang et al. Nov 2013 B2
8580765 Sofia et al. Nov 2013 B2
8609627 Cho et al. Dec 2013 B2
8618076 Ross et al. Dec 2013 B2
8629263 Ross et al. Jan 2014 B2
8637475 Storer et al. Jan 2014 B1
8642756 Ross et al. Feb 2014 B2
8658616 McGuigan et al. Feb 2014 B2
8680071 Surleraux et al. Mar 2014 B2
8691788 Sommadossi et al. Apr 2014 B2
8716262 Sofia et al. May 2014 B2
8716263 Chun et al. May 2014 B2
8728725 Paul et al. May 2014 B2
8735372 Du et al. May 2014 B2
8759318 Chamberlain et al. Jun 2014 B2
8759510 Du et al. Jun 2014 B2
8765710 Sofia et al. Jul 2014 B2
8765935 Wagner Jul 2014 B2
8772474 Beigelman et al. Jul 2014 B2
8802840 Francom et al. Aug 2014 B2
8816074 Chu et al. Aug 2014 B2
8841275 Du et al. Sep 2014 B2
8859756 Ross et al. Oct 2014 B2
8871737 Smith et al. Oct 2014 B2
8877731 Beigelman et al. Nov 2014 B2
8877733 Cho et al. Nov 2014 B2
8933052 Jonckers et al. Jan 2015 B2
8980865 Wang et al. Mar 2015 B2
20030008841 Devos et al. Jan 2003 A1
20030087873 Stuyver et al. May 2003 A1
20030129712 Poechlauer et al. Jul 2003 A1
20030236216 Devos et al. Dec 2003 A1
20040002596 Hong et al. Jan 2004 A1
20040006002 Sommadossi et al. Jan 2004 A1
20040014108 Eldrup et al. Jan 2004 A1
20040110718 Devos et al. Jun 2004 A1
20040121980 Martin et al. Jun 2004 A1
20040181052 Sourena et al. Sep 2004 A1
20040209904 Dunn et al. Oct 2004 A1
20040229839 Babu et al. Nov 2004 A1
20040229840 Bhat et al. Nov 2004 A1
20040259934 Olsen et al. Dec 2004 A1
20040266723 Otto et al. Dec 2004 A1
20050009737 Clark Jan 2005 A1
20050009775 Howes et al. Jan 2005 A1
20050038240 Connolly et al. Feb 2005 A1
20050124532 Sommadossi et al. Jun 2005 A1
20060040890 Martin et al. Feb 2006 A1
20060040944 Gosselin et al. Feb 2006 A1
20060205685 Phiasivongsa et al. Sep 2006 A1
20060234962 Olsen et al. Oct 2006 A1
20060264389 Bhat et al. Nov 2006 A1
20070004669 Carroll et al. Jan 2007 A1
20070027065 LaColla et al. Feb 2007 A1
20070027104 LaColla et al. Feb 2007 A1
20070032449 LaColla et al. Feb 2007 A1
20070037735 Gosselin et al. Feb 2007 A1
20070042988 Klumpp et al. Feb 2007 A1
20070042990 Gosselin et al. Feb 2007 A1
20070060503 Gosselin et al. Mar 2007 A1
20070060504 Gosselin et al. Mar 2007 A1
20070060505 Gosselin et al. Mar 2007 A1
20070060541 Gosselin et al. Mar 2007 A1
20070265222 MacCoss et al. Nov 2007 A1
20080070861 Clark Mar 2008 A1
20080139802 Axt et al. Jun 2008 A1
20080207554 Beigelman et al. Aug 2008 A1
20080253995 Clark Oct 2008 A1
20080255038 Hopkins et al. Oct 2008 A1
20080261913 Sommadossi et al. Oct 2008 A1
20080280842 MacCoss et al. Nov 2008 A1
20080286230 Sommadossi et al. Nov 2008 A1
20090004135 Clark Jan 2009 A1
20090036666 Clark Feb 2009 A1
20090048189 Keicher et al. Feb 2009 A1
20090076062 Maibaum et al. Mar 2009 A1
20090118223 Erion et al. May 2009 A1
20090162292 Thompson et al. Jun 2009 A1
20090169504 Sommadossi et al. Jul 2009 A1
