WO2016105547A1 - Deuterated dasabuvir - Google Patents

Deuterated dasabuvir Download PDF

Info

Publication number
WO2016105547A1
WO2016105547A1 PCT/US2015/000409 US2015000409W WO2016105547A1 WO 2016105547 A1 WO2016105547 A1 WO 2016105547A1 US 2015000409 W US2015000409 W US 2015000409W WO 2016105547 A1 WO2016105547 A1 WO 2016105547A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
deuterium
pharmaceutical composition
inhibitor
compounds
Prior art date
Application number
PCT/US2015/000409
Other languages
French (fr)
Inventor
I Robert SILVERMAN
Original Assignee
Concert Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Concert Pharmaceuticals, Inc. filed Critical Concert Pharmaceuticals, Inc.
Publication of WO2016105547A1 publication Critical patent/WO2016105547A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/52Two oxygen atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B59/00Introduction of isotopes of elements into organic compounds ; Labelled organic compounds per se
    • C07B59/002Heterocyclic compounds

Definitions

  • ADME absorption, distribution, metabolism and/or excretion
  • ADME limitation that affects many medicines is the formation of toxic or biologically reactive metabolites.
  • some patients receiving the drug may experience toxicities, or the safe dosing of such drugs may be limited such that patients receive a suboptimal amount of the active agent.
  • modifying dosing intervals or formulation approaches can help to reduce clinical adverse effects, but often the formation of such undesirable metabolites is intrinsic to the metabolism of the compound.
  • a metabolic inhibitor will be co-administered with a drug that is cleared too rapidly.
  • a drug that is cleared too rapidly.
  • the FDA recommends that these drugs be co- dosed with ritonavir, an inhibitor of cytochrome P450 enzyme 3A4 (CYP3A4), the enzyme typically responsible for their metabolism (see Kempf, D.J. et al.,
  • a potentially attractive strategy for improving a drug's metabolic properties is deuterium modification.
  • Deuterium is a safe, stable, non-radioactive isotope of hydrogen. Compared to hydrogen, deuterium forms stronger bonds with carbon. In select cases, the increased bond strength imparted by deuterium can positively impact the ADME properties of a drug, creating the potential for improved drug efficacy, safety, and/or tolerability.
  • the size and shape of deuterium are essentially identical to those of hydrogen,
  • biochemical potency and selectivity of the drug as compared to the original chemical entity that contains only hydrogen.
  • Dasabuvir also known as ExvieraTM, ABT-333 and N-[6-[3-tert-Butyl-5-(2,4- dioxo-3,4-dihydropyrimidin-l(2H)-yl)-2-methoxyphenyl]naphthalen-2- yljmethanesulfonamide, is a non-nucleoside hepatitis C virus NS5B palm polymerase inhibitor.
  • This invention relates to novel deuterated pyrimidinedione derivatives, and pharmaceutically acceptable salts thereof.
  • This invention also provides compositions comprising a compound of this invention and the use of such compositions in methods of treating hepatitis C viral infections.
  • the invention also provides novel intermediates useful in the synthesis of the compounds of the invention. Such intermediates may also have hepatitis C virus inhibitory properties.
  • treat means decrease, suppress, attenuate, diminish, arrest, or stabilize the development or progression of a disease (e.g., a disease or disorder delineated herein), lessen the severity of the disease or improve the symptoms associated with the disease.
  • a disease e.g., a disease or disorder delineated herein
  • Disease means any condition or disorder that damages or interferes with the normal function of a cell, tissue, or organ.
  • any atom not specifically designated as a particular isotope is meant to represent any stable isotope of that atom.
  • a position is designated specifically as “H” or “hydrogen”
  • the position is understood to have hydrogen at its natural abundance isotopic composition.
  • a position is designated specifically as “D” or “deuterium”
  • the position is understood to have deuterium at an abundance that is at least 3340 times greater than the natural abundance of deuterium, which is 0.015% (i.e., at least 50.1 % incorporation of deuterium).
  • isotopic enrichment factor means the ratio between the isotopic abundance and the natural abundance of a specified isotope.
  • a compound of this invention has an isotopic enrichment factor for each designated deuterium atom of at least 3500 (52.5% deuterium incorporation at each designated deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium), at least 5500 (82.5% deuterium incorporation), at least 6000 (90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5% deuterium incorporation).
  • isotopologue refers to a species in which the chemical structure differs from a specific compound of this invention only in the isotopic composition thereof.
  • a compound represented by a particular chemical structure containing indicated deuterium atoms will also contain lesser amounts of isotopologues having hydrogen atoms at one or more of the designated deuterium positions in that structure.
  • the relative amount of such isotopologues in a compound of this invention will depend upon a number of factors including the isotopic purity of deuterated reagents used to make the compound and the efficiency of incorporation of deuterium in the various synthesis steps used to prepare the compound.
  • the relative amount of such isotopologues in toto will be less than 49.9% of the compound. In other embodiments, the relative amount of such isotopologues in toto will be less than 47.5%, less than 40%, less than 32.5%, less than 25%, less than 17.5%, less than 10%, less than 5%, less than 3%, less than 1%), or less than 0.5% of the compound.
  • the invention also provides salts of the compounds of the invention.
  • a salt of a compound of this invention is formed between an acid and a basic group of the compound, such as an amino functional group, or a base and an acidic group of the compound, such as a carboxyl functional group.
  • the compound is a pharmaceutically acceptable acid addition salt.
  • pharmaceutically acceptable refers to a component that is, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and other mammals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable salt means any non-toxic salt that, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention.
  • pharmaceutically acceptable counterion is an ionic portion of a salt that is not toxic when released from the salt upon administration to a recipient.
  • Acids commonly employed to form pharmaceutically acceptable salts include inorganic acids such as hydrogen bisulfide, hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid and phosphoric acid, as well as organic acids such as para-toluenesulfonic acid, salicylic acid, tartaric acid, bitartaric acid, ascorbic acid, maleic acid, besylic acid, fumaric acid, gluconic acid, glucuronic acid, formic acid, glutamic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, lactic acid, oxalic acid, para-bromophenylsulfonic acid, carbonic acid, succinic acid, citric acid, benzoic acid and acetic acid, as well as related inorganic and organic acids.
  • inorganic acids such as hydrogen bisulfide, hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid and phosphoric acid
  • Such pharmaceutically acceptable salts thus include sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, phosphate, monohydrogenphosphate, dihydrogenphosphate, metaphosphate, pyrophosphate, chloride, bromide, iodide, acetate, propionate, decanoate, caprylate, acrylate, formate, isobutyrate, caprate, heptanoate, propiolate, oxalate, malonate, succinate, suberate, sebacate, fumarate, maleate, butyne- 1 ,4-dioate, hexyne-l,6-dioate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, phthalate, terephthalate, sulfonate, xylene sulfonate, phenylacetate, phenyl
  • the pharmaceutically acceptable salt may also be a salt of a compound of the present invention having an acidic functional group, such as a carboxylic acid functional group, and a base.
  • exemplary bases include, but are not limited to, hydroxide of alkali metals including sodium, potassium, and lithium; hydroxides of alkaline earth metals such as calcium and magnesium; hydroxides of other metals, such as aluminum and zinc; ammonia, organic amines such as unsubstituted or hydroxyl-substituted mono-, di-, or tri-alkylamines, dicyclohexylamine; tributyl amine; pyridine; N-methyl, N-ethylamine; diethylamine; triethylamine; mono-, bis-, or tris-(2-OH-(Ci-C 6 )-alkylamine), such as N,N-dimethyl-N-(2-hydroxyethyl)amine or tri-(2-hydroxyethyl)amine; N-methyl
  • the pharmaceutically acceptable salt is a sodium salt, more specifically a monosodium salt.
  • the compounds of the present invention may contain an asymmetric carbon atom, for example, as the result of deuterium
  • compounds of this invention can exist as either individual enantiomers, or mixtures of the two enantiomers. Accordingly, a compound of the present invention may exist as either a racemic mixture or a scalemic mixture, or as individual respective stereoisomers that are substantially free from another possible stereoisomer. The term "substantially free of other
  • stereoisomers as used herein means less than 25% of other stereoisomers, preferably less than 10% of other stereoisomers, more preferably less than 5% of other stereoisomers and most preferably less than 2% of other stereoisomers are present.
  • Methods of obtaining or synthesizing an individual enantiomer for a given compound are known in the art and may be applied as practicable to final compounds or to starting material or intermediates.
  • stable compounds refers to compounds which possess stability sufficient to allow for their manufacture and which maintain the integrity of the compound for a sufficient period of time to be useful for the purposes detailed herein (e.g., formulation into therapeutic products, intermediates for use in production of therapeutic compounds, isolatable or storable intermediate compounds, treating a disease or condition responsive to therapeutic agents).
  • Substituted with deuterium refers to the replacement of one or more hydrogen atoms with a corresponding number of deuterium atoms.
  • each R may be referred to specifically (e.g., R 1 , R 2 , R 3 , etc.). Unless otherwise indicated, when a variable is referred to generally, it is meant to include all specific embodiments of that particular variable.
  • R 1 is a t-butyl moiety optionally comprising between 1 and 9 deuterium substitutions
  • each R 2 and R 3 is independently selected from -CH 3 , -CH 2 D, -CHD 2 and -CD 3 ;
  • each of Y', Y 2 , Y 3 , Y 4 , Y 5 , Y 6 , Y 7 , Y 8 and Y 9 is independently selected from hydrogen and deuterium
  • R 1 , R 2 and R 3 comprises a deuterium atom.
  • R 1 is selected from -C(CH 3 )3 and -C(CD 3 ) 3 . In one aspect of these embodiments, R 1 is -C(CH 3 ) 3 . In an alternate aspect of these embodiments, R 1 is -C(CD 3 ) 3 .
  • each of R 2 and R 3 are independently selected from -CH 3 and -CD 3 .
  • R 2 is -CD 3 and R 3 is -CH 3 .
  • R 2 is -CH 3 and R 3 is -CD 3 . In still another aspect of these embodiments, R 2 is -CD 3 and R 3 is -CD 3 . In still another aspect of these embodiments, R 2 is -CH 3 and R 3 is -CH 3 .
  • Y 1 and Y 2 are the same.
  • Y 4 , Y 5 , Y 6 , Y 7 , Y 8 and Y 9 are the same.
  • Y 1 and Y 2 are the same.
  • Y 1 and Y 2 are the same; R 1 is selected from -C(CH 3 ) 3 and -C(CD 3 ) 3 and each of R 2 and R 3 are independently selected from -CH 3 and -CD 3 .
  • each of Y 1 , Y 2 , Y 3 , Y 4 , Y 3 , Y 6 , Y 7 , Y 8 and Y 9 is hydrogen;
  • R 1 is selected from -C(CH 3 ) 3 and -C(CD 3 ) 3 and each of R 2 and R 3 are independently selected from -CH 3 and -CD 3 .
  • R 1 is -C(CD 3 ) 3
  • R 2 is -CD 3
  • R 3 is CD 3 .
  • R 1 is -C(CD 3 ) 3
  • R 2 is -CH 3 and R 3 is CD 3
  • R 1 is -C(CD 3 )3
  • R 2 is -CD 3 and R 3 is CH 3 .
  • R 1 is -C(CD 3 )3
  • R 2 is -CH 3
  • R 3 is CH 3 .
  • R 1 is -C(CH 3 ) 3
  • R 2 is -CD 3
  • R 3 is CD 3 .
  • R 1 is -C(CH 3 ) 3
  • R 2 is -CH 3
  • R 3 is CD 3 .
  • R 1 is -C(CH 3 ) 3
  • R 2 is -CD 3
  • R 3 is CH 3 .
  • the invention does not include a compound wherein R 1 is -C(CD 3 ) 3 , each of R 2 and R 3 is -CD 3 ; and each of
  • Y 1 , Y 2 , Y 3 , Y 4 , Y 5 , Y 6 , Y 7 , Y 8 and Y 9 is deuterium.
  • Y 1 and Y 2 are the same; Y 4 , Y 3 , Y 6 , Y 7 , Y 8 and Y 9 are the same; and the compound is selected from any one of the compounds (Cmpd) set forth in Table 1 (below):
  • any atom not designated as deuterium in any of the embodiments set forth above is present at its natural isotopic abundance.
  • Reagents and conditions (a) Nal, NaOH, NaOCl; (b) (3): (CD 3 )2S0 4 , K 2 C0 3 or CD3-I, NaOH; (c) K3PO4, Cul, Sodium ascorbate, N-(2-cyanophenyl)picolinamide or N-(2- cyanophenyl)benzamide; (d)K3P04, Pd2dba 3 , l,3,5,7-Tetramethyl-6-phenyl-2,4,8-trioxa-6- phosphaadamantane
  • Reagents and conditions (a) Br 2 , Ph 3 P; (b) acetyl chloride, A1C1 3 , NaOCl; (c) DPPA; (d) CD 3 S0 2 C1; (e) bis(pinacolato)diboron, KOAc, combiPhos-Pd6 catalyst
  • compositions comprising an effective amount of a compound of Formula I (e.g., including any of the formulae herein), or a pharmaceutically acceptable salt of said compound; and a
  • the carrier(s) are "acceptable” in the sense of being compatible with the other ingredients of the formulation and, in the case of a pharmaceutically acceptable carrier, not deleterious to the recipient thereof in an amount used in the medicament.
  • Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene- polyoxypropylene-block polymers, polyethylene glycol and wool fat.
  • ion exchangers alumina, aluminum stearate, lecithin
  • serum proteins such as human serum albumin
  • buffer substances such as phosphate
  • the solubility and bioavailability of the compounds of the present invention in pharmaceutical compositions may be enhanced by methods well-known in the art.
  • One method includes the use of lipid excipients in the formulation. See “Oral Lipid-Based Formulations: Enhancing the Bioavailability of Poorly Water- Soluble Drugs (Drugs and the Pharmaceutical Sciences),” David J. Hauss, ed. Informa Healthcare, 2007; and “Role of Lipid Excipients in Modifying Oral and Parenteral Drug Delivery: Basic Principles and Biological Examples," Kishor M. Wasan, ed. Wiley-Interscience, 2006.
