WO2008156632A1 - Azapeptide derivatives - Google Patents

Azapeptide derivatives Download PDF

Info

Publication number
WO2008156632A1
WO2008156632A1 PCT/US2008/007331 US2008007331W WO2008156632A1 WO 2008156632 A1 WO2008156632 A1 WO 2008156632A1 US 2008007331 W US2008007331 W US 2008007331W WO 2008156632 A1 WO2008156632 A1 WO 2008156632A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
composition
pharmaceutically acceptable
efavirenz
acceptable salt
Prior art date
Application number
PCT/US2008/007331
Other languages
English (en)
French (fr)
Inventor
Scott L. Harbeson
Roger D. Tung
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.
Priority to AU2008267048A priority Critical patent/AU2008267048C1/en
Priority to JP2010512186A priority patent/JP2010529196A/ja
Priority to MX2009013565A priority patent/MX2009013565A/es
Priority to CA2692028A priority patent/CA2692028C/en
Priority to CN2008800216013A priority patent/CN101711237B/zh
Priority to KR1020107000675A priority patent/KR101185899B1/ko
Priority to BRPI0813911-3A priority patent/BRPI0813911A2/pt
Publication of WO2008156632A1 publication Critical patent/WO2008156632A1/en
Priority to ZA2009/09079A priority patent/ZA200909079B/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/24Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D213/36Radicals substituted by singly-bound nitrogen atoms
    • C07D213/42Radicals substituted by singly-bound nitrogen atoms having hetero atoms attached to the substituent nitrogen atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4418Non condensed pyridines; Hydrogenated derivatives thereof having a carbocyclic group directly attached to the heterocyclic ring, e.g. cyproheptadine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • 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
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/24Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with substituted hydrocarbon radicals attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/553Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having one nitrogen atom as the only ring hetero atom
    • C07F9/576Six-membered rings
    • C07F9/58Pyridine rings

