WO1992015319A1 - Hiv protease inhibitors - Google Patents

Hiv protease inhibitors Download PDF

Info

Publication number
WO1992015319A1
WO1992015319A1 PCT/US1992/001738 US9201738W WO9215319A1 WO 1992015319 A1 WO1992015319 A1 WO 1992015319A1 US 9201738 W US9201738 W US 9201738W WO 9215319 A1 WO9215319 A1 WO 9215319A1
Authority
WO
WIPO (PCT)
Prior art keywords
benzyl
phenyl
dihydroxy
oxo
hexyl
Prior art date
Application number
PCT/US1992/001738
Other languages
French (fr)
Inventor
John Gerald Gleason
Robert Thomas Lum
Original Assignee
Smithkline Beecham Corporation
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 Smithkline Beecham Corporation filed Critical Smithkline Beecham Corporation
Priority to JP4508263A priority Critical patent/JPH06505495A/en
Publication of WO1992015319A1 publication Critical patent/WO1992015319A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C271/00Derivatives of carbamic acids, i.e. compounds containing any of the groups, the nitrogen atom not being part of nitro or nitroso groups
    • C07C271/06Esters of carbamic acids
    • C07C271/08Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms
    • C07C271/10Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms with the nitrogen atoms of the carbamate groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C271/22Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms with the nitrogen atoms of the carbamate groups bound to hydrogen atoms or to acyclic carbon atoms to carbon atoms of hydrocarbon radicals substituted by carboxyl groups
    • 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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C237/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups
    • C07C237/02Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton
    • C07C237/22Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton having nitrogen atoms of amino groups bound to the carbon skeleton of the acid part, further acylated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/02Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing at least one abnormal peptide link
    • C07K5/0207Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing at least one abnormal peptide link containing the structure -NH-(X)4-C(=0), e.g. 'isosters', replacing two amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • Retroviruses that is, viruses within the family of Retroviridae, are a class of viruses which transport their genetic material as ribonucleic acid rather than
  • RNA-tumor viruses also known as RNA-tumor viruses, their presence has been associated with a wide range of diseases in humans and animals. They are believed to be the causative agents in pathological states associated with infection by Rous sarcoma virus (RSV), murine leukemia virus (MLV), mouse mammary tumor virus (MMTV), feline leukemia virus (FeLV), bovine leukemia virus (BLV), Mason-Pfizer monkey virus (MPMV) , simian sarcoma virus (SSV), simian acquired immunodeficiency syndrome (SAIDS), human T-lymphotropic virus (HTLV-I, -II) and human immunodeficiency virus (HIV-1, HIV-2), which is the etiologic agent of AIDS (acquired immunodeficiency syndrome) and AIDS related
  • the extracellular virus particle is composed of an outer membrane studded with viral glycoproteins, a core of structural proteins, and a genome of single stranded ribonucleic acid.
  • the retroviral genome has a distinctive regional organization, referred to as the 5'- gag-pol-env-3' structure, wherein the gag region encodes the core structural proteins, the pol region encodes certain critical viral enzymes such as reverse transcriptase, integrase and protease, and the env region encodes the envelope glycoproteins.
  • Viral replication occurs only within host cells and is dependent upon host cellular functions.
  • the method of isosteric replacement has been disclosedas a strategy for the development of protease inhibitors for HIV-1.
  • Published European Patent applications EP-A 337 714, EP-A 352 000 and EP-A 357 332, EP-A 346 847, EP-A 342 541 and EP-A 393 445 are representative. Similar strategies have also been reported for inhibition of renin in U.S. Patents 4,713,445 and 4,661,473.
  • protease- inhibiting compounds which have a favorable balance of potency and pharmacokinetics properties.
  • the compounds of this invention have a unique 2,4 dihydroxy isostere wherein the hydroxyl group in the 2 position is further substituted to create a tertiary alcohol.
  • the compounds are potent inhibitors of retroviral protease and are able to penetrate affected cells.
  • This invention comprises compounds, hereinafter
  • This invention is also a pharmaceutical composition, which comprises a compound of formula (I) and a