  • Another known method of enhancing bioavailability is the use of an amorphous form of a compound of this invention optionally formulated with a poloxamer, such as LUTROLTM and PLURONICTM (BASF Corporation), or block copolymers of ethylene oxide and propylene oxide. See United States patent
  • the bioavailability enhancing agent is one or more polymers selected from copovidone, polyvinylpyrrolidone,
  • the bioavailability enhancing agent comprises copovidone.
  • compositions of the invention include those suitable for oral, rectal, nasal, topical (including buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration.
  • the compound of the formulae herein is administered transdermally (e.g., using a transdermal patch or iontophoretic techniques).
  • Other formulations may conveniently be presented in unit dosage form, e.g., tablets, sustained release capsules, and in liposomes, and may be prepared by any methods well known in the art of pharmacy. See, for example, Remington: The Science and Practice of Pharmacy, Lippincott Williams & Wilkins, Baltimore, MD (20th ed. 2000).
  • Such preparative methods include the step of bringing into association with the molecule to be administered ingredients such as the carrier that constitutes one or more accessory ingredients.
  • ingredients such as the carrier that constitutes one or more accessory ingredients.
  • the compositions are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers, liposomes or finely divided solid carriers, or both, and then, if necessary, shaping the product.
  • compositions of the present invention suitable for oral administration may be presented as discrete units such as capsules, sachets, or tablets each containing a predetermined amount of the active ingredient; a powder or granules; a solution or a suspension in an aqueous liquid or a non-aqueous liquid; an oil-in-water liquid emulsion; a water-in-oil liquid emulsion; packed in liposomes; or as a bolus, etc.
  • Soft gelatin capsules can be useful for containing such suspensions, which may beneficially increase the rate of compound absorption.
  • the oral composition is a tablet.
  • carriers that are commonly used include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried cornstarch.
  • aqueous suspensions are administered orally, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavoring and/or coloring agents may be added.
  • compositions suitable for oral administration include lozenges comprising the ingredients in a flavored basis, usually sucrose and acacia or tragacanth; and pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia.
  • compositions suitable for parenteral administration include aqueous .and nonaqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • the formulations may be presented in unit-dose or multi-dose containers, for example, sealed ampules and vials, and may be stored in a freeze dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets.
  • Such injection solutions may be in the form, for example, of a sterile injectable aqueous or oleaginous suspension.
  • This suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example, as a solution in 1,3- butanediol.
  • the acceptable vehicles and solvents that may be employed are mannitol, water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant.
  • compositions of this invention may be administered in the form of suppositories for rectal administration.
  • These compositions can be prepared by mixing a compound of this invention with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the active components.
  • suitable non-irritating excipient include, but are not limited to, cocoa butter, beeswax and polyethylene glycols.
  • compositions of this invention may be administered by nasal aerosol or inhalation.
  • Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizing or dispersing agents known in the art. See, e.g.: Rabinowitz JD and Zaffaroni AC, US Patent 6,803,031, assigned to Alexza Molecular Delivery Corporation.
  • Topical administration of the pharmaceutical compositions of this invention is especially useful when the desired treatment involves areas or organs readily accessible by topical application.
  • the pharmaceutical composition should be formulated with a suitable ointment containing the active components suspended or dissolved in a carrier.
  • Carriers for topical administration of the compounds of this invention include, but are not limited to, mineral oil, liquid petroleum, white petroleum, propylene glycol, polyoxyethylene polyoxypropylene compound, emulsifying wax, and water.
  • the pharmaceutical composition can be formulated with a suitable lotion or cream containing the active compound suspended or dissolved in a carrier.
  • Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol, and water.
  • the pharmaceutical compositions of this invention may also be topically applied to the lower intestinal tract by rectal suppository formulation or in a suitable enema formulation. Topically-transdermal patches and iontophoretic administration are also included in this invention.
  • a composition of this invention further comprises a second therapeutic agent.
  • the second therapeutic agent may be selected from any compound or therapeutic agent known to have or that demonstrates advantageous properties for the treatment of HCV infection.
  • the second therapeutic agent is an agent selected from a HCV NS3/NS4A protease inhibitor, a HCV NS5A inhibitor, a cytochrome P450 inhibitor, a second HCV NS5B polymerase inhibitor, and a guanosine analog.
  • the HCV NS3 NS4A protease inhibitor is paritaprevir.
  • the HCV NS5A inhibitor is ombitasvir.
  • the cytochrome P450 inhibitor is ritonavir.
  • the guanosine analog is ribavirin.
  • the invention provides separate dosage forms of a compound of this invention and one or more of any of the above-described second therapeutic agents, wherein the compound and second therapeutic agent are associated with one another.
  • the term "associated with one another" as used herein means that the separate dosage forms are packaged together or otherwise attached to one another such that it is readily apparent that the separate dosage forms are intended to be sold and administered together (within less than 24 hours of one another, consecutively or simultaneously).
  • the compound of the invention is packaged together with a HCV NS3/NS4A protease inhibitor, a HCV NS5A inhibitor, and a cytochrome P450 inhibitor.
  • the HCV NS3/NS4A protease inhibitor, HCV NS5A inhibitor, and cytochrome P450 inhibitor are co-formulated into a single dosage form and packaged with the compound of the invention in a separate dosage form.
  • the compound of the invention is a single dosage form packaged together with a separate single dosage form comprising paritaprevir, ombitasvir and ritonavir.
  • the compound of the invention is a single dosage form packaged together with a separate single dosage form comprising paritaprevir and ombitasvir, but lacking ritonavir or another cytochrome P450 inhibitor.
  • the invention provides a packaged pharmaceutical composition comprising two separate, but identical oral dosage forms of a compound of the invention; a third separate oral dosage form comprising a combination of paritaprevir, ombitasvir and ritonavir; and instructions for when during the day to take each of the first, second and third dosage forms.
  • the compound of the present invention is present in an effective amount.
  • the term is a pharmaceutical composition of the invention.
  • an “effective amount” refers to an amount which, when administered in a proper dosing regimen, is sufficient to treat the target disorder.
  • an effective amount of a compound of this invention for oral dosing to an adult can range from about 0.01 mg to about 1000 mg, specifically from about 25 mg to about 600 mg, from about 50 mg to about 600 mg, from about 25 mg to 500 mg, from about 50 mg to 500 mg, from about 25 mg to 400 mg, and from about 50 mg to 400 mg per day.
  • the daily dosage may be administered as single dose or in divided doses, e.g., two doses per day.
  • compositions that comprise a second therapeutic agent an effective amount of the second therapeutic agent is between about 20% and 100% of the dosage normally utilized in a monotherapy regime using just that agent.
  • an effective amount is between about 70% and 100% of the normal monotherapeutic dose.
  • the normal monotherapeutic dosages of these second therapeutic agents are well known in the art. See, e.g., Wells et al., eds.,
  • the invention provides a method of inhibiting the activity of HCV NS5B polymerase in a cell infected with HCV, comprising contacting a cell with one or more compounds of Formula I herein, or a pharmaceutically acceptable salt thereof.
  • the invention provides a method of treating a HCV infection.
  • the above method of treatment comprises the further step of co-administering to the subject in need thereof one or more second therapeutic agents.
  • the choice of second therapeutic agents may be made from any second therapeutic agent known to be useful to treat HCV infections.
  • the second therapeutic agent is one of more agents selected from a HCV NS3/NS4A protease inhibitor, a HCV NS5A inhibitor, a cytochrome P450 inhibitor, a second HCV NS5B polymerase inhibitor, and a guanosine analog.
  • the HCV NS3 S4A protease inhibitor is paritaprevir.
  • the HCV NS5A inhibitor is ombitasvir.
  • the cytochrome P450 inhibitor is ritonavir.
  • the guanosine analog is ribavirin.
  • the second therapeutic agents are a combination of a HCV NS3 NS4A protease inhibitor, a HCV NS5A inhibitor, and, optionally, a cytochrome P450 inhibitor.
  • the second therapeutic agents are a combination of paritaprevir, ombitasvir, and, optionally, ritonavir.
  • the second therapeutic agents are a combination of paritaprevir, ombitasvir, optionally ritonavir, and optionally ribavirin.
  • co-administered means that the second therapeutic agent may be administered together with a compound of this invention as part of a single dosage form (such as a composition of this invention comprising a compound of the invention and an second therapeutic agent as described above) or as separate, multiple dosage forms. Alternatively, the additional agent may be administered prior to, consecutively with, or following the administration of a compound of this invention. In such combination therapy treatment, both the compounds of this invention and the second therapeutic agent(s) are administered by conventional methods.
  • composition of this invention comprising both a compound of the invention and a second therapeutic agent
  • administration of a composition of this invention does not preclude the separate administration of that same therapeutic agent, any other second therapeutic agent or any compound of this invention to said subject at another time during a course of treatment.
  • not all doses of a compound of the invention administered each day need be co-administered with the one or more second therapeutic agent.
  • a compound of the invention may be administered twice a day, but only co-administered with one or more second therapeutic agents (e.g., a NS3/4A protease inhibitor (e.g., ombitasvir), a NS5A polymerase inhibitor (paritaprevir) and a cytochrome P450 inhibitor (e.g., ritonavir)) in one of the two dosings.
  • a second therapeutic agents e.g., a NS3/4A protease inhibitor (e.g., ombitasvir), a NS5A polymerase inhibitor (paritaprevir) and a cytochrome P450 inhibitor (e.g., ritonavir)
  • the effective amount of the compound of this invention is less than its effective amount would be where the second therapeutic agent is not administered. In another embodiment, the effective amount of the second therapeutic agent is less than its effective amount would be where the compound of this invention is not administered. In this way, undesired side effects associated with high doses of either agent may be minimized. Other potential advantages (including without limitation improved dosing regimens and/or reduced drug cost) will be apparent to those of skill in the art.
  • the invention provides the use of a compound of Formula I alone or together with one or more of the above-described second therapeutic agents in the manufacture of a medicament, either as a single composition or as separate dosage forms, for treatment in a subject of a disease, disorder or symptom set forth above.
  • Another aspect of the invention is a compound of Formula I for use in the treatment in a subject of a disease, disorder or symptom thereof delineated herein.
  • Microsomal Assay Human liver microsomes (20 mg/mL) are obtained from Xenotech, LLC (Lenexa, KS). ⁇ -nicotinamide adenine dinucleotide phosphate, reduced form (NADPH), magnesium chloride (MgCb), and dimethyl sulfoxide (DMSO) are purchased from Sigma-Aldrich.
  • 7.5 mM stock solutions of test compounds are prepared in DMSO.
  • the 7.5 mM stock solutions are diluted to 12.5- 50 ⁇ in acetonitrile (ACN).
  • ACN acetonitrile
  • the 20 mg/mL human liver microsomes are diluted to 0.625 mg/mL in 0.1 M potassium phosphate buffer, pH 7.4, containing 3 mM MgCl 2 .
  • the diluted microsomes are added to wells of a 96-well deep-well polypropylene plate in triplicate.
  • a 10 aliquot of the 12.5-50 ⁇ test compound is added to the microsomes and the mixture is pre-warmed for 10 minutes. Reactions are initiated by addition of pre-warmed NADPH solution.
  • the final reaction volume is 0.5 mL and contains 0.5 mg/mL human liver microsomes, 0.25-1.0 ⁇ test compound, and 2 mM NADPH in 0.1 M potassium phosphate buffer, pH 7.4, and 3 mM MgCl 2 .
  • the reaction mixtures are incubated at 37 °C, and 50 ⁇ ⁇ aliquots are removed at 0, 5, 10, 20, and 30 minutes and added to shallow-well 96-well plates which contain 50 ⁇ , of ice-cold ACN with internal standard to stop the reactions.
  • the plates are stored at 4 °C for 20 minutes after which 100 ⁇ L of water is added to the wells of the plate before centrifugation to pellet precipitated proteins.