Definitions

  • Atazanavir sulfate also known as
  • HIV-I infected cells prevents the formation of mature HIV virions in HIV-I infected cells by selectively inhibiting the virus-specific processing of certain polyproteins (viral Gag and Gag-Pol). Atazanavir sulfate is currently approved for the treatment of HIV infection.
  • Atazanavir is contraindicated for coadministration with drugs that are highly dependent on CYP3 A for clearance and for which elevated plasma concentrations are associated with serious and/or life-threatening events. Due to inhibition effects of atazanavir on CYP3A, CYP2C8, and UGTlAl, caution is advised when prescribing drugs primarily metabolized by CYP3A, CYP2C8, or UGTlAl for patients receiving atazanavir. Common adverse events associated with atazanavir include hyperbilirubinemia, rash, nausea, headache, and jaundice/scleral icterus. Adverse events experienced by some patients and for which a causal relationship has not been established include diabetes mellitus/hyperglycemia, PR interval prolongation, hemophilia, and fat redistribution.
  • This invention relates to novel compounds that are azapeptides, and pharmaceutically acceptable salts thereof. More specifically, the invention relates to novel azapeptide compounds that are derivatives of the HIV protease inhibitor atazanavir sulfate. This invention also provides pyrogen- free compositions comprising one or more compounds of the invention and a carrier, and the use of the disclosed compounds and compositions in methods of treating diseases and conditions that are treated by administering HIV protease inhibitors. The invention also relates to the use of one or more of the disclosed compounds as reagents in analytical studies involving atazanavir.
  • each of R la and R lb is independently selected from C 1 -C 3 alkyl, wherein one or more hydrogen atoms in the alkyl is optionally replaced with a deuterium atom; each of R 2 and R 3 is independently selected from isopropyl, sec-butyl, and tert-butyl wherein one or more hydrogen atoms in the isopropyl, sec-butyl, or tert-butyl is optionally replaced with a deuterium atom;
  • R 4 is selected from H, OH and -O-(CR 6 R 7 -O) n -R 8 ;
  • R 5 is selected from H and -(CR 6 R 7 -O) n -R 8 , wherein:
  • R 6 and R 7 are each independently selected from H, Ci-C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, and C 3 -C 7 cycloalkyl, or
  • R 6 and R 7 are taken together with the carbon to which they are attached to form a 3-7-membered cycloalkyl; each R 8 is independently selected from -C(O)H, -C(O)-(Ci-C 7 alkyl), -P(O)-(OH) 2 , -S(O)-OH, -S(O) 2 -OH, and A-R 11 , wherein A is an ⁇ -amino acid residue; and
  • R 11 is selected from H, Ci-C 6 alkyl, -C(O)-(C-C 7 alkyl), A-R 12 , wherein R 12 is selected from H, Ci-C 6 alkyl, and -C(O)-(Ci-C 7 alkyl); and n is O or 1 ; wherein any alkyl in R 5 is optionally substituted; each of Y la and Y lb is independently selected from H and D; R 9 is selected from 2-thienyl, 3-thienyl, thiazol-5-yl, thiazol-2-yl, pyridin-2-yl, pyridin-3-yl, pyridin-4-yl, pyrazin-2-yl, 2-methyl-2H-tetrazol-5-yl, 2-(c?
  • the compounds, pharmaceutically acceptable salts thereof and compositions of the invention are useful for treating diseases that are effectively treated by a compound that is an HIV protease inhibitor.
  • the present invention includes a method of treating a disease which is susceptible to treatment by a compound that is an HIV protease inhibitor, comprising administering to a subject in need thereof an effective amount of: (i) a compound or pharmaceutically acceptable salt thereof; or (ii) a pyrogen-free composition (e.g., a pharmaceutical composition) described herein.
  • HIV protease inhibitory activity diseases or conditions susceptible to treatment with a compound having HIV protease inhibitory activity include, but are not limited to, HIV infection.
  • compositions of this invention are also useful as reagents in methods for determining the concentration of atazanavir sulfate in solution, examining the metabolism of atazanavir sulfate and other analytical studies.
  • An additional utility of compounds of any of the formulae herein include their use as internal standards to determine the true concentrations atazanavir sulfate in biological matrices, such as plasma.
  • FIG. 1 is a graph showing the relative stability of compounds of this invention in human liver microsomes as compared to atazanavir.
  • FIG. 2 is a graph showing the relative stability of compounds of this invention in human liver microsomes as compared to atazanavir.
  • FIG. 3 is a graph showing the relative stability of compounds of this invention in human liver microsomes as compared to atazanavir.
  • FIG. 4 is a graph showing the relative plasma levels of compounds of this invention following oral administration to chimps as compared to atazanavir.
  • FIG. 5 is a graph showing the relative plasma levels of compounds of this invention following oral administration to chimps as compared to atazanavir.
  • ameliorate and “treat” are used interchangeably and include both - - therapeutic treatment and prophylactic treatment (reducing the likelihood of development). Both terms mean 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.
  • H hydrogen
  • D deuterium
  • deuterium the position is understood to have deuterium at an abundance that is at least 3500 times greater than the natural abundance of deuterium, which is 0.015% (i.e., at least 52.5% incorporation of deuterium).
  • isotopic enrichment factor means the ratio between the isotopic abundance of D at a specified position in a compound of this invention and the naturally occurring abundance of that isotope.
  • the natural abundance of deuterium is 0.015%.
  • a compound of this invention has an isotopic enrichment factor for each deuterium present at a site designated as a potential site of deuteration on the compound of 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).
  • the isotopic enrichment factor of each deuterium present at a site designated as a site of deuteration is independent of other deuterated sites. For example, if there are two sites of deuteration on a compound one site could be deuterated at 52.5% while the other could be deuterated at 75%. The resulting compound would be considered to be a compound wherein the isotopic enrichment factor is at least 3500 (52.5%).
  • isotopologue refers to a species that differs from a specific compound of this invention only in the isotopic composition thereof. Isotopologues can differ in the level of isotopic enrichment at one or more positions and/or in the positions(s) of isotopic enrichment.
  • the term "compound,” when referring to the compounds of the 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.
  • 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.
  • 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, -O- 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
  • 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-l,4-dioate, hexyne-l,6-dioate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, phthalate, terephthalate, sulfonate, xylene sulfonate, phenylacetate, phenylprop
  • suitable cationic moieties to form pharmaceutically acceptable salts include, but are not limited to, alkali metals such as sodium, potassium, and lithium; alkaline earth metals such as calcium and magnesium; other metals, such as aluminum and zinc; ammonia, and organic amines, such as mono-, di-, or trialkylamines; dicyclohexylamine; tributyl amine; pyridine; N-methyl,N-ethylamine; diethylamine; triethylamine; mono-, bis-, or tris-(2-hydroxy-lower alkyl amines), such as mono-, bis-, or tris-(2-hydroxyethyl)amine, 2-hydroxy-tert-butylamine, or tris-(hydroxymethyl)methylamine, N, N,-di-lower alkyl-N
  • hydrate means a compound which further includes a stoichiometric or non-stoichiometric amount of water bound by non-covalent intermolecular forces.
  • solvate means a compound which further includes a stoichiometric or non-stoichiometric amount of solvent such as water, acetone, ethanol, methanol, dichloromethane, 2-propanol, or the like, bound by non-covalent intermolecular forces.
  • solvent such as water, acetone, ethanol, methanol, dichloromethane, 2-propanol, or the like.
  • the disclosed compounds may exist in various stereoisomeric forms. Stereoisomers are compounds which differ only in their spatial arrangement. Enantiomers are pairs of stereoisomers whose mirror images are not superimposable, most commonly because they contain an asymmetrically substituted carbon atom that acts as a chiral center. "Enantiomer” means one of a pair of molecules that are mirror images of each other and are not superimposable.
  • Diastereomers are stereoisomers that are not related as mirror images, most commonly because they contain two or more asymmetrically substituted carbon atoms. "R” and “S” represent the configuration of substituents around one or more chiral carbon atoms. [31] When the stereochemistry of the disclosed compounds is named or depicted by structure, the named or depicted stereoisomer is at least 60%, 70%, 80%, 90%, 99% or 99.9% by weight pure relative to the other stereoisomers. When a single enantiomer is named or depicted by structure, the depicted or named enantiomer is at least 60%, 70%, 80%, 90%, 99% or 99.9% optically pure.
  • Percent optical purity by weight is the ratio of the weight of the enantiomer over the weight of the enantiomer plus the weight of its optical isomer.
  • substantially free of other stereoisomers 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, or less than "X"% of other stereoisomers (wherein X is a number between 0 and 100, inclusive) are present.
  • 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., -o- 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).
  • halo refers to any of -Cl, -F, -Br, or -I.
  • alkoxy refers to -O-alkyl
  • alkylamino refers to -NH-alkyl.
  • dialkylamino refers to N(alkyl)-alkyl, wherein the two alkyl moieties are the same or different.
  • alkyl refers to straight or branched alkyl chains of from 1 to 12 carbon atoms, preferably from 1 to 8 carbon atoms, more preferably from 1 to 4 carbon atoms unless otherwise specified.
  • straight chained and branched alkyl groups include methyl, ethyl, n-propyl, iso-propyl, n-butyl, sec-butyl, tert-butyl, pentyl, hexyl, heptyl and octyl.
  • Alkyl may be optionally substituted.
  • Alkyl or aryl groups that are optionally substituted will typically contain one to four substituents that are independently selected.
  • substituents include Ci -7 alkyl, halo, cyano, hydroxyl, carboxy, alkoxy, oxo, amino, alkylamino, dialkylamino, cycloheteroalkyl, alkylcycloheteroalkyl, aryl, alkylaryl, heteroaryl, and alkylheteroaryl.
  • cycloheteroalkyl refers to a non-aromatic monocyclic, bicyclic, tricyclic, spirocyclic, or tetracyclic ring system which includes one or more heteroatoms such as nitrogen, oxygen or sulfur in at least one of the rings.
  • Each ring can be four, five, six, seven or eight-membered. Examples include tetrahydrofuryl, tetrahydrothiophenyl, morpholino, thiomorpholino, pyrrolidinyl, piperazinyl, piperidinyl, and thiazolidinyl, along with the cyclic form of sugars.
  • alkylcycloheteroalkyl refers to a cycloheteroalkyl group comprising an alkyl substituent. Examples include 4-methylpiperazin-l-yl and 4-methylpiperidin-l-yl.
  • aryl refers carbocyclic aromatic groups such as phenyl and naphthyl.
  • alkylaryl refers to an aryl group linked to the rest of the molecule through an alkyl chain.
  • heteroaryl refers to monocyclic aromatic groups comprising one or more heteroatoms such as nitrogen, oxygen or sulfur in the ring, such as imidazolyl, thienyl, furyl, pyridyl, pyrimidyl, pyranyl, pyrazolyl, pyrrolyl, pyrazinyl, thiazolyl, oxazolyl, and tetrazolyl.
  • Heteroaryl groups also include fused polycyclic aromatic ring systems in which at least one ring comprises one or more heteroatoms such as nitrogen, oxygen or sulfur.
  • heteroatoms such as nitrogen, oxygen or sulfur.
  • examples include benzothienyl, benzofuryl, indolyl, quinolinyl, benzothiazole, benzoxazole, benzimidazole, quinolinyl, isoquinolinyl and isoindolyl.
  • alkylheteroaryl refers to a heteroaryl group linked to the rest of the molecule through an alkyl chain.
  • ⁇ -amino acid residue refers to a group of the general formula
  • ⁇ -amino acid includes ⁇ -amino acids having a
  • variable R 8 is an ⁇ -amino acid, it is linked to the rest of the molecule through the carbonyl carbon directly bonded to the ⁇ -carbon of the amino acid. In accordance with the structure of Formula I, such a linkage results in the formation of an ester
  • variable may be referred to generally (e.g., "each R") or 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.
  • each of R la and R lb is independently selected from Ci-C 3 alkyl, wherein one or more hydrogen atoms in the alkyl is optionally replaced with a deuterium atom; each of R 2 and R 3 is independently selected from isopropyl, sec-butyl, and tert-butyl wherein one or more hydrogen atoms in the isopropyl, sec-butyl, or tert-butyl is optionally replaced with a deuterium atom;
  • R 4 is selected from H, OH and -O-(CR 6 R 7 -O) n -R 8
  • R 5 is selected from H and -(CR 6 R 7 -O) n -R 8 , wherein:
  • R 6 and R 7 are each independently selected from H, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, and C 3 -C 7 cycloalkyl, or
  • R 6 and R 7 are taken together with the carbon to which they are attached to form a 3-7-membered cycloalkyl; each R 8 is independently selected from -C(O)H, -C(O)-(C 1 -C 7 alkyl), -P(O)-(OH) 2 , -S(O)-OH, -S(O) 2 -OH, and A-R 11 , wherein A is an ⁇ -amino acid residue; and