20090176732 Beigelman et al. Jul 2009 A1
20090181921 Blatt et al. Jul 2009 A1
20090238790 Sommadossi et al. Sep 2009 A2
20090306007 Wagner Dec 2009 A1
20090318380 Sofia et al. Dec 2009 A1
20100003217 Cretton-Scott et al. Jan 2010 A1
20100056468 Kotra et al. Mar 2010 A1
20100077085 Cohen Mar 2010 A1
20100081628 Du et al. Apr 2010 A1
20100151001 Schott et al. Jun 2010 A1
20100240604 Beigelman et al. Sep 2010 A1
20100249068 Beigelman et al. Sep 2010 A1
20100279969 Schinazi et al. Nov 2010 A1
20100279973 Chun et al. Nov 2010 A1
20100279974 Pierra et al. Nov 2010 A1
20100286083 Bao et al. Nov 2010 A1
20100297079 Almond et al. Nov 2010 A1
20100298257 Ross et al. Nov 2010 A1
20100316594 Sommadossi et al. Dec 2010 A1
20100331397 Beigelman et al. Dec 2010 A1
20110015146 Sofia et al. Jan 2011 A1
20110021454 Du et al. Jan 2011 A1
20110091943 Gallou et al. Apr 2011 A1
20110124592 McGuigan et al. May 2011 A1
20110150997 Shah et al. Jun 2011 A1
20110171192 Tomiyama et al. Jul 2011 A1
20110217261 Or et al. Sep 2011 A1
20110243886 Surleraux et al. Oct 2011 A1
20110244027 Chu et al. Oct 2011 A1
20110245484 Ross et al. Oct 2011 A1
20110251152 Ross et al. Oct 2011 A1
20110257121 Chang et al. Oct 2011 A1
20110269707 Stuyver et al. Nov 2011 A1
20110286962 Sommadossi et al. Nov 2011 A1
20110287927 Grasset et al. Nov 2011 A1
20110288308 Grasset et al. Nov 2011 A1
20110306541 Delaney, IV et al. Dec 2011 A1
20110306573 Avolio et al. Dec 2011 A1
20120010164 Surnma et al. Jan 2012 A1
20120034184 Devos et al. Feb 2012 A1
20120035115 Manoharan et al. Feb 2012 A1
20120040924 Cho et al. Feb 2012 A1
20120041184 Beigelman et al. Feb 2012 A1
20120052046 Chamberlain et al. Mar 2012 A1
20120070411 Beigelman et al. Mar 2012 A1
20120070415 Beigelman et al. Mar 2012 A1
20120071434 Smith et al. Mar 2012 A1
20120107274 Clarke et al. May 2012 A1
20120142626 Du et al. Jun 2012 A1
20120157511 Manoharan et al. Jun 2012 A1
20120165286 Beigelman et al. Jun 2012 A1
20120165515 Bhat et al. Jun 2012 A1
20120219568 Liu et al. Aug 2012 A1
20120225839 Jonckers et al. Sep 2012 A1
20120232029 Sofia et al. Sep 2012 A1
20120237480 Or et al. Sep 2012 A1
20120245335 Clark Sep 2012 A1
20120251487 Surleraux Oct 2012 A1
20120258928 Du et al. Oct 2012 A1
20120263678 Cho et al. Oct 2012 A1
20120316327 Chun et al. Dec 2012 A1
20130005677 Chu et al. Jan 2013 A1
20130017171 Sommadossi et al. Jan 2013 A1
20130029929 Sofia et al. Jan 2013 A1
20130064793 Surleraux et al. Mar 2013 A1
20130078217 Wang et al. Mar 2013 A1
20130137143 Gallou et al. May 2013 A1
20130149283 Sommadossi et al. Jun 2013 A1
20130164261 Wang et al. Jun 2013 A1
20130165400 Beigelman et al. Jun 2013 A1
20130203978 Wagner Aug 2013 A1
20130225520 Jonckers et al. Aug 2013 A1
20130244968 Jonckers et al. Sep 2013 A1
20130252920 Blatt et al. Sep 2013 A1
20130253181 Serebryany et al. Sep 2013 A1
20130273005 Delaney et al. Oct 2013 A1