Landscapes

  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Virology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Medicinal Chemistry (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • AIDS & HIV (AREA)
  • General Chemical & Material Sciences (AREA)
  • Molecular Biology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

This invention relates to novel deuterated pyrimidinedione derivatives of Formula (I), and pharmaceutically acceptable salts thereof. This invention also provides compositions comprising a compound of this invention and the use of such compositions in methods of treating hepatitis C viral infections. The invention also provides novel intermediates useful in the synthesis of the compounds of the invention. Such intermediates may also have hepatitis C virus inhibitory properties.

Description

DEUTERATED DASABUVIR
BACKGROUND OF THE INVENTION
[1] Many current medicines suffer from poor absorption, distribution, metabolism and/or excretion (ADME) properties that prevent their wider use or limit their use in certain indications. Poor ADME properties are also a major reason for the failure of drug candidates in clinical trials. While formulation technologies and prodrug strategies can be employed in some cases to improve certain ADME properties, these approaches often fail to address the underlying ADME problems that exist for many drugs and drug candidates. One such problem is rapid metabolism that causes a number of drugs, which otherwise would be highly effective in treating a disease, to be cleared too rapidly from the body. A possible solution to rapid drug clearance is frequent or high dosing to attain a sufficiently high plasma level of drug. This, however, introduces a number of potential treatment problems such as poor patient compliance with the dosing regimen, side effects that become more acute with higher doses, and increased cost of treatment. A rapidly metabolized drug may also expose patients to undesirable toxic or reactive metabolites.
[2] Another ADME limitation that affects many medicines is the formation of toxic or biologically reactive metabolites. As a result, some patients receiving the drug may experience toxicities, or the safe dosing of such drugs may be limited such that patients receive a suboptimal amount of the active agent. In certain cases, modifying dosing intervals or formulation approaches can help to reduce clinical adverse effects, but often the formation of such undesirable metabolites is intrinsic to the metabolism of the compound.
[3] In some select cases, a metabolic inhibitor will be co-administered with a drug that is cleared too rapidly. Such is the case with the protease inhibitor class of drugs that are used to treat HIV infection. The FDA recommends that these drugs be co- dosed with ritonavir, an inhibitor of cytochrome P450 enzyme 3A4 (CYP3A4), the enzyme typically responsible for their metabolism (see Kempf, D.J. et al.,
Antimicrobial agents and chemotherapy, 1997, 41(3): 654-60). Ritonavir, however, causes adverse effects and adds to the pill burden for HIV patients who must already take a combination of different drugs. Similarly, the CYP2D6 inhibitor quinidine has been added to dextromethorphan for the purpose of reducing rapid CYP2D6 metabolism of dextromethorphan in a treatment of pseudobulbar affect. Quinidine, however, has unwanted side effects that greatly limit its use in potential combination therapy (see Wang, L et al., Clinical Pharmacology and Therapeutics, 1994, 56(6 Pt 1): 659-67; and FDA label for quinidine at www.accessdata.fda.gov).
[4] In general, combining drugs with cytochrome P450 inhibitors is not a satisfactory strategy for decreasing drug clearance. The inhibition of a CYP enzyme's activity can affect the metabolism and clearance of other drugs metabolized by that same enzyme. CYP inhibition can cause other drugs to accumulate in the body to toxic levels.
[5] A potentially attractive strategy for improving a drug's metabolic properties is deuterium modification. In this approach, one attempts to slow the CYP -mediated metabolism of a drug or to reduce the formation of undesirable metabolites by replacing one or more hydrogen atoms with deuterium atoms. Deuterium is a safe, stable, non-radioactive isotope of hydrogen. Compared to hydrogen, deuterium forms stronger bonds with carbon. In select cases, the increased bond strength imparted by deuterium can positively impact the ADME properties of a drug, creating the potential for improved drug efficacy, safety, and/or tolerability. At the same time, because the size and shape of deuterium are essentially identical to those of hydrogen,
replacement of hydrogen by deuterium would not be expected to affect the
biochemical potency and selectivity of the drug as compared to the original chemical entity that contains only hydrogen.
[6] Over the past 35 years, the effects of deuterium substitution on the rate of metabolism have been reported for a very small percentage of approved drugs (see, e.g., Blake, MI et al, J Pharm Sci, 1975, 64:367-91 ; Foster, AB, Adv Drug Res 1985, 14: 1-40 ("Foster"); Kushner, DJ et al, Can J Physiol Pharmacol 1999, 79-88; Fisher, MB et al, Curr Opin Drug Discov Devel, 2006, 9: 101-09 ("Fisher")). The results have been variable and unpredictable. For some compounds deuteration caused decreased metabolic clearance in vivo. For others, there was no change in
metabolism. Still others demonstrated increased metabolic clearance. The variability in deuterium effects has also led experts to question or dismiss deuterium
modification as a viable drug design strategy for inhibiting adverse metabolism (see Foster at p. 35 and Fisher at p. 101).
[7] The effects of deuterium modification on a drug's metabolic properties are not predictable even when deuterium atoms are incorporated at known sites of
metabolism. Only by actually preparing and testing a deuterated drug can one determine if and how the rate of metabolism will differ from that of its non-deuterated counterpart. See, for example, Fukuto et al. (J. Med. Chem. 1991 , 34, 2871-76). Many drugs have multiple sites where metabolism is possible. The site(s) where deuterium substitution is required and the extent of deuteration necessary to see an effect on metabolism, if any, will be different for each drug.
[8] Dasabuvir, also known as Exviera™, ABT-333 and N-[6-[3-tert-Butyl-5-(2,4- dioxo-3,4-dihydropyrimidin-l(2H)-yl)-2-methoxyphenyl]naphthalen-2- yljmethanesulfonamide, is a non-nucleoside hepatitis C virus NS5B palm polymerase inhibitor. It has recently been approved in the United States and is recommended for approval in the EU for the treatment of humans with genotype 1 or 4 chronic hepatitis C infection and cirrhosis in a combination regimen with paritaprevir (an NS3/4A protease inhibitor), ritonavir, and ombitasvir (an NS5A inhibitor) with and without ribavirin. The approved dosage form requires a subject to take one tablet comprising dasabuvir and one tablet comprising a combination of paritaprevir, ritonavir, arid ombitasvir simultaneously in the morning and then a second table comprising dasabuvir in the evening. This combination regimen (without ribavirin) is also known as VIEKIRA PA ®. Despite the prospects for dasabuvir and VIEKIRA PAK®, it would be advantageous to reduce the dosing of dasabuvir to once a day.
SUMMARY OF THE INVENTION
[9] This invention relates to novel deuterated pyrimidinedione derivatives, and pharmaceutically acceptable salts thereof. This invention also provides compositions comprising a compound of this invention and the use of such compositions in methods of treating hepatitis C viral infections. The invention also provides novel intermediates useful in the synthesis of the compounds of the invention. Such intermediates may also have hepatitis C virus inhibitory properties. DETAILED DESCRIPTION OF THE INVENTION
Definitions
[10] The term "treat" means decrease, suppress, attenuate, diminish, arrest, or stabilize the development or progression of a disease (e.g., a disease or disorder delineated herein), lessen the severity of the disease or improve the symptoms associated with the disease.
[11] "Disease" means any condition or disorder that damages or interferes with the normal function of a cell, tissue, or organ.
[12] It will be recognized that some variation of natural isotopic abundance occurs in a synthesized compound depending upon the origin of chemical materials used in the synthesis. Thus, a preparation of Compound 1 will inherently contain small amounts of deuterated isotopologues. The concentration of naturally abundant stable hydrogen and carbon isotopes, notwithstanding this variation, is small and immaterial as compared to the degree of stable isotopic substitution of compounds of this invention. See, for instance, Wada, E et al., Seikagaku, 1994, 66: 15; Gannes, LZ et al., Comp Biochem Physiol Mol Integr Physiol, 1998, 119:725.
[13] In the compounds of this invention any atom not specifically designated as a particular isotope is meant to represent any stable isotope of that atom. Unless otherwise stated, when a position is designated specifically as "H" or "hydrogen", the position is understood to have hydrogen at its natural abundance isotopic composition. Also unless otherwise stated, when a position is designated specifically as "D" or "deuterium", the position is understood to have deuterium at an abundance that is at least 3340 times greater than the natural abundance of deuterium, which is 0.015% (i.e., at least 50.1 % incorporation of deuterium).
[14] The term "isotopic enrichment factor" as used herein means the ratio between the isotopic abundance and the natural abundance of a specified isotope.
[15] In other embodiments, a compound of this invention has an isotopic enrichment factor for each designated deuterium atom of at least 3500 (52.5% deuterium incorporation at each designated deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium), at least 5500 (82.5% deuterium incorporation), at least 6000 (90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5% deuterium incorporation). [16] The term "isotopologue" refers to a species in which the chemical structure differs from a specific compound of this invention only in the isotopic composition thereof.
[17] The term "compound," when referring to a compound of this invention, refers to a collection of molecules having an identical chemical structure, except that there may be isotopic variation among the constituent atoms of the molecules. Thus, it will be clear to those of skill in the art that a compound represented by a particular chemical structure containing indicated deuterium atoms, will also contain lesser amounts of isotopologues having hydrogen atoms at one or more of the designated deuterium positions in that structure. The relative amount of such isotopologues in a compound of this invention will depend upon a number of factors including the isotopic purity of deuterated reagents used to make the compound and the efficiency of incorporation of deuterium in the various synthesis steps used to prepare the compound. However, as set forth above the relative amount of such isotopologues in toto will be less than 49.9% of the compound. In other embodiments, the relative amount of such isotopologues in toto will be less than 47.5%, less than 40%, less than 32.5%, less than 25%, less than 17.5%, less than 10%, less than 5%, less than 3%, less than 1%), or less than 0.5% of the compound.