  • R 11 is selected from H, C 1 -C 6 alkyl, -C(O)-(C 1 -C 7 alkyl), A-R 12 , wherein R 12 is selected from H, C 1 -C 6 alkyl, and -C(O)-(Ci-C 7 alkyl); and n is O or 1 ; wherein any alkyl in R 5 is optionally substituted; each of Y la and Y lb is independently selected from H and D; R 9 is selected from 2-thienyl, 3-thienyl, thiazol-5-yl, thiazol-2-yl, pyridin-2-yl, pyridin-3-yl, pyridin-4-yl, pyrazin-2-yl, 2-methyl-2H-tetrazol-5-yl, 2-(c?j-methyl)-2H-tetrazol-5-yl, 1 -methyl- lH-tetrazol-5-yl, and 1 -t ⁇ -methyl)-
  • R 2 and R 3 comprise a deuterium atom
  • each of R 2 and R 3 is independently selected from -C(CH 3 ) 3 , -C(CD 3 ) 3 , -CH(CH 3 ) 2 , -CD(CD 3 ) 2 , -CH 2 CH 2 (CH 3 ) 2 , and -CD 2 CD 2 (CD 3 );
  • each of R la and R lb is independently selected from -CH 3 , -CD 3 , -CH 2 CH 3 , -CD 2 CD 3 , -CD 2 CD 2 CD 3 , and -CH 2 CH 2 CH 3
  • R 5 is H, P(O)-(OH) 2 , -CH 2 -O-P(O)-(OH) 2 , or a pharmaceutically acceptable salt thereof
  • each of R la and R lb is independently selected from CH 3 , CH 2 D, CHD 2 , and CD 3 ; each of R 2 and R 3 is independently -C(CH 3 ) 3 , wherein from 1 to 9 hydrogen atoms are optionally replaced with a deuterium atom;
  • R 4 is selected from H, OH and -O-(CR 6 R 7 -O) n -R 8
  • R 5 is selected from H and -(CR 6 R 7 -O) n -R 8
  • R 6 and R 7 are independently selected from H and Ci-C 3 alkyl
  • each R is independently selected from an ⁇ -amino acid, -C(O)H, -C(O)-(C 1 -C 7 alkyl), wherein said Ci-C 7 alkyl is optionally substituted, -P(O)-(OH) 2 , and -S(O)-OH
  • n is O or 1 ;
  • Y la and Y lb are independently selected from H and D; and at least one of R la , R lb , R 2 , R 3 or Y variable comprises a deuterium atom.
  • Specific embodiments of Formula I include a compound wherein: i. each of R la and R lb is independently selected from CH 3 and CD 3 ; ii. each of R 2 and R 3 is independently selected from -C(CH 3 ) 3 and -C(CD 3 ) 3 ; iii. R 2 is -C(CD 3 ) 3 ; iv. Y la and Y lb are the same; v. each of Y la and Y lb is deuterium; vi.
  • R 4 is selected from H and -O-(CR 6 R 7 -O) n -R 8 ; vii. R 4 and R 5 are simultaneously H; viii. each R 6 and each R 7 is H; ix. each R 8 is independently selected from an ⁇ -amino acid having (L)- configuration; -C(O)H; -C(O)-(Cj-C 3 alkyl), wherein said C]-C 3 alkyl is optionally substituted with cyano, hydroxyl, carboxy, alkoxy, amino, alkylamino, dialkylamino, cycloheteroalkyl, alkyl cycloheteroalkyl, aryl, alkylaryl, heteroaryl, and alkylheteroaryl; -P(O)-(OH) 2 ; a salt of -P(O)-(OH) 2 , wherein the cation is selected from Na + , Mg 2+ , or ammonium; -S(
  • each R 8 is independently selected from L-Serine; L-Lysine; L-Tyrosine; L- Valine; L-Glutamic acid; L-Aspartic acid; L-3-Pyridylalanine; L-Histidine; -C(O)H; -C(O)-(C 1 -C 3 alkyl); -C(O)CH 2 OCH 3 ; -C(O)CH 2 CH 2 OCH 3 ; -C(O)CH 2 CH 2 C(O)OH; -C(O)CH 2 CH 2 NH 2 ;
  • Example embodiments where two or more of the above parameters are met include, but are not limited to, the following particular embodiments.
  • R 2 and R 3 are independently selected from -C(CH 3 ) 3 and -C(CD 3 ) 3 , and R la and R lb are independently selected from CH 3 and CD 3 .
  • R 2 is -C(CD 3 ) 3
  • R la is CD 3
  • R 2 is -C(CD 3 ) 3
  • R la is CD 3
  • R lb is CD 3 .
  • Y la and Y lb are the same (i.e., both are simultaneously deuterium or simultaneously H), and either R la and R lb are independently selected from CH 3 and CD 3 , or R 2 and R 3 are independently selected from -C(CH 3 ) 3 and -C(CD 3 ) 3 .
  • Y la and Y lb are the same (e.g., both are deuterium), R la and R lb are independently selected from CH 3 and CD 3 , and R 2 and R 3 are independently selected from -C(CH 3 ) 3 and -C(CD 3 ) 3 .
  • R 4 is selected from H and -O-(CR 6 R 7 -O) n -R 8 , and either R la and R lb are independently selected from CH 3 and CD 3 , or R and R 3 are independently selected from -C(CH 3 ) 3 and -C(CD 3 ) 3 .
  • R 4 is selected from H and -O-(CR 6 R 7 -O) n -R 8 , R la and R lb are independently selected from CH 3 and CD 3 , and R 2 and R 3 are independently selected from -C(CH 3 ) 3 and -C(CD 3 ) 3 .
  • R 4 is selected from H and -O-(CR 6 R 7 -O) n -R 8 , and Y la and Y lb are the same. In a more particular embodiment, R 4 is selected from H and -O-(CR 6 R 7 -O) n -R 8 , and Y la and Y lb are deuterium.
  • R 4 is selected from H and -O-(CR 6 R 7 -O) n -R 8 , Y la and Y lb are the same (i.e., both are simultaneously deuterium or simultaneously H), and either R la and R lb are independently selected from CH 3 and CD 3 or R 2 and R 3 are independently selected from -C(CH 3 ) 3 and -C(CD 3 ) 3 .
  • R 4 is selected from H and -O-(CR 6 R 7 -O) n -R 8 , Y la and Y lb are the same (i.e., both are simultaneously deuterium or simultaneously H), R la and R are independently selected from CH 3 and CD 3 , and R and R are independently selected from -C(CH 3 ) 3 and -C(CD 3 ) 3 .
  • R 6 and R 7 are each H, either R la and R lb are independently selected from CH 3 and CD 3 , or R 2 and R 3 are independently selected from -C(CH 3 ) 3 and -C(CD 3 ) 3 .
  • R 6 and R 7 are each H, R la and R lb are independently selected from CH 3 and CD 3 , and R 2 and R 3 are independently selected from -C(CH 3 ) 3 and -C(CD 3 ) 3 .
  • R 6 and R 7 are each H, and Y la and Y lb are the same. In a more particular embodiment, R 6 and R 7 are each H, and Y la and Y lb are deuterium. In an even more particular embodiment, R 6 and R 7 are each H, Y la and Y lb are the same (i.e.., both are simultaneously deuterium or simultaneously H), and either R la and R lb are independently selected from CH 3 and CD 3 , or R 2 and R 3 are independently selected from -C(CH 3 ) 3 and -C(CD 3 ) 3 .
  • R 6 and R 7 are each H, Y la and Y lb are the same (e.g., both are deuterium), R la and R lb are independently selected from CH 3 and CD 3 , and R 2 and R 3 are independently selected from -C(CH 3 ) 3 and -C(CD 3 ) 3 .
  • R 6 and R 7 are each H, and R 4 is selected from H and -O-(CR 6 R 7 -O) n -R 8 .
  • R 6 and R 7 are each H, R 4 is selected from H and -O-(CR 6 R 7 -O) n -R 8 ,and either R la and R lb are independently selected from CH 3 and CD 3 or R 2 and R 3 are independently selected from -C(CH 3 ) 3 and -C(CD 3 ) 3 .
  • R 6 and R 7 are each H, R 4 is selected from H and -O-(CR 6 R 7 -O) n -R 8 , R la and R lb are independently selected from CH 3 and CD 3 , and R 2 and R 3 are independently selected from -C(CH 3 ) 3 and -C(CD 3 ) 3 .
  • R 6 and R 7 are each H, R 4 is selected from H and -O-(CR 6 R 7 -O) n -R 8 , and Y la and Y lb are the same, hi a more particular embodiment, R 6 and each R 7 are each H, R 4 is selected from H and -0-(CR 6 R 7 O) n -R 8 , and Y la and Y lb are deuterium, hi an even more particular embodiment, R 6 and R 7 are each H, R 4 is selected - - from H and -O-(CR 6 R 7 -O) n -R 8 , Y la and Y lb are the same (i.e., both are simultaneously deuterium or simultaneously H), and either R la and R lb are independently selected from CH 3 and CD 3 , or R 2 and R 3 are independently selected from -C(CH 3 ) 3 and -C(CD 3 ) 3 .
  • R 6 and R 7 are each H
  • R 4 is selected from H and -O-(CR 6 R 7 -O) n -R 8
  • Y la and Y lb are the same (i.e.,both are simultaneously deuterium or simultaneously H)
  • R la and R lb are independently selected from CH 3 and CD 3
  • R 2 and R 3 are independently selected from -C(CH 3 ) 3 and -C(CD 3 ) 3 .
  • R 8 is independently selected from an ⁇ -amino acid having (L)- configuration; -C(O)H; -C(O)-(CpC 3 alkyl), wherein said Cj-C 3 alkyl is optionally substituted with cyano, hydroxyl, carboxy, alkoxy, amino, alkylamino, dialkylamino, cycloheteroalkyl, alkyl cycloheteroalkyl, aryl, alkylaryl, heteroaryl, and alkylheteroaryl; -P(O)-(OH) 2 ; a salt of -P(O)-(OH) 2 wherein the cation is selected from Na + , K + , or Ca 2+ ; -S(O)-OH; and a salt of -S(O)-OH wherein the cation is selected from Na + , K + , or Ca 2+ .
  • R 8 is independently selected from L-Serine; L-Lysine; L-Tyrosine; L- Valine; L-Glutamic acid; L-Aspartic acid; L-3-Pyridylalanine; L-Histidine; -C(O)H; -C(O)-(Ci-C 3 alkyl); -C(O)CH 2 OCH 3 ; -C(O)CH 2 CH 2 OCH 3 ; -C(O)CH 2 CH 2 C(O)OH; -C(O)CH 2 CH 2 NH 2 ;
  • R 4 and R 5 are simultaneously H, and either R la and R Ib are independently selected from CH 3 and CD 3j or R 2 and R 3 are independently selected from -C(CH 3 ) 3 and -C(CD 3 ) 3 .
  • R 4 and R 5 are simultaneously H, R la and R 1 are independently selected from CH 3 and CD 3 , and R 2 and R 3 are independently selected from -C(CH 3 ) 3 and -C(CD 3 ) 3 .
  • R 4 and R 5 are simultaneously H, and Y la and Y lb are the same.
  • R 4 and R 5 are simultaneously H, and Y la and Y lb are simultaneously deuterium
  • R 4 and R 5 are simultaneously H, Y la and Y lb are the same (i.e., both are simultaneously deuterium or - - simultaneously H) and either R la and R lb are independently selected from CH 3 and CD 3 , or R 2 and R 3 are independently selected from -C(CHa) 3 and -C(CD 3 ) 3 .
  • R 4 and R 5 are simultaneously H
  • Y la and Y lb are the same (i.e., both are simultaneously deuterium or simultaneously H)
  • R la and R lb are independently selected from CH 3 and CD 3
  • R 2 and R 3 are independently selected from -C(CH 3 ) 3 and -C(CD 3 ) 3
  • the compound is a compound of the Formula Ia:
  • the compound is a compound of the Formula Ib:
  • each of R la and R lb is independently selected from -CD 3 and -CH 3 ;
  • R 3 is selected from -C(CD 3 ) 3 and -C(CH 3 ) 3 ;
  • Y la and Y lb are the same and are selected from H and D. - -
  • each of R la and R lb is independently selected from -CD 3 and -CH 3 ;
  • R 3 is selected from -C(CD 3 ) 3 and -C(CH 3 ) 3 ;
  • R 5 is -P(O)-(OH) 2 , -CH 2 -P(O)-(OH) 2 , or a pharmaceutically acceptable salt of either of the foregoing;
  • Y la and Y lb are the same and are selected from H and D.
  • the compound of this invention is selected from the following:
  • the compound of the invention is selected from the following:
  • the compound is selected from Compound 114, Compound 120, Compound 122 and Compound 131. - -
  • any atom not designated as deuterium in any of the embodiments set forth above is present at its natural isotopic abundance.
  • the synthesis of compounds of Formula I can be readily achieved by synthetic chemists of ordinary skill. Relevant procedures and intermediates are disclosed, for instance, in United States Patent 5,849,911; PCT Intl Publication WO 97/46514; Bold, G et al., J Med Chem 1998, 41 :3387; Xu, Z et al., Org Process Res Dev 2002, 6:323; and PCT Intl Publication WO 2006/014282.
  • 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.
  • Certain intermediates can be used with or without purification (e.g., filtration, distillation, sublimation, crystallization, trituration, solid phase extraction, and chromatography).
  • R 2 R 3 .
  • Aldehyde X is treated with the commercially available t-butoxycarbonylhydrazide (XI) to produce a BOC-protected hydrazone intermediate XII, which is then reduced using either hydrogen or deuterium gas to form the appropriate BOC-protected hydrazide XIII.
  • the BOC-protected hydrazide XIII is then treated with the commercially available epoxide (XTV) to produce XV, which is then deprotected with hydrochloric acid to produce XVI.
  • the undeuterated aldehyde X may be oxidized to the carboxylic acid, converted to the Weinreb amide via the acyl chloride, and reduced with LiAlD 4 to afford the desired deuterated aldehyde as set forth below in Scheme 2b.
  • tert-leucine XXIII wherein R 2 and/or R 3 are -C(CDa) 3 , may be prepared starting from the commercially available dp-pivalic acid (XX).
  • XX is reduced to the alcohol XXI with lithium aluminum hydride as described in Brainard, RL et al., Organometallics 1986, 5:1481-1490.
  • This alcohol XXI is oxidized to the aldehyde XXII by any one of a number of mild conditions (see, for example, Herrerias, CI et al., Tet Lett 2005, - -
  • XXIIc is hydrolyzed with hydrochloric acid to produce the corresponding carboxylic acid (XXV), which is then reacted with a deuterated methyl chloroformate in the presence of NaOH to produce the deuterated intermediate XVII.
  • Prodrugs of the invention represented by Formula A where R 5 is -(CR 6 R 7 -O) n -R 8 , wherein: R 6 and R 7 are H and each R 8 is -P(O)-(OH) 2 or a salt thereof, may be prepared according to the procedures of Safadi, M et al., Pharmaceutical Research, 1993, 10(9): 1350. Other suitable methods for preparing prodrugs of the compounds of the invention can be found in PCT Intl Publication WO 2006/014282.
  • a compound of this invention has at least 52.5% deuterium incorporation, at least 60% deuterium incorporation, at least 67.5% deuterium incorporation, at least 75% deuterium incorporation, at least 82.5% deuterium incorporation, at least 90% deuterium incorporation, or at least 95% deuterium incorporation at each position designated as deuterium in a compound of this invention.
  • the compound of the invention may be in an amount of, for example, at least 1 OOmg, such as at least 200mg, preferably at least 400mg, more preferably at least 5 OOmg and optionally up to 10Kg.
  • reaction schemes and protocols may be determined by the skilled artisan by use of commercially available structure-searchable database software, for instance, SciFinder® (CAS division of the American Chemical Society), STN® (CAS division of the American Chemical Society), CrossFire Beilstein® (Elsevier MDL), or internet search engines such as Google® or keyword databases such as the US Patent and Trademark Office text database.