20130281686 Cho et al. Oct 2013 A1
20130281687 Serebryany et al. Oct 2013 A1
20130315862 Sommadossi et al. Nov 2013 A1
20130315866 Parsy et al. Nov 2013 A1
20130315867 Parsy et al. Nov 2013 A1
20130315868 Mayes et al. Nov 2013 A1
20130323836 Manoharan et al. Dec 2013 A1
20130330297 Storer et al. Dec 2013 A1
20140045783 Du et al. Feb 2014 A1
20140057863 Stuyver et al. Feb 2014 A1
20140086873 Mayes et al. Mar 2014 A1
20140088117 Burch et al. Mar 2014 A1
20140099283 Gosselin et al. Apr 2014 A1
20140112886 Moussa et al. Apr 2014 A1
20140112887 Mayes et al. Apr 2014 A1
20140113880 Storer et al. Apr 2014 A1
20140128339 Girijavallabhan et al. May 2014 A1
20140140951 Moussa et al. May 2014 A1
20140140952 Moussa et al. May 2014 A1
20140140955 McGuigan et al. May 2014 A1
20140154211 Girijavallabhan et al. Jun 2014 A1
20140161770 Girijavallabhan et al. Jun 2014 A1
20140178338 Mayes et al. Jun 2014 A1
20140179627 Beigelman et al. Jun 2014 A1
20140205566 Liao et al. Jul 2014 A1
20140206640 Girijavallabhan et al. Jul 2014 A1
20140212382 Schinazi et al. Jul 2014 A1
20140219958 Luly et al. Aug 2014 A1
20140221304 Verma et al. Aug 2014 A1
20140235567 Verma et al. Aug 2014 A1
20140271547 Dukhan et al. Sep 2014 A1
20140288020 Du et al. Sep 2014 A1
20140294769 Mayes et al. Oct 2014 A1
20140303113 Krop et al. Oct 2014 A1
20140309164 Deshpande et al. Oct 2014 A1
20140309189 Deshpande et al. Oct 2014 A1
20140315850 Huang et al. Oct 2014 A1
20140315852 Du et al. Oct 2014 A1
20150018300 Du et al. Jan 2015 A1
Foreign Referenced Citations (159)
Number Date Country
5181998 Nov 1998 AU
2026131 Mar 1991 CA
2087967 Jan 1992 CA
2600359 Sep 2006 CA
1133642 Jan 2004 CN
103102345 May 2013 CN
103848876 Jun 2014 CN
103848877 Jun 2014 CN
4232852 Mar 1994 DE
19855963 Jun 2000 DE
20121305 Sep 2002 DE
1178049 Feb 2002 EP
1323830 Jul 2003 EP
1980568 Oct 2008 EP
2264169 Dec 2010 EP
2266579 Dec 2010 EP
2388069 Nov 2011 EP
2392580 Dec 2011 EP
2977586 Jan 2013 FR
167775 Dec 1990 IN
05069681 Mar 1993 JP
07242544 Sep 1995 JP
2008214305 Sep 2008 JP
WO 9316075 Aug 1993 WO
WO 1993017651 Sep 1993 WO
WO 9428715 Dec 1994 WO
WO 9508540 Mar 1995 WO
WO 9515332 Jun 1995 WO
WO 9726883 Jul 1997 WO
WO 9816184 Apr 1998 WO
WO 9816186 Apr 1998 WO
WO 9914226 Mar 1999 WO
WO 9961583 Dec 1999 WO
WO 2000034276 Jun 2000 WO
WO 2000066604 Nov 2000 WO
WO 2001068663 Sep 2001 WO
WO 2002003997 Jan 2002 WO
WO 2002057287 Jul 2002 WO
WO 2002057425 Jul 2002 WO
WO 2002100415 Dec 2002 WO
WO 2003022859 Mar 2003 WO
WO 2003026589 Apr 2003 WO
WO 2003039523 May 2003 WO
WO 2003048315 Jun 2003 WO
WO 2003068244 Aug 2003 WO
WO 2003073989 Sep 2003 WO
WO 2003087119 Oct 2003 WO
WO 2003099840 Dec 2003 WO
WO 2003105770 Dec 2003 WO
WO 2004002999 Jan 2004 WO
WO 2004003138 Jan 2004 WO
WO 2004007512 Jan 2004 WO
WO 2004014312 Feb 2004 WO
WO 2004037159 May 2004 WO
WO 2004080466 Sep 2004 WO