[18] The invention also provides salts of the compounds of the invention.
[19] A salt of a compound of this invention is formed between an acid and a basic group of the compound, such as an amino functional group, or a base and an acidic group of the compound, such as a carboxyl functional group. According to another embodiment, the compound is a pharmaceutically acceptable acid addition salt.
[20] The term "pharmaceutically acceptable," as used herein, refers to a component that is, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and other mammals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. A "pharmaceutically acceptable salt" means any non-toxic salt that, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention. A "pharmaceutically acceptable counterion" is an ionic portion of a salt that is not toxic when released from the salt upon administration to a recipient.
[21] Acids commonly employed to form pharmaceutically acceptable salts include inorganic acids such as hydrogen bisulfide, hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid and phosphoric acid, as well as organic acids such as para-toluenesulfonic acid, salicylic acid, tartaric acid, bitartaric acid, ascorbic acid, maleic acid, besylic acid, fumaric acid, gluconic acid, glucuronic acid, formic acid, glutamic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, lactic acid, oxalic acid, para-bromophenylsulfonic acid, carbonic acid, succinic acid, citric acid, benzoic acid and acetic acid, as well as related inorganic and organic acids. Such pharmaceutically acceptable salts thus include sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, phosphate, monohydrogenphosphate, dihydrogenphosphate, metaphosphate, pyrophosphate, chloride, bromide, iodide, acetate, propionate, decanoate, caprylate, acrylate, formate, isobutyrate, caprate, heptanoate, propiolate, oxalate, malonate, succinate, suberate, sebacate, fumarate, maleate, butyne- 1 ,4-dioate, hexyne-l,6-dioate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, phthalate, terephthalate, sulfonate, xylene sulfonate, phenylacetate, phenylpropionate, phenylbutyrate, citrate, lactate, β- hydroxybutyrate, glycolate, maleate, tartrate, methanesulfonate, propanesulfonate, naphthalene- 1 -sulfonate, naphthalene-2- sulfonate, mandelate and other salts. In one embodiment, pharmaceutically acceptable acid addition salts include those formed with mineral acids such as hydrochloric acid and hydrobromic acid, and especially those formed with organic acids such as maleic acid.
[22] The pharmaceutically acceptable salt may also be a salt of a compound of the present invention having an acidic functional group, such as a carboxylic acid functional group, and a base. Exemplary bases include, but are not limited to, hydroxide of alkali metals including sodium, potassium, and lithium; hydroxides of alkaline earth metals such as calcium and magnesium; hydroxides of other metals, such as aluminum and zinc; ammonia, organic amines such as unsubstituted or hydroxyl-substituted mono-, di-, or tri-alkylamines, dicyclohexylamine; tributyl amine; pyridine; N-methyl, N-ethylamine; diethylamine; triethylamine; mono-, bis-, or tris-(2-OH-(Ci-C6)-alkylamine), such as N,N-dimethyl-N-(2-hydroxyethyl)amine or tri-(2-hydroxyethyl)amine; N-methyl-D-glucamine; morpholine; thiomorpholine; piperidine; pyrrolidine; and amino acids such as arginine, lysine, and the like.
[23] In one embodiment, the pharmaceutically acceptable salt is a sodium salt, more specifically a monosodium salt.
[24] The compounds of the present invention (e.g., compounds of Formula I), may contain an asymmetric carbon atom, for example, as the result of deuterium
substitution or otherwise. As such, compounds of this invention can exist as either individual enantiomers, or mixtures of the two enantiomers. Accordingly, a compound of the present invention may exist as either a racemic mixture or a scalemic mixture, or as individual respective stereoisomers that are substantially free from another possible stereoisomer. The term "substantially free of other
stereoisomers" as used herein means less than 25% of other stereoisomers, preferably less than 10% of other stereoisomers, more preferably less than 5% of other stereoisomers and most preferably less than 2% of other stereoisomers are present. Methods of obtaining or synthesizing an individual enantiomer for a given compound are known in the art and may be applied as practicable to final compounds or to starting material or intermediates.
[25] Unless otherwise indicated, when a disclosed compound is named or depicted by a structure without specifying the stereochemistry and has one or more chiral centers, it is understood to represent all possible stereoisomers of the compound.
[26] The term "stable compounds," as used herein, refers to compounds which possess stability sufficient to allow for their manufacture and which maintain the integrity of the compound for a sufficient period of time to be useful for the purposes detailed herein (e.g., formulation into therapeutic products, intermediates for use in production of therapeutic compounds, isolatable or storable intermediate compounds, treating a disease or condition responsive to therapeutic agents).
[27] "D" and "d" both refer to deuterium. "Stereoisomer" refers to both enantiomers and diastereomers. "Tert" and "t-" each refer to tertiary. "US" refers to the United States of America.
[28] "Substituted with deuterium" refers to the replacement of one or more hydrogen atoms with a corresponding number of deuterium atoms.
[29] Throughout this specification, a variable may be referred to generally
(e.g., "each R") or may be referred to specifically (e.g., R1, R2, R3, etc.). Unless otherwise indicated, when a variable is referred to generally, it is meant to include all specific embodiments of that particular variable. Therapeutic Compounds
[30] The present invention provides a compound of Formula I:
Figure imgf000009_0001
, or a pharmaceutically acceptable salt thereof, wherein:
R1 is a t-butyl moiety optionally comprising between 1 and 9 deuterium substitutions;
each R2 and R3 is independently selected from -CH3, -CH2D, -CHD2 and -CD3; and
each of Y', Y2, Y3, Y4, Y5, Y6, Y7, Y8 and Y9 is independently selected from hydrogen and deuterium,
wherein at least one of R1, R2 and R3 comprises a deuterium atom.
[31] In some embodiments R1 is selected from -C(CH3)3 and -C(CD3)3. In one aspect of these embodiments, R1 is -C(CH3)3. In an alternate aspect of these embodiments, R1 is -C(CD3)3.
[32] In some embodiments, each of R2 and R3 are independently selected from -CH3 and -CD3. In one aspect of these embodiments, R2 is -CD3 and R3 is -CH3.
In an alternate aspect of these embodiments, R2 is -CH3 and R3 is -CD3. In still another aspect of these embodiments, R2 is -CD3 and R3 is -CD3. In still another aspect of these embodiments, R2 is -CH3 and R3 is -CH3.
[33] In some embodiments, Y1 and Y2 are the same.
[34] In some embodiments, Y4, Y5, Y6, Y7, Y8 and Y9 are the same. In one aspect of these embodiments, Y1 and Y2 are the same. In a more specific aspect of these embodiments, Y1 and Y2 are the same; R1 is selected from -C(CH3)3 and -C(CD3)3 and each of R2 and R3 are independently selected from -CH3 and -CD3. In an even more specific aspect of these embodiments each of Y1, Y2, Y3, Y4, Y3, Y6, Y7, Y8 and Y9 is hydrogen; R1 is selected from -C(CH3)3 and -C(CD3)3 and each of R2 and R3 are independently selected from -CH3 and -CD3.
[35] In some embodiments, R1 is -C(CD3)3, R2 is -CD3 and R3 is CD3.
[36] In some embodiments, R1 is -C(CD3)3, R2 is -CH3 and R3 is CD3. [37] In some embodiments, R1 is -C(CD3)3, R2 is -CD3 and R3 is CH3.
[38] In some embodiments, R1 is -C(CD3)3, R2 is -CH3 and R3 is CH3.
[39] In some embodiments, R1 is -C(CH3)3, R2 is -CD3 and R3 is CD3.
[40] In some embodiments, R1 is -C(CH3)3, R2 is -CH3 and R3 is CD3.
[41] In some embodiments, R1 is -C(CH3)3, R2 is -CD3 and R3 is CH3.
[42] In one embodiment of a compound of Formula I, the invention does not include a compound wherein R1 is -C(CD3)3, each of R2 and R3 is -CD3; and each of
Y1, Y2, Y3, Y4, Y5, Y6, Y7, Y8 and Y9 is deuterium.
[43] In some embodiments, Y1 and Y2 are the same; Y4, Y3, Y6, Y7, Y8 and Y9 are the same; and the compound is selected from any one of the compounds (Cmpd) set forth in Table 1 (below):
Table 1 : Exemplary Embodiments of Formula I
Figure imgf000010_0001
Figure imgf000011_0001
or a pharmaceutically acceptable salt thereof.
[44] In another set of embodiments, any atom not designated as deuterium in any of the embodiments set forth above is present at its natural isotopic abundance.
[45] The synthesis of compounds of Formula I may be readily achieved by synthetic chemists of ordinary skill by reference to the Exemplary Synthesis and Examples disclosed herein. Relevant procedures analogous to those of use for the preparation of compounds of Formula I and intermediates thereof are disclosed, for instance in PCT patent publications WO2009039134, WO2009039127,
WO2012009699, and WO2014031791. [46] Such methods can be carried out utilizing corresponding deuterated and optionally, other isotope-containing reagents and/or intermediates to synthesize the compounds delineated herein, or invoking standard synthetic protocols known in the art for introducing isotopic atoms to a chemical structure.
Exemplary Synthesis
[47] A convenient method for synthesizing compounds of Formula I is depicted in Scheme 1 , below.
[48] Scheme 1 : General S nthesis of Compounds of Formula I
Figure imgf000012_0001
(1 ) (2) (4)
Figure imgf000012_0002
Figure imgf000012_0003
Reagents and conditions: (a) Nal, NaOH, NaOCl; (b) (3): (CD3)2S04, K2C03 or CD3-I, NaOH; (c) K3PO4, Cul, Sodium ascorbate, N-(2-cyanophenyl)picolinamide or N-(2- cyanophenyl)benzamide; (d)K3P04, Pd2dba3, l,3,5,7-Tetramethyl-6-phenyl-2,4,8-trioxa-6- phosphaadamantane
[49] Iodination of appropriately deuterated phenol intermediate (1) with Nal and NaOCl in methanolic NaOH provides corresponding diiodophenol intermediate (2), which is O-alkylated with reagent (3), Dimethyl sulfate-d6 (99 atom % D) and K2C03 or with Iodomethane-d3 (99.5 atom % D) and NaOH to afford correspondingly deuterated diiodo aryl ether intermediate (4). Goldberg reaction of aryl iodide (4) with Uracil-5,6-d2 (98 atom % D) (5) in the presence of Cul, K3P04 and N-(2- cyanophenyl)picolinamide or using N-(2-cyanophenyl)benzamide and a trace of sodium ascorbate furnishes appropriately deuterated uracil intermediate (6). Suzuki coupling of aryl iodide intermediate (6) with appropriately deuterated
naphthylboronate (7) using Pd2dba3, phosphine ligand and K3P04, affords
correspondingly deuterated compounds of Formula (I)
[50] Scheme 2: Pre aration of Intermediate (1
Figure imgf000013_0001
Reagents and conditions: (a) t-BuLi, CD3OD; (b) cation exchange resin Amberlyst 15
[51] Lithiation of starting bromophenol (8) followed by deuteration in accordance with the prodecure described by Shimizu, H. et al., Tetrahedron Letters, 47(33), 5927-5931 ; 2006 affords appropriately deuterated intermediate (9), which is subsequently treated with starting material (10), 2-Methylpropene-d8 ( 99 atom % D) in a manner analogous to the method described by Chaudhuri, B. et al., Industrial & Engineering Chemistry Research, 30(1), 227-31 ; 1991, to furnish intermediate (1).
[52] Scheme 3: Preparation of Intermediate (7)
Figure imgf000014_0001