  • SciFinder® CAS division of the American Chemical Society
  • STN® CAS division of the American Chemical Society
  • CrossFire Beilstein® Elsevier MDL
  • internet search engines such as Google® or keyword databases such as the US Patent and Trademark Office text database.
  • the methods described herein may also additionally include steps, either before or after the steps described specifically herein, to add or remove suitable protecting groups in order to ultimately allow synthesis of the compounds herein.
  • various synthetic steps may be performed in an alternate sequence or order to give the desired compounds.
  • the invention also provides pyrogen-free compositions comprising an effective amount of a compound of any one of Formulae A, I, Ia, Ib or Ic (e.g., including any of the formulae herein), or a pharmaceutically acceptable salt of said compound; and an acceptable carrier.
  • a composition of this invention is formulated for pharmaceutical use ("a pharmaceutical composition"), wherein the carrier is 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.
  • 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 phosphat
  • 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 7,014,866; and United States patent publications 20060094744 and 20060079502.
  • compositions of the invention include those suitable for oral, rectal, nasal, topical (including buccal and sublingual), pulmonary, 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's Pharmaceutical Sciences, Mack Publishing Company, Philadelphia, PA (17th ed. 1985).
  • 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.
  • 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.
  • 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.
  • 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 non-aqueous 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.
  • 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 iri 1,3-butanediol.
  • suitable 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.
  • the pharmaceutical compositions of this invention may be administered in the form of suppositories for rectal administration.
  • 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.
  • the patient therapeutics may be local, so as to be administered at the site of interest.
  • Various techniques can be used for providing the patient compositions at the site of interest, such as injection, use of catheters, trocars, projectiles, pluronic gel, stents, sustained drug release polymers or other device which provides for internal access.
  • the compounds of this invention may be incorporated into compositions for coating an implantable medical device, such as prostheses, artificial valves, vascular grafts, stents, or catheters. Suitable coatings and the general preparation of coated implantable devices are known in the art and are exemplified in US Patents 6,099,562; 5,886,026; and 5,304,121.
  • the coatings are typically biocompatible polymeric materials such as a hydrogel polymer, polymethyldisiloxane, polycaprolactone, polyethylene glycol, polylactic acid, ethylene vinyl acetate, and mixtures thereof.
  • the coatings may optionally be further covered by a suitable topcoat of fluorosilicone, polysaccharides, polyethylene glycol, phospholipids or combinations thereof to impart controlled release characteristics in the composition.
  • Coatings for invasive devices are to be included within the definition of pharmaceutically acceptable carrier, adjuvant or vehicle, as those terms are used herein.
  • the invention provides a method of coating an implantable medical device comprising the step of contacting said device with the coating composition described above. It will be obvious to those skilled in the art that the coating of the device will occur prior to implantation into a mammal.
  • the invention provides a method of impregnating an implantable drug release device comprising the step of contacting said drug release device with a compound or composition of this invention.
  • Implantable drug release devices include, but are not limited to, biodegradable polymer capsules or bullets, non-degradable, diffusible polymer capsules and biodegradable polymer wafers.
  • the invention provides an implantable medical device coated with a compound or a composition comprising a compound of this invention, such that said compound is therapeutically active.
  • the invention provides an implantable drug release device impregnated with or containing a compound or a composition comprising a compound of this invention, such that said compound is released from said device and is therapeutically active.
  • composition of this invention may be painted onto the organ, or a composition of this invention may be applied in any other convenient way.
  • a composition of this invention further comprises a second therapeutic agent.
  • the second therapeutic agent is one or more additional compounds of the invention.
  • each of the two or more compounds of the invention present in such compositions differs from all others in the positions of isotopic enrichment. Commonly, such a composition comprises three, four, five or more different compounds of this invention.
  • the second therapeutic agent may be selected from any compound or therapeutic agent known to have or that demonstrates advantageous properties when administered with a compound having the same mechanism of action as atazanavir.
  • Such agents include those indicated as being useful in combination with atazanavir, including but not limited to, those described in PCT publications WO 2003020206, WO 2005058248,
  • the second therapeutic agent is an agent useful in the treatment or prevention of HIV infection (i.e., an antiretroviral agent).
  • the second therapeutic agent is selected from other anti-retroviral agents including, but not limited to, a second HIV protease inhibitor (e.g., amprenavir, _
  • NRTI non-nucleoside reverse transcriptase inhibitor
  • NRTI nucleoside/nucleotide reverse transcriptase inhibitor
  • zidovudine lamivudine, emtricitabine, tenofovir disoproxil fumarate, didanosine, stavudine, abacavir, racivir, amdoxovir, apricitabine, entecavir, adefovir or elvucitabine
  • a viral entry inhibitor e.g., enfuvirtide, maraviroc, vicriviroc,
  • the second therapeutic agent is selected from ritonavir, efavirenz, didanosine, tenofovir disoproxil, nelfinavir mesilate, amprenavir, raltegravir, saquinavir, lopinavir, nevirapine, emtricitabine, abacavir, lamivudine, zidovudine, maraviroc, stavudine, darunavir,. fosamprenavir, vicriviroc, pharmaceutically acceptable salts of any of the foregoing, and combinations thereof.
  • the second therapeutic agent is selected from ritonavir, efavirenz, didanosine, raltegravir, tenofovir disoproxil lamivudine, abacavir, zidovudine, emtricitabine, efavirenz, pharmaceutically acceptable salts of any of the foregoing, and combinations thereof.
  • the compositions of this invention comprise a compound of any one of Formulae A, I, Ia, Ib, or Ic, and two to three of the second therapeutic agents set forth above in this paragraph.
  • the compositions of this invention comprise a compound of any one of Formulae A, I, Ia, Ib, or Ic, and two of the second therapeutic agents set forth above in this paragraph.
  • 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.
  • association with one another 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 invention provides a pharmaceutical composition comprising an effective amount of a compound of any one of Formulae A, I, Ia, Ib or Ic, the administration of which to a test subject results in a serum terminal elimination half-life of - o- the compound that is greater than the serum terminal elimination half-life of atazanavir when atazanavir is administered to an equivalent test subject in a pharmaceutical composition comprising a molar equivalent amount of atazanavir and that is administered in the same dosing regimen as the compound of any one of Formulae A, I, Ia, Ib or Ic.
  • the serum terminal elimination half-life of a compound of any one of Formulae A, I, Ia, Ib or Ic is at least 110%, 120%, 130%, 140%, 150% or 160% or more of the serum terminal elimination half-life of atazanavir produced by a molar equivalent atazanavir composition administered in the same dosing regimen.
  • the compound of any one of Formulae A, I, Ia, Ib or Ic is administered in a single dose.
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of a compound of any one of Formulae A, I, Ia, Ib or Ic, or a pharmaceutically acceptable salt thereof, wherein the serum terminal elimination half-life of the compound following administration of a single dose of the composition to a test subject is greater than 5.0 hours, greater than 6.0 hours, greater than 7.0 hours or greater than 8.0 hours.
  • the AUC 0- ⁇ produced by a composition of this invention is at least 120%, 130%, 140%, 150%, 160% or more of the AUC 0- ⁇ produced by a molar equivalent atazanavir composition administered in the same dosing regimen.
  • the compound of any one of Formulae A, I, Ia, Ib or Ic is administered once-daily.
  • the invention provides a pharmaceutical composition comprising an effective amount of a compound of any one of Formulae A, I, Ia, Ib or Ic, the oral administration of which to a test subject results in a maximum serum concentration of the compound (C m3x ) that is greater than the maximum serum concentration of atazanavir when atazanavir is orally administered to an equivalent test subject in a molar equivalent pharmaceutical composition and that is administered in the same dosing regimen as the compound of any one of Formulae A, I, Ia, Ib or Ic.
  • the maximum serum concentration a compound of any one of Formulae A, I, Ia, Ib or Ic produced by oral administration of a composition of this invention is at least 120%, 125%, 130%, 135%, or more than the maximum serum concentration of atazanavir produced by oral administration of a molar equivalent atazanavir composition administered in the same dosing regimen.
  • the compound of any one of Formulae A, I, Ia, Ib or Ic is administered once daily.
  • the invention provides a pharmaceutical composition comprising an effective amount of a compound of any one of Formulae A, I, Ia, Ib or Ic, the oral administration of which to a test subject results in a minimum serum concentration of the compound (C m i n ) that is greater than the minimum serum concentration of atazanavir when atazanavir is orally administered to an equivalent test subject in a molar equivalent pharmaceutical composition and that is administered in the same dosing regimen as the compound of any one of Formulae A, I, Ia, Ib or Ic.
  • the minimum serum concentration a compound of any one of Formulae A, I, Ia, Ib or Ic produced by oral administration of a composition of this invention is at least 125%, 150%, 175%, 200%, or more than the minimum serum concentration of atazanavir produced by oral administration of a molar equivalent atazanavir composition administered in the same dosing regimen.
  • the compound of any one of Formulae A, I, Ia, Ib or Ic is administered once daily.
  • the invention provides a pharmaceutical composition comprising an effective amount of a compound of any one of Formulae A, I, Ia, Ib or Ic, the oral administration of which to a test subject results in a rate of serum clearance of the compound following oral dosing that is less than the rate of serum clearance of atazanavir following oral administration of atazanavir to an equivalent test subject in a molar equivalent pharmaceutical composition and that is administered in the same dosing regimen as the compound of any one of Formulae A, I, Ia, Ib or Ic.
  • the rate of serum clearance of a compound following oral administration of a composition of this invention is less than 90%, less than 80%, less than 70% , or less than 60% of the serum clearance rate of atazanavir following oral administration of a molar equivalent atazanavir composition administered in the same dosing regimen.
  • the compound of any one of Formulae A, I, Ia, Ib or Ic is administered once daily.
  • the invention provides a pharmaceutical composition comprising 150 mg of a compound of any one of Formulae A, I, Ia, Ib or Ic, or a _ _
  • the rate of serum clearance of the compound following oral administration of a single dose of the composition to a chimpanzee is less than 90 ml/h/kg, less than 80 ml/h/kg, less than 75 ml/h/kg, or less than 70 ml/h/kg.
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising 50 mg of a compound of any one of Formulae A, I, Ia, Ib or Ic, or a pharmaceutically acceptable salt thereof, wherein the rate of serum clearance of the compound following oral administration of a single dose of the composition to a chimpanzee is less than 350/h/kg, less than 325/h/kg, less than 300/h/kg, or less than 275/h/kg.
  • the invention provides a pharmaceutical composition comprising an effective amount of a compound of any one of Formulae A, I, Ia, Ib or Ic, the oral administration of which to a test subject results in an amount of compound excreted intact in 24 hours following administration that is greater than the amount of atazanavir excreted intact in 24 hours following oral administration of atazanavir to an equivalent test subject in a molar equivalent pharmaceutical composition and that is administered in the same dosing regimen as the compound of any one of Formulae A, I, Ia, Ib or Ic.