WO 2004091499 Oct 2004 WO
WO 2004106356 Dec 2004 WO
WO 2005007810 Jan 2005 WO
WO 2005009418 Feb 2005 WO
WO 2005020884 Mar 2005 WO
WO 2005020885 Mar 2005 WO
WO 2005021568 Mar 2005 WO
WO 2005034878 Apr 2005 WO
WO 2006000922 Jan 2006 WO
WO 2006012078 Feb 2006 WO
WO 2006044968 Apr 2006 WO
WO 2006063717 Jun 2006 WO
WO 2006094347 Sep 2006 WO
WO 2006105440 Oct 2006 WO
WO 2006116512 Nov 2006 WO
WO 2007027248 Mar 2007 WO
WO 2007113538 Oct 2007 WO
WO 2008012555 Jan 2008 WO
WO 2008054808 May 2008 WO
WO 2008089439 Jul 2008 WO
WO 2008095040 Aug 2008 WO
WO 2008117047 Oct 2008 WO
WO 2008121634 Oct 2008 WO
WO 2009001097 Dec 2008 WO
WO 2009003042 Dec 2008 WO
WO 2009010299 Jan 2009 WO
WO 2009040269 Apr 2009 WO
WO 2009058800 May 2009 WO
WO 2009067409 May 2009 WO
WO 2009086201 Jul 2009 WO
WO 2009105712 Aug 2009 WO
WO 2009129120 Oct 2009 WO
WO 2009152095 Dec 2009 WO
WO 2010002877 Jan 2010 WO
WO 2010026153 Mar 2010 WO
WO 2010027005 Mar 2010 WO
WO 2010075554 Jul 2010 WO
WO 2010084115 Jul 2010 WO
WO 2010088924 Aug 2010 WO
WO 2010089128 Aug 2010 WO
WO 2010091386 Aug 2010 WO
WO 2010108140 Sep 2010 WO
WO 2010130726 Nov 2010 WO
WO 2011005860 Jan 2011 WO
WO 2011029537 Mar 2011 WO
WO 2011057204 May 2011 WO
WO 2011119869 Sep 2011 WO
WO 2011133871 Oct 2011 WO
WO 2012012465 Jan 2012 WO
WO 2012012776 Jan 2012 WO
WO 2012025857 Mar 2012 WO
WO 2012041965 Apr 2012 WO
WO 2012048013 Apr 2012 WO
WO 2012062869 May 2012 WO
WO 2012062870 May 2012 WO
WO 2012074547 Jun 2012 WO
WO 2012075140 Jun 2012 WO
WO-2012075140 Jun 2012 WO
WO 2012092484 Jul 2012 WO
WO 2012094248 Jul 2012 WO
WO 2012099630 Jul 2012 WO
WO 2012142075 Oct 2012 WO
WO 2012142085 Oct 2012 WO
WO 2012142093 Oct 2012 WO
WO 2012142523 Oct 2012 WO
WO 2012158811 Nov 2012 WO
2012167133 Dec 2012 WO
WO 2013009735 Jan 2013 WO
WO 2013009737 Jan 2013 WO
WO 2013013009 Jan 2013 WO
WO 2013019874 Feb 2013 WO
WO 2013071169 May 2013 WO
WO 2013072466 May 2013 WO
WO 2013087765 Jun 2013 WO
WO 2013092447 Jun 2013 WO
WO 2013092481 Jun 2013 WO
WO 2013106344 Jul 2013 WO
WO 2013138210 Sep 2013 WO
WO 2013142124 Sep 2013 WO
WO 2013142159 Sep 2013 WO
WO 2013142525 Sep 2013 WO
WO 2013174962 Nov 2013 WO
WO 2013177219 Nov 2013 WO
WO 2014008236 Jan 2014 WO
WO 2014047117 Mar 2014 WO
WO 2014048532 Apr 2014 WO
WO 2014059901 Apr 2014 WO
WO 2014059902 Apr 2014 WO
WO 2014062596 Apr 2014 WO
WO 2014070771 May 2014 WO
WO 2014099941 Jun 2014 WO
WO 2014100498 Jun 2014 WO
WO 2014100505 Jun 2014 WO
WO 2014124430 Aug 2014 WO
WO 2014164533 Oct 2014 WO
WO 2014169278 Oct 2014 WO
WO 2014169280 Oct 2014 WO
WO 2014186637 Nov 2014 WO
WO 2014209979 Dec 2014 WO
WO 2014209983 Dec 2014 WO
WO 2015038596 Mar 2015 WO
WO 2015054465 Apr 2015 WO
WO 2015061683 Apr 2015 WO
WO 2015081133 Jun 2015 WO
Non-Patent Literature Citations (49)
Entry
Lohmann et al., “Replication of Subgenomic Hepatitis C Virus RNAs in a Hepatoma Cell Line”, Science, 285, pp. 110-113 (1999).