Reagents and conditions: (a) Br2, Ph3P; (b) acetyl chloride, A1C13, NaOCl; (c) DPPA; (d) CD3S02C1; (e) bis(pinacolato)diboron, KOAc, combiPhos-Pd6 catalyst
[53] Starting material (11) 2-Naphthol-l ,3,4,5,6,7,8-d7 (97 atom %D) is treated with a brominating reagent such as Bromine in a manner analogous to the procedure described by Schaefer, J. et al., Journal of Organic Chemistry, 32(5), 1607-8; 1967 to produce appropriately deuterated intermediate (12). Friedel Crafts acylation of intermediate (12), followed by oxidation affords appropriately deuterated intermediate (13) in a manner analogous to the procedure described in U.S. 4454341. Diazotization of intermediate (13) with diphenyphosphoryl azide provides aminonaphthalene intermediate (14) which is treated with Methane-d3-sulfonyl chloride (98 atom % D) followed by bis(pinacolato)diboron, in the presence of KOAc, and combiPhos-Pd6 catalyst to afford intermediate (7).
[54] The specific approaches and compounds shown above are not intended to be limiting. The chemical structures in the schemes herein depict variables that are hereby defined commensurately with chemical group definitions (moieties, atoms, etc.) of the corresponding position in the compound formulae herein, whether identified by the same variable name (i.e., R1, R2, R3, etc.) or not. The suitability of a chemical group in a compound structure for use in the synthesis of another compound is within the knowledge of one of ordinary skill in the art.
[55] Additional methods of synthesizing compounds of Formula I and their synthetic precursors, including those within routes not explicitly shown in schemes herein, are within the means of chemists of ordinary skill in the art. Synthetic chemistry transformations and protecting group methodologies (protection and deprotection) useful in synthesizing the applicable compounds are known in the art and include, for example, those described in Larock R, Comprehensive Organic Transformations, VCH Publishers (1989); Greene, TW et al., Protective Groups in Organic Synthesis, 3rd Ed., John Wiley and Sons (1999); Fieser, L et al., Fieser and Fieser 's Reagents for Organic Synthesis, John Wiley and Sons (1994); and Paquette, L, ed., Encyclopedia of Reagents for Organic Synthesis, John Wiley and Sons (1995) and subsequent editions thereof.
[56] Combinations of substituents and variables envisioned by this invention are only those that result in the formation of stable compounds.
Compositions
[57] The invention also provides pharmaceutical compositions comprising an effective amount of a compound of Formula I (e.g., including any of the formulae herein), or a pharmaceutically acceptable salt of said compound; and a
pharmaceutically acceptable carrier. The carrier(s) are "acceptable" in the sense of being compatible with the other ingredients of the formulation and, in the case of a pharmaceutically acceptable carrier, not deleterious to the recipient thereof in an amount used in the medicament.
[58] Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene- polyoxypropylene-block polymers, polyethylene glycol and wool fat.
[59] If required, the solubility and bioavailability of the compounds of the present invention in pharmaceutical compositions may be enhanced by methods well-known in the art. One method includes the use of lipid excipients in the formulation. See "Oral Lipid-Based Formulations: Enhancing the Bioavailability of Poorly Water- Soluble Drugs (Drugs and the Pharmaceutical Sciences)," David J. Hauss, ed. Informa Healthcare, 2007; and "Role of Lipid Excipients in Modifying Oral and Parenteral Drug Delivery: Basic Principles and Biological Examples," Kishor M. Wasan, ed. Wiley-Interscience, 2006.
[60] Another known method of enhancing bioavailability is the use of an amorphous form of a compound of this invention optionally formulated with a poloxamer, such as LUTROL™ and PLURONIC™ (BASF Corporation), or block copolymers of ethylene oxide and propylene oxide. See United States patent
7,014,866; and United States patent publications 20060094744 and 20060079502.
[61] In one embodiment, the bioavailability enhancing agent is one or more polymers selected from copovidone, polyvinylpyrrolidone,
hydroxymethylpropylcellulose, SOLUPLUS®, and combinations thereof; wherein the hydroxymethylpropylcellulose has a viscosity less than about 100 centipoise in a 2% solution of at a temperature of about 20°C. In a more specific embodiment, the bioavailability enhancing agent comprises copovidone.
[62] The pharmaceutical compositions of the invention include those suitable for oral, rectal, nasal, topical (including buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration. In certain embodiments, the compound of the formulae herein is administered transdermally (e.g., using a transdermal patch or iontophoretic techniques). Other formulations may conveniently be presented in unit dosage form, e.g., tablets, sustained release capsules, and in liposomes, and may be prepared by any methods well known in the art of pharmacy. See, for example, Remington: The Science and Practice of Pharmacy, Lippincott Williams & Wilkins, Baltimore, MD (20th ed. 2000).
[63] Such preparative methods include the step of bringing into association with the molecule to be administered ingredients such as the carrier that constitutes one or more accessory ingredients. In general, the compositions are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers, liposomes or finely divided solid carriers, or both, and then, if necessary, shaping the product.
[64] In certain embodiments, the compound is administered orally. Compositions of the present invention suitable for oral administration may be presented as discrete units such as capsules, sachets, or tablets each containing a predetermined amount of the active ingredient; a powder or granules; a solution or a suspension in an aqueous liquid or a non-aqueous liquid; an oil-in-water liquid emulsion; a water-in-oil liquid emulsion; packed in liposomes; or as a bolus, etc. Soft gelatin capsules can be useful for containing such suspensions, which may beneficially increase the rate of compound absorption. In a specific aspect of these embodiments, the oral composition is a tablet.
[65] In the case of tablets for oral use, carriers that are commonly used include lactose and corn starch. Lubricating agents,, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried cornstarch. When aqueous suspensions are administered orally, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavoring and/or coloring agents may be added.
[66] Compositions suitable for oral administration include lozenges comprising the ingredients in a flavored basis, usually sucrose and acacia or tragacanth; and pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia.
[67] Compositions suitable for parenteral administration include aqueous .and nonaqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. The formulations may be presented in unit-dose or multi-dose containers, for example, sealed ampules and vials, and may be stored in a freeze dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use.
Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets.
[68] Such injection solutions may be in the form, for example, of a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example, as a solution in 1,3- butanediol. Among the acceptable vehicles and solvents that may be employed are mannitol, water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono- or diglycerides. Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant.
[69] The pharmaceutical compositions of this invention may be administered in the form of suppositories for rectal administration. These compositions can be prepared by mixing a compound of this invention with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the active components. Such materials include, but are not limited to, cocoa butter, beeswax and polyethylene glycols.
[70] The pharmaceutical compositions of this invention may be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizing or dispersing agents known in the art. See, e.g.: Rabinowitz JD and Zaffaroni AC, US Patent 6,803,031, assigned to Alexza Molecular Delivery Corporation.
[71] Topical administration of the pharmaceutical compositions of this invention is especially useful when the desired treatment involves areas or organs readily accessible by topical application. For topical application topically to the skin, the pharmaceutical composition should be formulated with a suitable ointment containing the active components suspended or dissolved in a carrier. Carriers for topical administration of the compounds of this invention include, but are not limited to, mineral oil, liquid petroleum, white petroleum, propylene glycol, polyoxyethylene polyoxypropylene compound, emulsifying wax, and water. Alternatively, the pharmaceutical composition can be formulated with a suitable lotion or cream containing the active compound suspended or dissolved in a carrier. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol, and water. The pharmaceutical compositions of this invention may also be topically applied to the lower intestinal tract by rectal suppository formulation or in a suitable enema formulation. Topically-transdermal patches and iontophoretic administration are also included in this invention.
[72] In other embodiments, a composition of this invention further comprises a second therapeutic agent. The second therapeutic agent may be selected from any compound or therapeutic agent known to have or that demonstrates advantageous properties for the treatment of HCV infection. In one aspect of these embodiments, the second therapeutic agent is an agent selected from a HCV NS3/NS4A protease inhibitor, a HCV NS5A inhibitor, a cytochrome P450 inhibitor, a second HCV NS5B polymerase inhibitor, and a guanosine analog. In a more specific aspect of these embodiments, the HCV NS3 NS4A protease inhibitor is paritaprevir. In another more specific aspect of these embodiments, the HCV NS5A inhibitor is ombitasvir. In yet another more specific aspect of these embodiments, the cytochrome P450 inhibitor is ritonavir. In still another more specific aspect of these embodiments, the guanosine analog is ribavirin.
[73] In other embodiments, the invention provides separate dosage forms of a compound of this invention and one or more of any of the above-described second therapeutic agents, wherein the compound and second therapeutic agent are associated with one another. The term "associated with one another" as used herein means that the separate dosage forms are packaged together or otherwise attached to one another such that it is readily apparent that the separate dosage forms are intended to be sold and administered together (within less than 24 hours of one another, consecutively or simultaneously). In a specific aspect of these embodiments, the compound of the invention is packaged together with a HCV NS3/NS4A protease inhibitor, a HCV NS5A inhibitor, and a cytochrome P450 inhibitor. In a more specific aspect of these embodiments, the HCV NS3/NS4A protease inhibitor, HCV NS5A inhibitor, and cytochrome P450 inhibitor are co-formulated into a single dosage form and packaged with the compound of the invention in a separate dosage form. In an even more specific aspect of these embodiments, the compound of the invention is a single dosage form packaged together with a separate single dosage form comprising paritaprevir, ombitasvir and ritonavir. In another even more specific aspect of these embodiments, the compound of the invention is a single dosage form packaged together with a separate single dosage form comprising paritaprevir and ombitasvir, but lacking ritonavir or another cytochrome P450 inhibitor.