  • the amount of a compound of any one of Formulae A, I, Ia, Ib or Ic excreted intact in 24 hours following oral administration of a composition of this invention is greater than 140%, greater than 160%, greater than 180%, greater than 200%, or greater than 250% or more of the amount of atazanavir excreted intact 24 hours following oral administration of a molar equivalent atazanavir composition administered in the same dosing regimen.
  • the compound of any one of Formulae A, I, Ia, Ib or Ic is administered once daily.
  • the invention provides a pharmaceutical composition comprising an effective amount of a compound of any one of Formulae A, I, Ia, Ib or Ic, the administration of which to a test subject results in either a) a similar AUCo- 12 , b) a similar Cm ax , or c) a similar C m ; n (the lowest concentration within the dosing interval) as atazanavir when atazanavir is administered to an equivalent test subject in a pharmaceutical composition comprising an amount of atazanavir that is greater than the amount of the compound of any one of Formulae A, I, Ia, Ib or Ic on a mole basis of active ingredient and that is administered in the same dosing regimen as the compound of any one of Formulae A, I, Ia, Ib or Ic.
  • the effective amount of a compound of any one of Formulae A, I, Ia, Ib or Ic is no more than 80%, 70%, 60%, 50%, 40%, or less of the amount of atazanavir required to produce a similar AUC 0-I2 , a similar C nUn and/or a similar C m8x when administered in the same dosing regimen as the compound of any one of Formulae A, I, Ia, Ib or Ic.
  • the compound of any one of Formulae A, I, Ia, Ib or Ic is administered once daily.
  • the invention provides a pharmaceutical composition comprising between 250 mg and 275 mg of a compound of any one of Formulae A, I, Ia, Ib or Ic, the administration of which once a day to a test subject in the absence of coadministration of ritonavir results in a C m i n of between 275 and 625 ng/mL of plasma and/or a mean plasma concentration at steady state ("C ss, " also defined as AUCo - ⁇ , where ⁇ is the time of the dosing interval) of between 925 and 1425 ng/mL of plasma.
  • C ss mean plasma concentration at steady state
  • the invention provides a pharmaceutical composition comprising between 275 mg and 300 mg of a compound of any one of Formulae A, I, Ia, Ib or Ic, the administration of which once a day to a test subject results in a C m i n of between 300 and 675 ng/mL of plasma and/or a C ss of between 1000 and 1550 ng/mL of plasma.
  • the invention provides a pharmaceutical composition comprising between 300 mg and 325 mg of a compound of any one of Formulae A, I, Ia, Ib or Ic, the administration of which once a day to a test subject results in a C m i n of between 350 and 750 ng/mL of plasma and/or a C ss of between 1100 and 1675 ng/mL of plasma.
  • the invention provides a pharmaceutical composition comprising between 325 mg and 350 mg of a compound of any one of Formulae A, I, Ia, Ib or Ic, the administration of which once a day to a test subject in the absence of coadministration of ritonavir results in a C nUn of between 375 and 800 ng/mL of plasma and/or a C ss of between 1200 and 1800 ng/mL of plasma.
  • the invention provides a pharmaceutical composition comprising between 350 mg and 375 mg of a compound of any one of Formulae A, I, Ia, Ib or Ic, the administration of which once a day to a test subject in the absence of coadministration of ritonavir results in a C n Un of between 400 and 850 ng/mL of plasma and/or a C ss of between 1300 and 1925 ng/mL of plasma.
  • the invention provides a pharmaceutical composition comprising between 375 mg and 400 mg of a compound of any one of Formulae A, I, Ia, Ib or Ic, the administration of which once a day to a test subject in the absence of coadministration of ritonavir results in a C m i n of between 425 and 900 ng/mL of plasma and/or a C ss of between 1400 and 2050 ng/mL of plasma.
  • the invention provides a pharmaceutical composition comprising between 400 mg and 425 mg of a compound of any one of Formulae A, I, Ia, Ib or Ic, the administration of which once a day to a test subject in the absence of coadministration of ritonavir results in a C m ; n of between 450 and 975 ng/mL of plasma and/or a C 5S of between 1500 and 2175 ng/mL of plasma.
  • the invention provides a pharmaceutical composition comprising between 425 mg and 450 mg of a compound of any one of Formulae A, I, Ia, Ib or Ic, the administration of which once a day to a test subject in the absence of coadministration of ritonavir results in a C 1nJn of between 500 and 1025 ng/mL of plasma and/or a C ss of between 1575 and 2300 ng/mL of plasma.
  • a pharmaceutically acceptable salt of a compound of any one of Formulae A, I, Ia, Ib or Ic, and/or atazanavir may be used instead of the free base form.
  • the compound in each of the compositions set forth above, is a compound of Formula I. In an even more specific embodiment, in each of the compositions set forth above, the compound is a compound of Formula Ib. In a still more specific embodiment, in each of the compositions set forth above, the compound selected from Compound 114, Compound 120, Compound 122, and Compound 131. [153]
  • the term "molar equivalent amount" as used herein means an amount present in a first composition that is the same as the amount present in a second composition on a mole basis of active ingredient.
  • test subject is any mammal, preferably a chimpanzee or a human.
  • An "equivalent test subject” is defined herein as being of the same species and sex as the test subject, and which shows no more than 10% variability as compared to the test subject in the pharmacokinetic parameter being tested after administration of an equal amount of atazanavir to both the test subject and the equivalent subject. The skilled artisan will recognize that one way of reducing variability is to co-dose the compound of the invention along with atazanavir.
  • the compound of the present invention is present in an effective amount.
  • the term "effective amount” refers to an amount which, when administered in a proper dosing regimen, is sufficient to treat (therapeutically or prophylactically) the target disorder.
  • effective amount is sufficient to reduce or ameliorate the severity, duration or progression of the disorder being treated, prevent the advancement of the disorder being treated, cause the regression of the disorder being treated, or enhance or improve the prophylactic or therapeutic effect(s) of another therapy.
  • the compound is present in the composition in an amount of from 0.1 to 50wt.%, more preferably from 1 to 30 wt.%, most preferably from 5 to 20wt.%.
  • an effective amount of a compound of this invention can range from about 200 to about 800 mg per treatment. In more specific embodiments, the range is from about 250 mg to about 600 mg, or from about 250 mg to about 400 mg, or from about 300 mg to about 500 mg, or most specifically from about 325 mg to about 450 mg.
  • Treatment is typically administered from one to two times daily. 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 patient, 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 atazanavir.
  • 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., 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.
  • the invention provides a method of treating HIV infection in a patient in need thereof comprising the step of administering to the patient an effective amount of compound of any one of Formulae A, I, Ia, Ib or Ic or a pharmaceutically acceptable composition comprising a compound of any one of Formulae A, I, Ia, Ib or Ic.
  • Methods delineated herein also include those wherein the patient is identified as in need of a particular stated treatment. Identifying a patient in need of such treatment can be in the judgment of a patient or a health care professional and can be subjective (e.g. opinion) or objective (e.g. measurable by a test or diagnostic method).
  • any of the above methods of treatment comprises the further step of co-administering to said patient one or more second therapeutic agents.
  • the choice of second therapeutic agent may be made from any second therapeutic agent known to be useful for co-administration with atazanavir.
  • the choice of second therapeutic agent is also dependent upon the particular disease or condition to be treated. Examples of second therapeutic agents that may be employed in the methods of this invention are those set forth above for use in combination compositions comprising a compound of this invention and a second therapeutic agent.
  • the combination therapies of this invention include co-administering a compound of any one of Formulae A, I, Ia, Ib or Ic and a second HIV protease inhibitor (e.g., amprenavir, fosamprenavir, tipranavir, indinavir, saquinavir, lopinavir, ritonavir, darunavir, or nelfinavir), a non-nucleoside reverse transcriptase inhibitor ("NNRTI”) (e.g., etravirine, delavirdine, efavirenz, nevirapine, or rilpivirine), a nucleoside/nucleotide reverse transcriptase inhibitor (“NRTI”) (e.g., zidovudine, lamivudine, emtricitabine, zidovudine, tenofovir disoproxil fumarate, didanosine, stavudine, abaca
  • the combination therapies of this invention include co-administering a compound of any one of Formulae A, I, Ia, Ib or Ic and a second therapeutic agent selected from ritonavir, efavirenz, didanosine, tenofovir disoproxil, nelfinavir mesilate, amprenavir, raltegravir, saquinavir, lopinavir, nevirapine, emtricitabine, abacavir, lamivudine, zidovudine, maraviroc, stavudine, darunavir, fosamprenavir, vicriviroc, pharmaceutically acceptable salts of any of the foregoing, and combinations thereof to treat HIV infection in a patient in need thereof.
  • a second therapeutic agent selected from ritonavir, efavirenz, didanosine, tenofovir disoproxil, nelfinavir mesilate, amprenavir, ralte
  • the second therapeutic agent is selected from ritonavir, efavirenz, didanosine, raltegravir, tenofovir disoproxil lamivudine, abacavir, zidovudine, emtricitabine, efavirenz, pharmaceutically acceptable salts of any of the foregoing, and combinations thereof.
  • the method comprises co-administering a compound of any one of Formulae A, I, Ia, Ib, or Ic, and two to three of the second therapeutic agents set forth above in this paragraph.
  • the method comprises co-administering a compound of any one of Formulae A, I, Ia, Ib, or Ic, and two of the second therapeutic agents set forth above in this paragraph.
  • 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.
  • the additional agent may be administered prior to, consecutively with, or following the administration of a compound of this invention.
  • 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, to a patient does not preclude the separate administration of that same therapeutic agent, any other second therapeutic agent or any compound of this invention to said patient at another time during a course of treatment.
  • 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.
  • the recommended dose of Reyataz® (atazanavir sulfate) for the treatment of HIV-I infection is 400 mg once daily with food.
  • the recommended dose is Reyataz 300 mg and ritonavir 100 mg.
  • the recommended dose of Reyataz for the treatment of HIV-I infection is 300 mg with ritonavir 100 mg once daily with food.
  • certain compounds of this invention following a once daily dose in the range of 325 mg to 450 mg, are expected to have the advantage in humans of achieving a Cmin and/or AUC that is comparable to the C m j n and/or AUC achieved with a once-daily dose of 300 mg dose of atazanavir boosted with 100 mg ritonavir.
  • one embodiment of this invention provides a method of treating HIV infection by administering to a subject in need thereof a composition comprising a compound of this invention at a once daily dose in the range of 325 mg to 450 mg. In one embodiment, such a composition is administered without co-administration of ritonavir.
  • Another embodiment relates to a method of treating HIV infection by administering a composition comprising a compound of this invention at a once daily dose in the range of 250 mg to 400 mg.
  • 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 or prevention in a patient of a disease, disorder or symptom set forth above.
  • Another aspect of the invention is a compound of any one of Formulae A, I, Ia, Ib or Ic for use in the treatment or prevention in a patient of a disease, disorder or symptom thereof delineated herein.
  • the compounds of the invention may be used in medicine, such as in therapy. In any of these uses, the compound is preferably administered without co-administration of ritonavir.
  • the invention provides a method of determining the concentration, in a solution or a biological sample, of atazanavir, comprising the steps of: a) adding a known concentration of a compound of Formula A to the solution of biological sample; b) subjecting the solution or biological sample to a measuring device that distinguishes atazanavir from a compound of Formula A; c) calibrating the measuring device to correlate the detected quantity of the compound of Formula A with the known concentration of the compound of Formula A added to the biological sample or solution; and d) measuring the quantity of atazanavir in the biological sample with said calibrated measuring device; and e) determining the concentration of atazanavir in the solution of sample using the correlation between detected quantity and concentration obtained for a compound of Formula A.
  • Measuring devices that can distinguish atazanavir from the corresponding compound of Formula A include any measuring device that can distinguish between two compounds that differ from one another only in isotopic abundance.