Kreiger et al., “Enhancement of Hepatitis C Virus RNA Replication by Cell Culture-Adaptive Mutations”, Journal of Virology 75, pp. 4614-4624 (2001).
Inoue et al., “Evaluation of a Cyclophilin Inhibitor in Hepatitis C Virus-Infected Chimeric Mice in Vivo”, Hepatology, 45(4), pp. 921-8 (2007).
Taneto et al., “Near Completely Humanized Liver in Mice Shows Human-Type Metabolic Responses to Drugs”, American Journal of Pathology, vol. 165, No. 3, pp. 901-912 (2004).
Versteeg et al., “Synthesis, structure, and sugar dynamics of a 2′-spiroisoxazolidine thymidine analog”, Tetrahedron, vol. 66, pp. 8145-8150 (2010).
Babu et al., “2′ -Spiro ribo- and arabinonucleosides: synthesis, molecular modelling and incorporation into oligodeoxynucleotides”, Organic and Biomolecular Chemistry, vol. 1 (20), pp. 3514-3526 (2003).
U.S. Appl. No. 11/005,443, La Colla et al., filed Dec. 6, 2004.
Lewis W, et al. “Mitochondrial toxicity of NRTI antiviral drugs: an integrated cellular perspective” Nature Reviews Drug Discovery (2003) 2:812-22.
Moyle G. “Clinical manifestations and management of antiretroviral nucleoside analog-related mitochondrial toxicity” Clin. Ther. (2000) 22(8):911-36.
Arnold et al.; “Sensitivity of Mitochondrial Transcription and Resistance of RNA Polymerase II Dependent Nuclear Transcription to Antiviral Ribonucleosides” PLOS | Pathogens (2012) 8(11) e1003030 (12 pages).
Lee et al.; “A Concise Synthesis of 4′-Fiuoro Nucleosides” Organic Letters (2007), 9(24), 5007-5009.
Owen et al.; “4′-Substituted nucleosides. Synthesis of some 4′-fluorouridine derivatives”, J. Org. Chem. (1976), 41(18), 3010-17.
Congiatu et al., Novel Potential Anticancer Naphthyl Phosphoramidates of BVdU: Separation of Diastereoisomers and Assignment of the Absolute Configuration of the Phosphorus Center (2006) Journal of Medicinal Chemistry 49:452-455.
Eldrup et al., Structure-Activity Relationship of Purine Ribonucleosides for Inhibition of Hepatitis C Virus RNA-Dependent RNA Polymerase (2004) Journal of Medicinal Chemistry 47:2283-2295.
Gardelli et al., Phosphoramidate Prodrugs of 2′-C-Methylcytidine for Therapy of Hepatitis C Virus Infection (2009) Journal of Medicinal Chemistry 52:5394-5407.
Hollecker et al., Synthesis of -enantiomers of N4 -hydroxy-3′-deoxypyrimidine nucleosides and their evaluation against bovine viral diarrhoea virus and hepatitis C virus in cell culture (2004) Antiviral Chemistry & Chemotherapy 14:43-55.