[74] In still other embodiments, the invention provides a packaged pharmaceutical composition comprising two separate, but identical oral dosage forms of a compound of the invention; a third separate oral dosage form comprising a combination of paritaprevir, ombitasvir and ritonavir; and instructions for when during the day to take each of the first, second and third dosage forms.
[75] In the pharmaceutical compositions of the invention, the compound of the present invention is present in an effective amount. As used herein, the term
"effective amount" refers to an amount which, when administered in a proper dosing regimen, is sufficient to treat the target disorder.
[76] The interrelationship of dosages for animals and humans (based on milligrams per meter squared of body surface) is described in Freireich et al., Cancer Chemother. Rep, 1966, 50: 219. Body surface area may be approximately determined from height and weight of the subject. See, e.g., Scientific Tables, Geigy Pharmaceuticals, Ardsley, N.Y., 1970, 537.
[77] In one embodiment, an effective amount of a compound of this invention for oral dosing to an adult can range from about 0.01 mg to about 1000 mg, specifically from about 25 mg to about 600 mg, from about 50 mg to about 600 mg, from about 25 mg to 500 mg, from about 50 mg to 500 mg, from about 25 mg to 400 mg, and from about 50 mg to 400 mg per day. The daily dosage may be administered as single dose or in divided doses, e.g., two doses per day.
[78] Effective doses will also vary, as recognized by those skilled in the art, depending on the diseases treated, the severity of the disease, the route of
administration, the sex, age and general health condition of the subject, excipient usage, the possibility of co-usage with other therapeutic treatments such as use of other agents and the judgment of the treating physician. For example, guidance for selecting an effective dose can be determined by reference to the prescribing information for Compound 1.
[79] For pharmaceutical compositions that comprise a second therapeutic agent, an effective amount of the second therapeutic agent is between about 20% and 100% of the dosage normally utilized in a monotherapy regime using just that agent.
Preferably, an effective amount is between about 70% and 100% of the normal monotherapeutic dose. The normal monotherapeutic dosages of these second therapeutic agents are well known in the art. See, e.g., Wells et al., eds.,
Pharmacotherapy Handbook, 2nd Edition, Appleton and Lange, Stamford, Conn. (2000); PDR Pharmacopoeia, Tarascon Pocket Pharmacopoeia 2000, Deluxe Edition, Tarascon Publishing, Loma Linda, Calif. (2000), each of which references are incorporated herein by reference in their entirety.
[80] It is expected that some of the second therapeutic agents referenced above will act synergistically with the compounds of this invention. When this occurs, it will allow the effective dosage of the second therapeutic agent and/or the compound of this invention to be reduced from that required in a monotherapy. This has the advantage of minimizing toxic side effects of either the second therapeutic agent of a compound of this invention, synergistic improvements in efficacy, improved ease of administration or use and/or reduced overall expense of compound preparation or formulation.
Methods of Treatment
[81] In another embodiment, the invention provides a method of inhibiting the activity of HCV NS5B polymerase in a cell infected with HCV, comprising contacting a cell with one or more compounds of Formula I herein, or a pharmaceutically acceptable salt thereof.
[82] . According to another embodiment, the invention provides a method of treating a HCV infection.
[83] In other embodiments, the above method of treatment comprises the further step of co-administering to the subject in need thereof one or more second therapeutic agents. The choice of second therapeutic agents may be made from any second therapeutic agent known to be useful to treat HCV infections. In one aspect of these embodiments, the second therapeutic agent is one of more agents selected from a HCV NS3/NS4A protease inhibitor, a HCV NS5A inhibitor, a cytochrome P450 inhibitor, a second HCV NS5B polymerase inhibitor, and a guanosine analog. In a more specific aspect of these embodiments, the HCV NS3 S4A protease inhibitor is paritaprevir. In another more specific aspect of these embodiments, the HCV NS5A inhibitor is ombitasvir. In yet another more specific aspect of these embodiments, the cytochrome P450 inhibitor is ritonavir. In still another more specific aspect of these embodiments, the guanosine analog is ribavirin. In an even more specific aspect of these embodiments, the second therapeutic agents are a combination of a HCV NS3 NS4A protease inhibitor, a HCV NS5A inhibitor, and, optionally, a cytochrome P450 inhibitor. In another even more specific aspect of these embodiments, the second therapeutic agents are a combination of paritaprevir, ombitasvir, and, optionally, ritonavir. In still another even more specific aspect of these embodiments, the second therapeutic agents are a combination of paritaprevir, ombitasvir, optionally ritonavir, and optionally ribavirin.
[84] The term "co-administered" as used herein means that the second therapeutic agent may be administered together with a compound of this invention as part of a single dosage form (such as a composition of this invention comprising a compound of the invention and an second therapeutic agent as described above) or as separate, multiple dosage forms. Alternatively, the additional agent may be administered prior to, consecutively with, or following the administration of a compound of this invention. In such combination therapy treatment, both the compounds of this invention and the second therapeutic agent(s) are administered by conventional methods. The administration of a composition of this invention, comprising both a compound of the invention and a second therapeutic agent, to a subject does not preclude the separate administration of that same therapeutic agent, any other second therapeutic agent or any compound of this invention to said subject at another time during a course of treatment. Moreover, not all doses of a compound of the invention administered each day need be co-administered with the one or more second therapeutic agent. For example, in some embodiments, a compound of the invention may be administered twice a day, but only co-administered with one or more second therapeutic agents (e.g., a NS3/4A protease inhibitor (e.g., ombitasvir), a NS5A polymerase inhibitor (paritaprevir) and a cytochrome P450 inhibitor (e.g., ritonavir)) in one of the two dosings.
[85] Effective amounts of these second therapeutic agents are well known to those skilled in the art and guidance for dosing may be found in patents and published patent applications referenced herein, as well as in Wells et al., eds., Pharmacotherapy Handbook, 2nd Edition, Appleton and Lange, Stamford, Conn. (2000); PDR
Pharmacopoeia, Tarascon Pocket Pharmacopoeia 2000, Deluxe Edition, Tarascon Publishing, Loma Linda, Calif. (2000), and other medical texts. However, it is well within the skilled artisan's purview to determine the second therapeutic agent's optimal effective-amount range.
[86] In one embodiment of the invention, where a second therapeutic agent is administered to a subject, the effective amount of the compound of this invention is less than its effective amount would be where the second therapeutic agent is not administered. In another embodiment, the effective amount of the second therapeutic agent is less than its effective amount would be where the compound of this invention is not administered. In this way, undesired side effects associated with high doses of either agent may be minimized. Other potential advantages (including without limitation improved dosing regimens and/or reduced drug cost) will be apparent to those of skill in the art.
[87] In yet another aspect, the invention provides the use of a compound of Formula I alone or together with one or more of the above-described second therapeutic agents in the manufacture of a medicament, either as a single composition or as separate dosage forms, for treatment in a subject of a disease, disorder or symptom set forth above. Another aspect of the invention is a compound of Formula I for use in the treatment in a subject of a disease, disorder or symptom thereof delineated herein.
Example 1. Evaluation of Metabolic Stability
[88] Microsomal Assay : Human liver microsomes (20 mg/mL) are obtained from Xenotech, LLC (Lenexa, KS). β-nicotinamide adenine dinucleotide phosphate, reduced form (NADPH), magnesium chloride (MgCb), and dimethyl sulfoxide (DMSO) are purchased from Sigma-Aldrich.
[89] Determination of Metabolic Stability: 7.5 mM stock solutions of test compounds are prepared in DMSO. The 7.5 mM stock solutions are diluted to 12.5- 50 μΜ in acetonitrile (ACN). The 20 mg/mL human liver microsomes are diluted to 0.625 mg/mL in 0.1 M potassium phosphate buffer, pH 7.4, containing 3 mM MgCl2. The diluted microsomes are added to wells of a 96-well deep-well polypropylene plate in triplicate. A 10 aliquot of the 12.5-50 μΜ test compound is added to the microsomes and the mixture is pre-warmed for 10 minutes. Reactions are initiated by addition of pre-warmed NADPH solution. The final reaction volume is 0.5 mL and contains 0.5 mg/mL human liver microsomes, 0.25-1.0 μΜ test compound, and 2 mM NADPH in 0.1 M potassium phosphate buffer, pH 7.4, and 3 mM MgCl2. The reaction mixtures are incubated at 37 °C, and 50 μΐ^ aliquots are removed at 0, 5, 10, 20, and 30 minutes and added to shallow-well 96-well plates which contain 50 μΐ, of ice-cold ACN with internal standard to stop the reactions. The plates are stored at 4 °C for 20 minutes after which 100 μL of water is added to the wells of the plate before centrifugation to pellet precipitated proteins. Supernatants are transferred to another 96-well plate and analyzed for amounts of parent remaining by LC-MS/MS using an Applied Bio-systems API 4000 mass spectrometer. The same procedure is followed for the non-deuterated counterpart of the compound of Formula I and the positive control, 7-ethoxycoumarin (1 μΜ). Testing is done in triplicate.
[90] Data analysis: The in vitro ti/2s for test compounds are calculated from the slopes of the linear regression of % parent remaining (In) vs incubation time relationship.
in vitro t ½ = 0.693/k
k = -[slope of linear regression of % parent remaining (In) vs incubation time]
[91] Data analysis is performed using Microsoft Excel Software. [92] Without further description, it is believed that one of ordinary skill in the art can, using the preceding description and the illustrative examples, make and utilize the compounds of the present invention and practice the claimed methods. It should be understood that the foregoing discussion and examples merely present a detailed description of certain preferred embodiments. It will be apparent to those of ordinary skill in the art that various modifications and equivalents can be made without departing from the spirit and scope of the invention.