  • Exemplary measuring devices include a mass spectrometer, NMR spectrometer, or IR spectrometer.
  • the invention provides a method of evaluating the metabolic stability of a compound of Formula A comprising the steps of contacting the compound of Formula A with a metabolizing enzyme source for a period of time and comparing the amount of the compound of Formula A with the metabolic products of the compound of Formula I after the period of time.
  • the invention provides a method of evaluating the metabolic stability of a compound of Formula A in a patient following administration of the compound of Formula A.
  • This method comprises the steps of obtaining a serum, urine or feces sample from the patient at a period of time following the administration of the compound of Formula A to the subject; and comparing the amount of the compound of Formula A with the metabolic products of the compound of Formula A in the serum, urine or feces sample.
  • the present invention also provides kits for use to treat HIV infection.
  • kits comprise (a) a pharmaceutical composition comprising a compound of any one of Formulae A, I, Ia, Ib or Ic or a salt thereof, wherein said pharmaceutical composition is in a container; and (b) instructions describing a method of using the pharmaceutical composition to treat HIV infection.
  • the container may be any vessel or other sealed or sealable apparatus that can hold said pharmaceutical composition.
  • Examples include bottles, ampules, divided or multi-chambered holders bottles, wherein each division or chamber comprises a single dose of said composition, a divided foil packet wherein each division comprises a single dose of said composition, or a dispenser that dispenses single doses of said composition.
  • the container can be in any conventional shape or form as known in the art which is made of a pharmaceutically acceptable material, for example a paper or cardboard box, a glass or plastic bottle or jar, a re-sealable bag (for example, to hold a "refill" of tablets for placement into a different container), or a blister pack with individual doses for pressing out of the pack according to a therapeutic schedule.
  • the container employed can depend on the exact dosage form involved, for example a conventional cardboard box would not generally be used to hold a liquid suspension. It is feasible that more than one container can be used together in a single package to market a single dosage form. For example, tablets may be contained in a bottle, which is in turn contained within a box. In one embodiment, the container is a blister pack.
  • kits of this invention may also comprise a device to administer or to measure out a unit dose of the pharmaceutical composition.
  • a device to administer or to measure out a unit dose of the pharmaceutical composition may include an inhaler if said composition is an inhalable composition; a syringe and needle if said composition is an injectable composition; a syringe, spoon, pump, or a vessel with or without volume markings if said composition is an oral liquid composition; or any other measuring or delivery device appropriate to the dosage formulation of the composition present in the kit.
  • the kits of this invention may comprise in a separate vessel of container a pharmaceutical composition comprising a second therapeutic agent, such as one of those listed above for use for co-administration with a compound of this invention.
  • a second therapeutic agent such as one of those listed above for use for co-administration with a compound of this invention.
  • Compound 113 was prepared according to Scheme 1, above, following the General Method A described above. Deuterium gas (Cambridge Isotopes, 99.8 atom% D), MeOD (Aldrich, 99.5 atom% D), iPrOD (Aldrich, 98 atom% D) and deuterium chloride (Aldrich, 99 atom% D) were used in this synthesis. Deuterated aldehyde X was prepared according to Scheme 2b using LiAlD 4 (Cambridge Isotopes, 98 atom% D).
  • Example 9 Synthesis of 1 -Methyl- 14-(methyl-d ⁇ f 3S.8S.9S.12S)-3-(l .1 -dimethylethylV 12-IY1.1 -dimethylethyl ' )-dol-8-hvdroxy-4.11 -dioxo-9-fp henylmethylV6-f f4-(2-pyridinyl)phenyl "
  • Example 14 Synthesis of (S)-2-(methoxycarbonylaminoV3,3- ⁇ Q-dimethylbutanoic acid (XVII-rfg).
  • Example 15 Synthesis of (S)-2-(methoxycarbonylaminoV3,3- ⁇ Q-dimethylbutanoic acid (XVII-rfg).
  • microsome-test compound mixtures were added to wells of a 2 mL 96-well deep well polypropylene plate in triplicate. The plate was warmed to 37 0 C before initiating the reactions by addition of prewarmed NADPH in 0.1 M potassium phosphate buffer, pH 7.4, containing 3 mM MgCl 2 .
  • the final reaction mixture composition contained:
  • reaction mixtures were incubated at 37 0 C and 50 ⁇ L aliquots were removed at 0, 3, 7, 12, 20, and 30 minutes and added to shallow-well 96-well plates which contained 50 ⁇ L of ice-cold ACN with internal standard to stop the reactions.
  • the plates were stored at -20 0 C - - for 30 minutes, after which 100 ⁇ L of water was added to the wells of the plate before centrifugation to pellet precipitated proteins.
  • Supernatants were transferred to another 96-well plate and analyzed for amounts of parent remaining by LC-MS/MS using an Applied Biosystems API 4000 mass spectrometer.
  • Male Sprague-Dawley rats (body weight: 170 g to 235 g) were used in this study. Rats were dosed either orally or intravenously with either Compound 122 (2 mg/kg), atazanavir (2 mg/kg) or a 1 :1 combination of Compound 122 and atazanavir (1 mg/kg of each).
  • Plasma samples (300 ⁇ L) were collected via the retro-orbital vein at pre-dose and 0.083, 0.25, 0.5, 1, 2, 4, 6, 8, 10, 12 and 24 hours post-dose. Blood samples were placed into heparinized eppendorf tubes (evaporated) and then centrifuged at 8000 rpm for 6 minutes. 100 ⁇ L aliquots of plasma were transferred to clean Eppendorf tubes and stored with the dose formulation at -20 0 C until bioanalysis. For bioanalysis, plasma was thawed and added to it was 20 ⁇ L methanol and 500 ⁇ L of a 50 ng/ml internal standard solution (quetiapine in methanol). The sample was vortexed, centrifuged at 15,000 rpm for 5 minutes and the - - supernatant transferred to glass autosampler vials.
  • Analyses of plasma samples were performed using a high performance liquid chromatography/mass spectrometry (HPLC/MS/MS) method.
  • the LC system comprised an Agilent (Agilent Technologies Inc. USA) liquid chromatograph equipped with an isocratic pump (1100 series), an autosampler (1100 series) and a degasser (1100 series).
  • Mass spectrometric analysis was performed using an API3000 (triple-quadrupole) instrument from AB Inc (Canada) with an ESI interface.
  • the data acquisition and control system were created using Analyst 1.4 software from ABI Inc. Following intravenous co-administration of Compound 122 and atazanavir, atazanavir disappeared more rapidly from the blood. The accelerated reduction of atazanavir as compared to Compound 122 began between 1 and 2 hours post-IV administration.
  • Compound 122 demonstrated a significant increase in C max as compared to atazanavir following oral co-dosing.
  • the C max , half-life and AUC of the two compounds following oral co-administration is shown in the table below.
  • Compound 122 showed a 43% increase in half-life, a 67% increase in C max , and an 81 % increase in AUC as compared to atazanavir after oral co-dosing of the two compounds in rats.
  • Run B Same as Run A, except the dose was 150 mg orally of each of atazanavir and compounds 114 and 120, and the vehicle was 10 percent ethanol, 40 percent polypropylglycol in 2.5 percent citric acid.
  • the C max , C nUn , half-life, AUC, and clearance (CL, mL/minute/kg) of the compounds following oral co-administration are shown in table 8 and 9 below and Figures 4 and 5.
  • Compounds 114 and 120 had significantly longer half lives Cm a x, C m i ⁇ , and AUC, and had slower clearance rates than atazanavir when co-dosed in chimps.
  • Example 18 HIV Anti- viral Activity.
  • the HIV antiviral activity of compound of the present invention was tested in CEM-SS cells infected with HIV-I.
  • CEM-SS cells were passaged in T-75 flasks in RPMI 1640 medium supplemented with 10% heat inactivated fetal bovine serum, 2 mmol/L L-glutamine, 100 U/mL penicillin and 100 flg/mL streptomycin prior to use in the antiviral assay.
  • the cells were split 1 :2 to assure they were in an exponential growth phase at the time of infection. Total cell and viability quantification was performed using a hemocytometer and Trypan Blue dye exclusion.
  • Cell viability was greater than 95% for the cells to be utilized in the assay.
  • the cells were resuspended at 5 x 10 4 cells per mL in tissue culture medium and added to the drug-containing microtiter plates in a volume of 50 ⁇ L.
  • the virus used for the assay was the lymphocyte-tropic virus strain HIV-I R F.
  • the virus was obtained from the NIH AIDS Research and Reference Reagent Program and stock virus pools were produced in CEM-SS cells. A pre-titered aliquot of virus was removed from the freezer (-80 0 C) and allowed to thaw slowly to room temperature in a biological safety cabinet. Virus was resuspended and diluted into tissue culture medium such that the amount of virus added to each well in a volume of 50 ⁇ L was the amount determined to yield 85 to 95% cell killing at 6 days post-infection.
  • Each plate contains cell control wells (cells only), virus control wells (cells plus virus), compound toxicity wells (cells plus compound only), compound colorimetric control wells (compound only) as well as experimental wells (compound plus cells plus virus).
  • Samples were tested in triplicate with eleven half-log dilutions per compound starting at 0.1 ⁇ M of compound.
  • Compounds 104, 120 and 122 were tested, as was atazanavir and AZT. All compounds were also tested in the presence of 2 mg/mL Ot 1 acid glycoprotein (AAGP), 10 mg/mL human serum albumin (HSA) or a combination of AAGP plus HAS.
  • AAGP Ot 1 acid glycoprotein
  • HSA human serum albumin
  • test plates were stained with the tetrazolium dye XTT
  • XTT-tetrazolium was metabolized by the mitochondrial enzymes of metabolically active cells to a soluble formazan product, allowing rapid quantitative analysis of the inhibition of HIV induced cell killing by anti-HIV test substances.
  • XTT solution was prepared daily as a stock of 1 mg/mL in RPMI 1640.
  • Phenazine methosulfate (PMS) solution was prepared at 0.15 mg/mL in PBS and stored in the dark at -20 °C.
  • XTT/PMS stock was prepared immediately before use by adding 40 ⁇ L of PMS per mL of XTT solution. Fifty microliters of XTT/PMS was added to each well of the plate and the plate was reincubated for 4 hours at 37 °C. Plates were sealed with adhesive plate sealers and shaken gently or inverted several times to mix the soluble formazan product and the plate was read spectrophotometrically at 450/650 nm with a Molecular Devices Vmax plate reader. [255] Raw data was collected from the Softmax Pro 4.6 software and imported into a Microsoft Excel 2003 spreadsheet for analysis by linear curve fit calculations. The results of the assay are shown in the table 10 below.
  • Compounds 122 and 120 yielded EC 50 values of less than 0.4 and 0.5 nM, respectively, in cell culture medium and a 5 to 6-fold increase to 2 and 3 nM, respectively, in the presence of 0.5 mg/mL AAGP.
  • Compound 104 yielded an EC 5O value of less than 0.3 nM in cell culture medium and a greater than 7-fold increase to 2 nM in the presence of AAGP.
  • Compounds 104, 120 and 122 yielded EC 50 values of 0.8, 1 and 0.9 nM, respectively, which was two- to greater than three-fold less potent than in cell culture medium alone.
  • Antiviral activity decreased 8 to 15-fold for compound 122 and 120 in the presence of AAGP plus HSA with EC 5O values of 6 and 4 nM, respectively.
  • Compound 104 yielded an EC 50 value of 4 nM in the presence of AAGP plus HSA, which was greater - - than 13 -fold less potent than in cell culture medium alone.
  • the presence of AAGP, alone or in combination with HSA, resulted in the most significant protein binding and loss of antiviral activity for Compounds 104, 120 and 122.
  • Each of these serum protein affects is simlar to that observed for atazanavir.
  • Each of the compounds of this invention tested in this assay were at least as potent as atazanavir.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Molecular Biology (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Biochemistry (AREA)
  • Virology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • AIDS & HIV (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Pyridine Compounds (AREA)
PCT/US2008/007331 2007-06-12 2008-06-12 Azapeptide derivatives WO2008156632A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
AU2008267048A AU2008267048C1 (en) 2007-06-12 2008-06-12 Azapeptide derivatives
JP2010512186A JP2010529196A (ja) 2007-06-12 2008-06-12 アザペプチド誘導体
MX2009013565A MX2009013565A (es) 2007-06-12 2008-06-12 Derivados de azapeptidos.
CA2692028A CA2692028C (en) 2007-06-12 2008-06-12 Azapeptide derivatives
CN2008800216013A CN101711237B (zh) 2007-06-12 2008-06-12 氮杂肽衍生物
KR1020107000675A KR101185899B1 (ko) 2007-06-12 2008-06-12 아자펩티드 유도체
BRPI0813911-3A BRPI0813911A2 (pt) 2007-06-12 2008-06-12 Composto derivado de azapeptídeos e composição farmacêutica contendo o mesmo
ZA2009/09079A ZA200909079B (en) 2007-06-12 2009-12-18 Azapeptide derivatives