Ivanov et al., Synthesis and biological properties of pyrimidine 4′-fluoronucleosides and 4′-fluorouridine 5′-0-triphosphate (2010) Russian Journal of Bioorganic Chemistry 36:488-496.
King et al., Inhibition of the replication of a hepatitis C virus-like RNA template by interferon and 3′- deoxycytidine (2002) Antiviral Chemistry & Chemotherapy 13:363-370.
Leisvuori et al., Synthesis of 3′,5′-Cyclic Phosphate and Thiophosphate Esters of 2′-C-Methyl Ribonucleosides (2012) Helvetica Chimica Acta 95:1512-1520.
McGuigan et al., Phosphoramidate ProTides of 2′-C-Methylguanosine as Highly Potent Inhibitors of Hepatitis C Virus. Study of Their in Vitro and in Vivo Properties (2010) Journal of Medicinal Chemistry 53:4949-4957.
McGuigan et al., Phosphorodiamidates as a Promising New Phosphate Prodrug Motif for Antiviral Drug Discovery: Application to Anti-HCV Agents (2011) Journal of Medicinal Chemistry 54:8632-8645.
McGuigan et al., the application of phosphoramidate ProTide technology to the potent anti-HCV compound 4′-azidocytidine (R1479) (2009) Bioorganic & Medicinal Chemistry Letters 19:4250-4254.
Mehellou et al., Phosphoramidates of 2′- -D-arabinouridine (AraU) as phosphate prodrugs; design, synthesis, in vitro activity and metabolism (2010) Bioorganic & Medicinal Chemistry 18:2439-2446.
Mehellou et al., the design, synthesis and antiviral evaluation of a series of 5-trimethylsilyl-1- -o- (arabinofurano syl)uracil phosphoramidate ProTides (2010) Antiviral Chemistry & Chemotherapy 20:153-160.
Murakami et al., Mechanism of Activation of PSI-7851and Its Diastereoisomer PSI-7977 (2010) Journal of Biological Chemistry 285:34337-34347.
Murakami et al., Mechanism of Activation of -D-2′-Deoxy-2′-Fiuoro-2′-C-Methylcytidine and Inhibition of Hepatitis C Virus NSSB RNA polymerase (2007) Antimicrobial Agents and Chemotherapy 51:503-509.
Olsen et al., A 7-Deaza-Adenosine Analog Is a Potent and Selective Inhibitor of Hepatitis C Virus Replication with Excellent Pharmacokinetic Properties (2004) Antimicrobial Agents and Chemotherapy 28:3944-3953.
Perrone et al., Application of the Phosphoramidate ProTide Approach to 4′-Azidouridine Confers Sub-micromolar Potency versus Hepatitis C Virus on an Inactive Nucleoside (2007) Journal of Medicinal Chemistry 50:1840-1849.
Prakash et al., Synthesis and Evaluation of 5-Acyl-2-thioethyl Esters of Modified Nucleoside 5′Monophosphates as Inhibitors of Hepatitis C Virus RNA Replication (2005) J. Med. Chem. 48:1199-1210.
Saboulard et al., Characterization of the Activation Pathway of Phosphoramidate Triester Prodrugs of Stavudine and Zidovudine (2009) Molecular Pharmacology 56:693-704.
Shen et al., Design and synthesis of vidarabine prodrugs as antiviral agents (2009) Bioorganic & Medicinal Chemistry Letters 19:792-796.
Sofia et al., Discovery of a 6-o-2′-Deoxy-2′-a-fluoro-2'-6-C-methyluridine Nucleotide Prodrug (PSI-7977) for the Treatment of Hepatitis CVirus (2010) Journal of Medicinal Chemistry 53:7202-7218.
Stein et al., Phosphorylation of Nucleoside Analog Antiretrovirals: A Review for Clinicians (2001) Pharmacotherapy 21:11-34.
Tomassini et al., Inhibitory Effect of 2′-Substituted Nucleosides on Hepatitis C Virus Replication Correlates with Metabolic Properties in Replicon Cells (2005) Antimicrobial Agents and Chemotherapy 49:2050-2058.