Claims

We claim:
1. A compound of Formula I:
Figure imgf000026_0001
, or a pharmaceutically acceptable salt thereof, wherein:
R1 is a t-butyl moiety optionally comprising between 1 and 9 deuterium substitutions;
each R2 and R3 is independently selected from -CH3, -CH2D, -CHD2 and -CD3; and
each of Y1, Y2, Y3, Y4, Y5, Y6, Y7, Y8 and Y9 is independently selected from hydrogen and deuterium, wherein
at least one of R1, R2 and R3 comprises a deuterium atom.
2. The compound of claim 1, wherein R1 is selected from -C(CH3)3
and -C(CD3)3.
3. The compound of claim 1 or 2, wherein each of R2 and R3 is independently selected from -CH3 and -CD3.
4. The compound of any one of claims 1-3, wherein Y1 and Y2 are the same.
5. The compound of any one of claims 1 -4, wherein Y4, Y\ Y6, Y7, Y8 and Y9 are the same.
6. The compound of claim 5, wherein each of Y1, Y2, Y3, Y4, Y5, Y6, Y7, Y8 and Y9 is hydrogen.
7. The compound of any one of claims 1-6, wherein R1 is -C(CD3)3, R2 is -CD3 and R3 is CD3.
8. The compound of any one of claims 1-6, wherein R1 is -C(CD3)3, R2 is -CH3 and R3 is CD3.
9. The compound of any one of claims 1-6, wherein R1 is -C(CD3)3, R2 is -CD3 and R3 is CH3.
10. The compound of any one of claims 1-6, wherein R1 is -C(CD3)3, R2 is -CH3 and R3 is CH3.
1 1. The compound of any one of claims 1-6, wherein R1 is -C(CH3)3, R2 is -CD3 and R3 is CD3.
12. The compound of any one of claims 1-6, wherein R1 is -C(CH3)3, R2 is -CH3 and R3 is CD3.
13. The compound of any one of claims 1-6, wherein R1 is -C(CH3)3, R2 is -CD3 and R3 is CH3.
14. The compound of any one of claims 1 -6, wherein Y1 and Y2 are the same; Y4, Y5, Y6, Y7, Y8 and Y9 are the same; and the compound is selected from any one of the compounds (Cmpd) set forth in Table 1 (below):
Figure imgf000027_0001
Figure imgf000028_0001
Compound No. R1 R2 R3 Y'/Y2 Y3 γ4_γ9
163 C(CH3)3 CH3 CH3 D D D or a pharmaceutically acceptable salt thereof, wherein any atom not designated as deuterium is present at its natural isotopic abundance.
15. The compound of any one of claims 1-14, wherein any atom not designated as deuterium is present at its natural isotopic abundance.
16. The compound of any one of claims 1-15, wherein the compound is a monosodium salt.
17. A pharmaceutical composition comprising a compound of any one of claims 1-16; and a pharmaceutically acceptable carrier.
18. The pharmaceutical composition of claim 17, wherein the composition is formulated for oral administration.
19. The pharmaceutical composition of claim 18, wherein the composition is a tablet.
20. The pharmaceutical composition of any one of claims 17-19, wherein the composition additionally comprises one or more second therapeutic agents selected from a HCV NS3/4A protease inhibitor, a HCV NS5A polymerase inhibitor, and a cytochrome P450 inhibitor.
21. The pharmaceutical composition of claim 20, wherein the one or more second therapeutic agents is a, combination of paritaprevir, ombitasvir, and optionally ritonavir.
22. A packaged pharmaceutical composition comprising:
a. a first oral dosage form comprising a compound of any one of claims 1-16; and b. a separate, second oral dosage form comprising a combination of paritaprevir, ombitasvir, and ritonavir.
23. The packaged pharmaceutical composition of claim 22, further comprising a separate third oral dosage form comprising a compound of any one of claims 1-16, wherein the first and third dosage forms are identical; and instructions for when during the day to take each of the first, second and third dosage forms.
24. A method of treating a HCV infection comprising the step of administering to a subject in need thereof a pharmaceutical composition of any one of claims 17 to 23.
PCT/US2015/000409 2014-12-24 2015-12-24 Deuterated dasabuvir WO2016105547A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201462096676P 2014-12-24 2014-12-24
US62/096,676 2014-12-24

Publications (1)

Publication Number Publication Date
WO2016105547A1 true WO2016105547A1 (en) 2016-06-30

Family

ID=55236890

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/000409 WO2016105547A1 (en) 2014-12-24 2015-12-24 Deuterated dasabuvir

Country Status (1)

Country Link
WO (1) WO2016105547A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017202207A1 (en) * 2016-05-27 2017-11-30 深圳市塔吉瑞生物医药有限公司 Substituted naphthalene ring compound, pharmaceutical composition, and application thereof
WO2020029912A1 (en) * 2018-08-10 2020-02-13 广东东阳光药业有限公司 Disodium salt crystal form c of hcv inhibitor and preparation method therefor

Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4454341A (en) 1983-03-03 1984-06-12 Sri International Naphthyl or tetrahydronaphthyl-substituted naphthoic acid and derivatives
US6803031B2 (en) 2001-05-24 2004-10-12 Alexza Molecular Delivery Corporation Delivery of erectile dysfunction drugs through an inhalation route
US7014866B2 (en) 2001-05-03 2006-03-21 Hoffmann-La Roche Inc. High dose solid unit oral pharmaceutical dosage form of amorphous nelfinavir mesylate and process for making same
US20060079502A1 (en) 1999-11-02 2006-04-13 Steffen Lang Pharmaceutical compositions
US20060094744A1 (en) 2004-09-29 2006-05-04 Maryanoff Cynthia A Pharmaceutical dosage forms of stable amorphous rapamycin like compounds
WO2009039127A1 (en) 2007-09-17 2009-03-26 Abbott Laboratories Uracil or thymine derivative for treating hepatitis c
WO2009039134A1 (en) 2007-09-17 2009-03-26 Abbott Laboratories Anti-infective pyrimidines and uses thereof
WO2011109274A1 (en) * 2010-03-01 2011-09-09 Concert Pharmaceuticals Inc. Fluorouracil derivatives
WO2012009699A2 (en) 2010-07-16 2012-01-19 Abbott Laboratories Process for preparing antiviral compounds
WO2014031791A1 (en) 2012-08-21 2014-02-27 Abbvie Inc. Process for preparing antiviral compounds
US20140219958A1 (en) * 2013-02-01 2014-08-07 Enanta Pharmaceuticals, Inc. 5, 6-d2 uridine nucleoside/tide derivatives

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4454341A (en) 1983-03-03 1984-06-12 Sri International Naphthyl or tetrahydronaphthyl-substituted naphthoic acid and derivatives
US20060079502A1 (en) 1999-11-02 2006-04-13 Steffen Lang Pharmaceutical compositions
US7014866B2 (en) 2001-05-03 2006-03-21 Hoffmann-La Roche Inc. High dose solid unit oral pharmaceutical dosage form of amorphous nelfinavir mesylate and process for making same
US6803031B2 (en) 2001-05-24 2004-10-12 Alexza Molecular Delivery Corporation Delivery of erectile dysfunction drugs through an inhalation route
US20060094744A1 (en) 2004-09-29 2006-05-04 Maryanoff Cynthia A Pharmaceutical dosage forms of stable amorphous rapamycin like compounds
WO2009039127A1 (en) 2007-09-17 2009-03-26 Abbott Laboratories Uracil or thymine derivative for treating hepatitis c
WO2009039134A1 (en) 2007-09-17 2009-03-26 Abbott Laboratories Anti-infective pyrimidines and uses thereof
WO2011109274A1 (en) * 2010-03-01 2011-09-09 Concert Pharmaceuticals Inc. Fluorouracil derivatives
WO2012009699A2 (en) 2010-07-16 2012-01-19 Abbott Laboratories Process for preparing antiviral compounds
WO2014031791A1 (en) 2012-08-21 2014-02-27 Abbvie Inc. Process for preparing antiviral compounds
US20140219958A1 (en) * 2013-02-01 2014-08-07 Enanta Pharmaceuticals, Inc. 5, 6-d2 uridine nucleoside/tide derivatives

Non-Patent Citations (28)

* Cited by examiner, † Cited by third party
Title
"Drugs and the Pharmaceutical Sciences", 2007, INFORMA HEALTHCARE, article "Oral Lipid-Based Formulations: Enhancing the Bioavailability of Poorly WaterSoluble Drugs"
"Encyclopedia of Reagents for Organic Synthesis", 1995, JOHN WILEY AND SONS
"PDR Pharmacopoeia, Tarascon Pocket Pharmacopoeia 2000", 2000, TARASCON PUBLISHING
"PDR Pharmacopoeia, Tarascon Pocket Pharmacopoeia 2000", 2000, TARASCON PUBLISHING, LOMA LINDA, CALIF.
"Pharmacotherapy Handbook", 2000, APPLETON AND LANGE, STAMFORD, CONN
"Pharmacotherapy Handbook", 2000, APPLETON AND LANGE, STAMFORD, CONN.
"Role of Lipid Excipients in Modifying Oral and Parenteral Drug Delivery: Basic Principles and Biological Example", 2006, WILEY-INTERSCIENCE
"Scientific Tables, Geigy Pharmaceuticals", 1970, ARDSLEY, N.Y., pages: 537
BLAKE, MI ET AL., J PHARM SCI, vol. 64, 1975, pages 367 - 91
CHAUDHURI, B. ET AL., INDUSTRIAL & ENGINEERING CHEMISTRY RESEARCH, vol. 30, no. 1, 1991, pages 227 - 31
DATABASE CAPLUS [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; GENTILE, IVAN ET AL: "Dasabuvir: A Non-Nucleoside Inhibitor of NS5B for the Treatment of Hepatitis C Virus Infection", XP002754504, retrieved from STN Database accession no. 2014:1759955 *
FIESER, L ET AL.: "Fieser and Fieser's Reagents for Organic Synthesis", 1994, JOHN WILEY AND SONS
FISHER, MB ET AL., CURR OPIN DRUG DISCOV DEVEL, vol. 9, 2006, pages 101 - 09
FOSTER, AB, ADV DRUG RES, vol. 14, 1985, pages 1 - 40
FREIREICH ET AL., CANCER CHEMOTHER. REP, vol. 50, 1966, pages 219
FUKUTO ET AL., J. MED. CHEM., vol. 34, 1991, pages 2871 - 76
GANNES, LZ ET AL., COMP BIOCHEM PHYSIOL MOL INTEGR PHYSIOL, vol. 119, 1998, pages 725
GENTILE, IVAN ET AL: "Dasabuvir: A Non-Nucleoside Inhibitor of NS5B for the Treatment of Hepatitis C Virus Infection", REVIEWS ON RECENT CLINICAL TRIALS , 9(2), 115-123 CODEN: RRCTB2; ISSN: 1574-8871, 2014, DOI: 10.2174/1574887109666140529222602 10.2174/1574887109666140529222602 *
GREENE, TW ET AL.: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY AND SONS
KEMPF, D.J. ET AL., ANTIMICROBIAL AGENTS AND CHEMOTHERAPY, vol. 41, no. 3, 1997, pages 654 - 60
KUSHNER, DJ ET AL., CAN J PHYSIOL PHARMACOL, 1999, pages 79 - 88
LAROCK R: "Comprehensive Organic Transformations", 1989, VCH PUBLISHERS
REMINGTON: "The Science and Practice of Pharmacy", 2000, LIPPINCOTT WILLIAMS & WILKINS, BALTIMORE, MD
SCHAEFER, J. ET AL., JOURNAL OF ORGANIC CHEMISTRY, vol. 32, no. 5, 1967, pages 1607 - 8
SCOTT L. HARBESON ET AL: "Deuterium Medicinal Chemistry: A New Approach to Drug Discovery and Development", MEDCHEM NEWS, no. 2, 1 May 2014 (2014-05-01), pages 8 - 22, XP055246294 *
SHIMIZU, H. ET AL., TETRAHEDRON LETTERS, vol. 47, no. 33, 2006, pages 5927 - 5931
WADA, E ET AL., SEIKAGAKU, vol. 66, 1994, pages 15
WANG, L ET AL., CLINICAL PHARMACOLOGY AND THERAPEUTICS, vol. 56, no. 6, 1994, pages 659 - 67

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017202207A1 (en) * 2016-05-27 2017-11-30 深圳市塔吉瑞生物医药有限公司 Substituted naphthalene ring compound, pharmaceutical composition, and application thereof
CN108290844A (en) * 2016-05-27 2018-07-17 深圳市塔吉瑞生物医药有限公司 A kind of substituted naphthalene cycle compound and pharmaceutical composition and its application
CN108290844B (en) * 2016-05-27 2021-04-30 深圳市塔吉瑞生物医药有限公司 Substituted naphthalene ring compound, pharmaceutical composition and application thereof
WO2020029912A1 (en) * 2018-08-10 2020-02-13 广东东阳光药业有限公司 Disodium salt crystal form c of hcv inhibitor and preparation method therefor
CN112513019A (en) * 2018-08-10 2021-03-16 广东东阳光药业有限公司 HCV inhibitor disodium salt crystal form C and preparation method thereof
CN112513019B (en) * 2018-08-10 2022-09-27 广东东阳光药业有限公司 HCV inhibitor disodium salt crystal form C and preparation method thereof

Similar Documents

Publication Publication Date Title
AU2013296627B2 (en) Deuterated ibrutinib
AU2014235462C1 (en) Deuterated palbociclib
WO2012151361A1 (en) Carbamoylpyridone derivatives
WO2014011971A2 (en) Deuterated carfilzomib
WO2016168553A1 (en) Deuterated obeticholic acid
CA2897814A1 (en) Deuterated momelotinib
WO2011017612A1 (en) Substituted diphenylpyrazine derivatives
WO2018005328A1 (en) Deuterated bictegravir
WO2016179207A1 (en) Deuterated filgotinib
WO2011109274A1 (en) Fluorouracil derivatives
WO2010132810A1 (en) Deuterium modified benzimidazoles
WO2018013625A1 (en) Deuterated miconazole
WO2016105547A1 (en) Deuterated dasabuvir
EP3416629A1 (en) Deuterated gft-505
WO2012129381A1 (en) Deuterated preladenant
WO2012009446A1 (en) Novel pyrimidinecarboxamide derivatives
EP3265440A1 (en) Deuterated emricasan
EP2970213A1 (en) Deuterated pacritinib
WO2015009889A1 (en) Deuterated intedanib derivatives and their use for the treatment of proliferative disorders
WO2010132663A1 (en) Pegylated azapeptide derivatives as hiv protease inhibitors
EP2885267B1 (en) Deuterated idebenone
WO2014152275A1 (en) Deuterium modified derivatives of the ns5b polymerase inhibitor tmc647055
WO2011159920A1 (en) [5,6]-dihydro-2h-pyran-2-one derivatives
WO2011103457A1 (en) Pyrimidine derivatives
EP2678337A1 (en) 2-amino-naphthyridine derivatives

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 15828698

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 15828698

Country of ref document: EP

Kind code of ref document: A1