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US93420107P 2007-06-12 2007-06-12
US60/934,201 2007-06-12
US6762708P 2008-02-29 2008-02-29
US61/067,627 2008-02-29

Publications (1)

Publication Number Publication Date
WO2008156632A1 true WO2008156632A1 (en) 2008-12-24

Family

ID=39730784

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/007331 WO2008156632A1 (en) 2007-06-12 2008-06-12 Azapeptide derivatives

Country Status (26)

Country Link
US (4) US20090036357A1 (US20080242721A1-20081002-C00053.png)
EP (3) EP2003120B9 (US20080242721A1-20081002-C00053.png)
JP (1) JP2010529196A (US20080242721A1-20081002-C00053.png)
KR (2) KR101185899B1 (US20080242721A1-20081002-C00053.png)
CN (2) CN102424668A (US20080242721A1-20081002-C00053.png)
AR (1) AR066972A1 (US20080242721A1-20081002-C00053.png)
AT (2) ATE447554T1 (US20080242721A1-20081002-C00053.png)
AU (1) AU2008267048C1 (US20080242721A1-20081002-C00053.png)
BR (2) BRPI0813911A2 (US20080242721A1-20081002-C00053.png)
CA (1) CA2692028C (US20080242721A1-20081002-C00053.png)
CO (1) CO6241121A2 (US20080242721A1-20081002-C00053.png)
CY (1) CY1109766T1 (US20080242721A1-20081002-C00053.png)
DE (1) DE602008000255D1 (US20080242721A1-20081002-C00053.png)
DK (1) DK2003120T3 (US20080242721A1-20081002-C00053.png)
ES (3) ES2356334T3 (US20080242721A1-20081002-C00053.png)
HK (2) HK1136576A1 (US20080242721A1-20081002-C00053.png)
HR (1) HRP20100065T1 (US20080242721A1-20081002-C00053.png)
MX (1) MX2009013565A (US20080242721A1-20081002-C00053.png)
PL (1) PL2003120T3 (US20080242721A1-20081002-C00053.png)
PT (1) PT2003120E (US20080242721A1-20081002-C00053.png)
RS (1) RS51226B (US20080242721A1-20081002-C00053.png)
RU (2) RU2448958C2 (US20080242721A1-20081002-C00053.png)
SI (1) SI2003120T1 (US20080242721A1-20081002-C00053.png)
TW (1) TW200908970A (US20080242721A1-20081002-C00053.png)
WO (1) WO2008156632A1 (US20080242721A1-20081002-C00053.png)
ZA (1) ZA200909079B (US20080242721A1-20081002-C00053.png)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010135424A1 (en) * 2009-05-19 2010-11-25 Glaxosmithkline Llc Chemical compounds
WO2011080562A1 (en) 2009-12-29 2011-07-07 Hetero Research Foundation Novel aza-peptides containing 2,2-disubstituted cyclobutyl and/or substituted alkoxy benzyl derivatives as antivirals
US8158805B2 (en) 2007-06-12 2012-04-17 Concert Pharmaceuticals, Inc. Azapeptide derivatives
US8575174B2 (en) 2011-06-20 2013-11-05 H. Lundbeck A/S Deuterated 1-piperazino-3-phenyl-indanes for treatment of schizophrenia
US10294234B2 (en) 2017-02-06 2019-05-21 Gilead Sciences, Inc. HIV inhibitor compounds
US11052087B2 (en) 2018-07-30 2021-07-06 Gilead Sciences, Inc. Anti-HIV compounds
EP4135740A4 (en) * 2020-04-16 2024-05-22 The Medical College of Wisconsin, Inc. AEROSOL FORMULATIONS OF HIV PROTEASE INHIBITORS FOR THE TREATMENT OF RESPIRATORY REFLUX

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW200835693A (en) * 2007-02-23 2008-09-01 Auspex Pharmaceuticals Inc Preparation and utility of non-nucleoside reverse transcriptase inhibitors
EP2334318B1 (en) * 2008-10-06 2016-02-10 Yissum Research Development Company of The Hebrew University of Jerusalem Ltd. Hiv-1 integrase derived stimulatory peptides interfering with integrase - rev protein binding
WO2010132663A1 (en) * 2009-05-13 2010-11-18 Concert Pharmaceticals, Inc. Pegylated azapeptide derivatives as hiv protease inhibitors
US8784858B2 (en) * 2009-12-21 2014-07-22 Janssen R&D Ireland Degradable removable implant for the sustained release of an active compound
CN106543073A (zh) * 2015-09-17 2017-03-29 宁波杰尔盛化工有限公司 2-[4-(2-吡啶基)苄基]-肼羧酸叔丁酯的制备方法
HRP20211687T1 (hr) * 2015-12-02 2022-03-04 Merck Sharp & Dohme Corp. Farmaceutske kompozicije koje sadrže doravirin, tenofovir dizoproksil fumarat i lamivudin
WO2019025250A1 (en) 2017-08-04 2019-02-07 Basf Se SUBSTITUTED TRIFLUOROMETHYLOXADIAZOLES FOR COMBATING PHYTOPATHOGENIC FUNGI
AR117600A1 (es) * 2018-06-29 2021-08-18 Incyte Corp Formulaciones de un inhibidor de axl / mer

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006014282A2 (en) * 2004-07-06 2006-02-09 Abbott Laboratories Prodrugs of hiv protease inhibitors