Cahard et al., Aryloxy Phosphoramidate Triesters as Pro-Tides (2004) Mini-Reviews in Medicinal Chemistry 4:371-381.
Kakefuda et al., Nucleosides and nucleotides. 120. Stereoselective radical deoxygenation of tert- alcohols in the sugar moiety of nucleosides: synthesis of 2′,3′-dideoxy-2′-C-methyl- -2′-C-ethynyl- -d- threo-pentofuranosyl pyrimidines and adenine as potential antiviral and antitumor agents (1993) Tetrahedron 49:8513-8528.
Kawana et al., The deoxygenations of tosylated adenosine derivatives with Grignard reagents (1986) Nucleic Acids Symp Ser. 17:37-40.
Kawana et al., The Synthesis of C-Methyl Branched-Chain Deoxy Sugar Nucleosides by the Deoxygenative Methylation of 0-Tosylated Adenosines with Grignard Reagents (1988) Bull. Chem. Soc. Jpn. 61:2437-2442.
Madela et al., Progress in the development of anti-hepatitis C virus nucleoside and nucleotide prodrugs (2012) Future Med. Chem. 4:625-650.
Pierra et al., Synthesis of 2′-C-Methylcytidine and 2′-C-Methyluridine Derivatives Modified in the 3′-Position as Potential Antiviral Agents (2006) Collection of Czechoslovak Chemical Communications 71:991-1010.
Tong et al., Nucleosides of thioguanine and other 2-amino-6-substituted purines from 2-acetamido-5- chloropurine (1967) J Org Chem. 32:859-62.
Vernachio et al., INX-08189, a Phosphoramidate Prodrug of 6-0-Methyi-2′-C-Methyl Guanosine, Is a Potent Inhibitor of Hepatitis C Virus Replication with Excellent Pharmacokinetic and Pharmacodynamic Properties (2011) Antimicrobial Agents and Chemotherapy 55:1843-1851.
Dang, Q., et al., “Syntheses of Nucleosides with 2′-Spirolactam and 2′-Spiropyrroliine Moieties as Potential Inhibitors of Hepatitis C Virus NS5B Polymerase”, Tetrahedron Letters, vol. 55, pp. 3813-3816 (2014).
Du, J., et al., “Use of 2′-Spirocyclic Ethers in HCV Nucleoside Design”, Journal of Medicinal Chemistry, vol. 57, pp. 1826-1835 (2014).
Jonckers, T., et al., “Nucleotide Prodrugs of 2′-Dooxy-2′-Spirooxetane Ribonucleosides as Novel Inhibitors of the HCV NS5 Polymerase”, Journal of Medicinal Chemistry, vol. 57, pp. 1836-1844 (2014).
Zheng, Y., et al., “The Use of Spirocyclic Scaffolds in Drug Discovery”, Bioorganic & Medicinal Chemistry Letters, vol. 24, pp. 3673-3682 (2014).
International search report dated Jul. 9, 2013, for corresponding international application PCT/EP2013/060704.
European search report dated Aug. 13, 2012, for corresponding European application 12169425.1.
Jonckers, T., et al., “Discovery of 1- ((2R,4aR,6R,7R,7aR)- 2-Isopropoxy-2- oxidodihydro- 4H,6H- spiro[furo[3,2- d][1,3,2]dioxaphosphinine-7,2'- oxetan]-6-yl)pyrimidine-2,4(1H,3H)- dione (JNJ-54257099), a 3′-5′- Cyclic Phosphate Ester Prodrug of 2′-Deoxy-2′-Spirooxetane Uridine Triphosphate Useful for HCV Inhibition”, J. of Medicinal Chemistry, vol. 59, pp. 5790-5798 (2016).
Related Publications (1)
Number Date Country
20190092804 A1 Mar 2019 US
Continuations (5)
Number Date Country
Parent 16044262 Jul 2018 US
Child 16212554 US
Parent 15809730 Nov 2017 US
Child 16044262 US
Parent 15423985 Feb 2017 US
Child 15809730 US
Parent 15238553 Aug 2016 US
Child 15423985 US
Parent 14403587 US
Child 15238553 US