Family Cites Families (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0206497B1 (en) 1985-05-15 1994-07-20 The Wellcome Foundation Limited Therapeutic nucleosides and their preparation
GB8815265D0 (en) 1988-06-27 1988-08-03 Wellcome Found Therapeutic nucleosides
US5539122A (en) * 1989-05-23 1996-07-23 Abbott Laboratories Retroviral protease inhibiting compounds
US5304121A (en) 1990-12-28 1994-04-19 Boston Scientific Corporation Drug delivery system making use of a hydrogel polymer coating
GB9009861D0 (en) 1990-05-02 1990-06-27 Glaxo Group Ltd Chemical compounds
KR100333016B1 (ko) 1992-12-29 2002-11-22 아보트 러보러터리즈 레트로바이러스성프로테아제억제화합물,이의제조방법및이를함유하는약제학적조성물
US5716981A (en) 1993-07-19 1998-02-10 Angiogenesis Technologies, Inc. Anti-angiogenic compositions and methods of use
DE69535592T2 (de) 1994-03-25 2008-06-12 Isotechnika, Inc., Edmonton Verbesserung der effektivität von arzneimitteln duren deuterierung
US6221335B1 (en) * 1994-03-25 2001-04-24 Isotechnika, Inc. Method of using deuterated calcium channel blockers
US6099562A (en) 1996-06-13 2000-08-08 Schneider (Usa) Inc. Drug coating with topcoat
US5849911A (en) 1996-04-22 1998-12-15 Novartis Finance Corporation Antivirally active heterocyclic azahexane derivatives
TW409125B (en) * 1996-04-22 2000-10-21 Novartis Ag Antivirally active heterocyclic azahexane derivatives
AU2959397A (en) 1996-05-31 1998-01-05 Novartis Ag Process for the preparation of hydrazine derivatives useful as intermediates for the preparation of peptide analogues
US6087383A (en) * 1998-01-20 2000-07-11 Bristol-Myers Squibb Company Bisulfate salt of HIV protease inhibitor
US6440710B1 (en) * 1998-12-10 2002-08-27 The Scripps Research Institute Antibody-catalyzed deuteration, tritiation, dedeuteration or detritiation of carbonyl compounds
GB9914821D0 (en) 1999-06-24 1999-08-25 Glaxo Group Ltd Compounds
GB9925962D0 (en) 1999-11-02 1999-12-29 Novartis Ag Organic compounds
ES2193921T3 (es) * 1999-12-03 2003-11-16 Pfizer Prod Inc Compuestos de sulfamoilheteroaril-pirazol como agentes antinflamatorios/analgesicos.
ATE282428T1 (de) 2001-05-03 2004-12-15 Hoffmann La Roche Pharmazeutische dosierungsform von amorphem nilfenavir mesylat
CA2446904A1 (en) 2001-05-24 2003-04-03 Alexza Molecular Delivery Corporation Delivery of drug esters through an inhalation route
CA2458807A1 (en) * 2001-08-31 2003-03-13 Bristol-Myers Squibb Company Use of atazanavir in hiv therapy
TW200413273A (en) * 2002-11-15 2004-08-01 Wako Pure Chem Ind Ltd Heavy hydrogenation method of heterocyclic rings
CA2516642C (en) 2003-02-21 2010-11-23 Jarrow Formulas, Inc. Methods for treatment of hiv or malaria using combinations of chloroquine and protease inhibitors
US20050131017A1 (en) 2003-12-11 2005-06-16 Degoey David A. HIV protease inhibiting compounds
US20050148523A1 (en) 2003-12-15 2005-07-07 Colonno Richard J. Method of treating HIV infection in atazanavir-resistant patients using a combination of atazanavir and another protease inhibitor
US7829720B2 (en) * 2004-05-04 2010-11-09 Bristol-Myers Squibb Company Process for preparing atazanavir bisulfate and novel forms
CN101076319A (zh) 2004-09-29 2007-11-21 科迪斯公司 稳定的非晶形雷帕霉素样化合物的药物剂型
CA2588466A1 (en) 2004-12-03 2006-06-08 Merck & Co., Inc. Use of atazanavir for improving the pharmacokinetics of drugs metabolized by ugt1a1
AU2006299424A1 (en) * 2005-10-06 2007-04-12 Auspex Pharmaceuticals, Inc. Deuterated inhibitors of gastric H+, K+-ATPase with enhanced therapeutic properties
US7750168B2 (en) * 2006-02-10 2010-07-06 Sigma-Aldrich Co. Stabilized deuteroborane-tetrahydrofuran complex
JO2630B1 (en) 2006-04-13 2012-06-17 نوفارتيس ايه جي Organic compounds
CA2661404A1 (en) * 2006-09-05 2008-03-13 Schering Corporation Pharmaceutical combinations for lipid management and in the treatment of atherosclerosis and hepatic steatosis
WO2008156632A1 (en) * 2007-06-12 2008-12-24 Concert Pharmaceuticals, Inc. Azapeptide derivatives
US20090076097A1 (en) * 2007-09-14 2009-03-19 Protia, Llc Deuterium-enriched atazanavir
WO2010132663A1 (en) 2009-05-13 2010-11-18 Concert Pharmaceticals, Inc. Pegylated azapeptide derivatives as hiv protease inhibitors

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006014282A2 (en) * 2004-07-06 2006-02-09 Abbott Laboratories Prodrugs of hiv protease inhibitors

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
CIHLAR ET AL: "Suppression of HIV-1 Protease Inhibitor Resistance by Phosphonate-mediated Solvent Anchoring", JOURNAL OF MOLECULAR BIOLOGY, LONDON, GB, vol. 363, no. 3, 27 October 2006 (2006-10-27), pages 635 - 647, XP005683454, ISSN: 0022-2836 *
XU ZHONGMIN ET AL: "Process Research and Development for an Efficient Synthesis of the HIV Protease Inhibitor BMS-232632", ORGANIC PROCESS RESEARCH AND DEVELOPMENT, CAMBRIDGE, GB, vol. 6, no. 3, 1 January 2002 (2002-01-01), pages 323 - 328, XP002429422 *
ZHANG HUIPING ET AL: "A facile and efficient synthesis of d3-labelled Reyataz", JOURNAL OF LABELLED COMPOUNDS AND RADIOPHARMACEUTICALS, JOHN WILEY, CHICHESTER, GB, vol. 48, no. 14, 1 January 2005 (2005-01-01), pages 1041 - 1047, XP002429423, ISSN: 0362-4803 *

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8158805B2 (en) 2007-06-12 2012-04-17 Concert Pharmaceuticals, Inc. Azapeptide derivatives
US8258309B2 (en) 2007-06-12 2012-09-04 Concert Pharmaceuticals, Inc. Azapeptide derivatives
WO2010135424A1 (en) * 2009-05-19 2010-11-25 Glaxosmithkline Llc Chemical compounds
WO2011080562A1 (en) 2009-12-29 2011-07-07 Hetero Research Foundation Novel aza-peptides containing 2,2-disubstituted cyclobutyl and/or substituted alkoxy benzyl derivatives as antivirals
US10118907B2 (en) 2011-06-20 2018-11-06 H. Lundbeck A/S Deuterated 1-piperazino-3-phenyl indanes for treatment of schizophrenia
US9012453B2 (en) 2011-06-20 2015-04-21 H. Lundbeck A/S Deuterated 1-piperazino-3-phenyl indanes for treatment of schizophrenia
US9216961B2 (en) 2011-06-20 2015-12-22 H. Lundbeck A/S Deuterated 1-piperazino-3-phenyl indanes for treatment of schizophrenia
US9617231B2 (en) 2011-06-20 2017-04-11 H. Lundbeck A/S Deuterated 1-piperazino-3-phenyl indanes for treatment of schizophrenia
US8575174B2 (en) 2011-06-20 2013-11-05 H. Lundbeck A/S Deuterated 1-piperazino-3-phenyl-indanes for treatment of schizophrenia
US10501427B2 (en) 2011-06-20 2019-12-10 H. Lundbeck A/S Deuterated 1-piperazino-3-phenyl indanes for treatment of schizophrenia
US11059798B2 (en) 2011-06-20 2021-07-13 H. Lundbeck A/S Deuterated 1-piperazino-3-phenyl indanes for treatment of schizophrenia
US10294234B2 (en) 2017-02-06 2019-05-21 Gilead Sciences, Inc. HIV inhibitor compounds
US10752636B2 (en) 2017-02-06 2020-08-25 Gilead Sciences, Inc. HIV inhibitor compounds
US11078208B1 (en) 2017-02-06 2021-08-03 Gilead Sciences, Inc. HIV inhibitor compounds
US11052087B2 (en) 2018-07-30 2021-07-06 Gilead Sciences, Inc. Anti-HIV compounds
EP4135740A4 (en) * 2020-04-16 2024-05-22 The Medical College of Wisconsin, Inc. AEROSOL FORMULATIONS OF HIV PROTEASE INHIBITORS FOR THE TREATMENT OF RESPIRATORY REFLUX

Also Published As

Publication number Publication date
US8158805B2 (en) 2012-04-17
CN102424668A (zh) 2012-04-25
SI2003120T1 (sl) 2010-03-31
RS51226B (sr) 2010-12-31
RU2012101881A (ru) 2013-07-27
KR101185899B1 (ko) 2012-09-27
PL2003120T3 (pl) 2010-04-30
EP2116532A1 (en) 2009-11-11
RU2448958C2 (ru) 2012-04-27
HK1136576A1 (en) 2010-07-02
TW200908970A (en) 2009-03-01
ATE536343T1 (de) 2011-12-15
ES2356334T3 (es) 2011-04-07
AU2008267048C1 (en) 2013-01-17
CA2692028C (en) 2013-06-04
BRPI0813911A2 (pt) 2012-02-22
EP2003120B9 (en) 2010-06-02
EP2322509B1 (en) 2012-08-22
BRPI0823520A2 (pt) 2013-12-17
ZA200909079B (en) 2011-05-25
AU2008267048A1 (en) 2008-12-24
CA2692028A1 (en) 2008-12-24
PT2003120E (pt) 2010-02-11
ES2394952T3 (es) 2013-02-07
HK1127345A1 (en) 2009-09-25
DK2003120T3 (da) 2010-03-15
US20090036357A1 (en) 2009-02-05
KR20120029480A (ko) 2012-03-26
EP2003120B1 (en) 2009-11-04
ATE447554T1 (de) 2009-11-15
JP2010529196A (ja) 2010-08-26
MX2009013565A (es) 2010-06-02
HRP20100065T1 (hr) 2010-03-31
RU2010100821A (ru) 2011-07-20
US20120165288A1 (en) 2012-06-28
US20130041156A1 (en) 2013-02-14
AU2008267048B2 (en) 2012-05-31
CY1109766T1 (el) 2014-09-10
US20110009355A1 (en) 2011-01-13
KR20100020033A (ko) 2010-02-19
DE602008000255D1 (de) 2009-12-17
CN101711237B (zh) 2013-08-07
CN101711237A (zh) 2010-05-19
AR066972A1 (es) 2009-09-23
CO6241121A2 (es) 2011-01-20
ES2395137T3 (es) 2013-02-08
EP2322509A1 (en) 2011-05-18
US8258309B2 (en) 2012-09-04
EP2116532B1 (en) 2011-12-07
EP2003120A1 (en) 2008-12-17

Similar Documents

Publication Publication Date Title
CA2692028C (en) Azapeptide derivatives
EP2891658B1 (en) Tenofovir prodrug and pharmaceutical uses thereof
JPH11269146A (ja) 分化誘導剤
JPH01168689A (ja) K−252誘導体
HUT71891A (en) 3-phenylpyrrolidine derivatives and pharmaceutical compositions containing them
JP2021531234A (ja) ヒトインテグリンα4β7のアンタゴニスト
JP2021532107A (ja) ジカチオン化合物及びその製造方法と使用
WO2009148600A2 (en) Deuterated lysine-based compounds
US20110257111A1 (en) Hydroxyethlamino Sulfonamide Derivatives
US8592487B2 (en) Deuterated darunavir
AU2012216511A1 (en) Azapeptide derivatives
JP2023518851A (ja) Hiv治療に有用な化合物
EP2163553B1 (en) Arene connected polyamine macroring derivatives, preparation methods and pharmaceutical uses thereof
WO2010132663A1 (en) Pegylated azapeptide derivatives as hiv protease inhibitors
WO2021257669A1 (en) Synthesis of substituted arylmethylureas, analogues, and crystalline forms thereof and methods of using same

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200880021601.3

Country of ref document: CN

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

Ref document number: 08768383

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2010512186

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2692028

Country of ref document: CA

Ref document number: 09141805

Country of ref document: CO

Ref document number: MX/A/2009/013565

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 8182/DELNP/2009

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2008267048

Country of ref document: AU

ENP Entry into the national phase

Ref document number: 20107000675

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2010100821

Country of ref document: RU

ENP Entry into the national phase

Ref document number: 2008267048

Country of ref document: AU

Date of ref document: 20080612

Kind code of ref document: A

122 Ep: pct application non-entry in european phase

Ref document number: 08768383

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: PI0813911

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20091214