  • This invention further constitutes a method for treating retroviral disease, which comprises administering to a mammal in need thereof an effective amount of a compound of formula
  • R 1 is A, A-B or A-B-D
  • A is R 11 , R 11 CO, R 11 - (CHR 11 ' ) n -CO, R 1 1 OCO, R 1 1 OCH (R 11 ' ) CO, R 11 NHCH ( R 11 ' ) CO, R 11 SCH ( R 11 ' ) CO, R 1 1 SO 2 , R 1 1 SO or R 1 1 C (O) CO;
  • R 2 and R 4 are C 1-6 alkyl, optionally substituted with 1-5 fluorine atoms, C 3-6 alkenyl, C 1-6 alkyl-O-CH 2 or (CH 2 ) n -T, wherein T is phenyl, naphthyl, C 5-6 cycloalkyl or indolyl, optionally substituted with nitro, halogen, C 1-4 alkyl,
  • R 5 is H, C 1-6 alkyl, C 1-6 alkyl-O-CH 2 or J-CH 2 (CH 2 )n, wherein J is CONHR', CO 2 R', NHR', SR', or phenyl, naphthyl, C 5-6 cycloalkyl or indolyl, optionally substituted with nitro, halogen, hydroxy, C 1-4 alkyl, C 1-4 alkoxy, C 1-4 alkylthio or trifluoromethyl;
  • R 3 is G, X-G or X-Y-G; X and Y are
  • G is R 10 , NR'R 10 , NR'NR'-A, OR 10 or SR 10 ;
  • R 10 is (CR 11 R 11 ') n -W;
  • R 11 and R 11 ' are H, Alk, Ar, Het, Ar-C 1-5 alkyl,
  • W is H, CH 2 OR", COR", OR 11 , OCOR 11 , NR'R 11 , NR'COR 11 , SR 11 , Ar or Het;
  • Alk is C 1-6 alkyl or C ⁇ -6alkyl substituted by one or two hydroxy, nitro or C 1-4 alkoxy groups, or one to five fluoro atoms;
  • R' is H, C 1-6 alkyl or (CH 2 ) n -Ar;
  • R" is H, C 1-6 alkyl or (CH 2 ) n -Ph;
  • R 1 is A or A-B.
  • R 2 is C 1-6 alkyl or (CH 2 ) n -phenyl.
  • R 3 is G.
  • R 3 is NHCH(R 11 )-W, wherein R 11 is H or Alk and W is CH 2 OH, CONH 2 or phenyl.
  • R 1 is A. More suitably R 1 is C 1-6 alkoxycarbonyl. Preferably R 2 is benzyl.
  • R 3 is NHCH(i-pr)CH 2 OH.
  • R 4 is benzyl
  • Prodrugs are considered to be any covalently bonded carriers which release the active parent drug according to formula (I) in vivo.
  • Ar indicates aryl, Ar is phenyl or naphthyl, or phenyl or naphthyl substituted by one to three C 1-4 alkyl, C 1-4 alkoxy, C 1-4 alkylthio, trifluoromethyl, nitro, mercapto, hydroxy, halogen, cyano, CO 2 R" or CON(R") 2 .
  • Het indicates an
  • heterocyclic ring is a saturated or unsaturated, five or six membered ring or nine or ten-membered bicyclic ring, containing one to three heteroatoms chosen from the group of nitrogen, oxygen and sulfur, which are stable and available by conventional chemical synthesis.
  • Illustrative heterocycles are pyridyl, furyl, thienyl, morpholinyl, pyrrolinyl, pyrrolidinyl, pyrazolyl,
  • Boc refers to the t-butyloxycarbonyl radical
  • Z and Cbz refer to the carbobenzyloxy radical
  • BrZ refers to the o-bromobenzyloxy-carbonyl radical
  • C1Z is the
  • C12Z refers to the 2,4-dichlorocarbobenzyloxy radical
  • Bn refers to the benzyl radical
  • Ac refers to acetyl
  • Ph refers to phenyl
  • BrMgDA refers to bromomagnesium diisopropylamide
  • DCC refers to dicyclohexyl-carbodiimide
  • DMAP refers to dimethylaminopyridine
  • HOBT 1-hydroxybenzotriazole
  • OSU is
  • 1-hydroxysuccinimide NMM is N-methylmorpholine
  • DTT is dithiothreitol
  • EDTA is ethylenediamine tetraacetic acid
  • DIEA is diisopropyl ethylamine
  • DBU is 1,8 diazobicyclo [5.4.0]undec-7-ene
  • DMSO is dimethylsulfoxide
  • DMF is dimethyl formamide
  • HMPA hexamethylphosphoramide
  • DMAPEC is 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide
  • DCC dicyclohexylcarbodiimide
  • EDC N-ethyl-N'(dimethylaminopropyl)-carbodiimide
  • PPA 1-propanephosphonic acid cyclic anhydride
  • DPPA diphenylphosphoryl azide
  • BOP refers to
  • TMS-Imid N-trimethysilylimidazole
  • valinol (2S)-2-amino-3-methyl-butanol
  • THF is
  • HF refers to hydrofluoric acid
  • TFA refers to trifluoroacetic acid.
  • C 1-6 alkyl as applied herein is meant to include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tertiary butyl, n-pentyl, isopentyl arid hexyl. As used herein in the compounds of this invention.
  • Representative compounds of this invention are:
  • Preferred compounds of this invention are:
  • the chiral centers of the compounds of the invention may be racemic or nonracemic. Racemic mixtures, mixtures of diastereomers, as well as single diastereomers and
  • Pr 1 is an amino protecting group
  • Pr 2 is a carboxy protecting group
  • Pr 3 and Pr 5 are H or a hydroxy
  • Pr2 is generally OH, O-alkyl, O-benzyl or O-phenyl.
  • Methods of general synthetic utility are found in U.S. Patents 4,661,473 and 4,713,445, published European Patent applications EP-A 352 000 and 337 714,
  • Pr 3 is H or a hydroxy-protecting group
  • Pr 5 is H or a hydroxy-protecting group
  • A, B, D, R 4 and R 2 are as defined for formula (I), with any reactive groups protected
  • Pr 3 is H or a hydroxy-protecting group
  • X, Y, G, R 2 and R 4 are as defined in formula (I), with any reactive groups protected
  • G, X, Y, A and R 10 are as defined for formula (I), with any reactive groups protected, and
  • Typical coupling agents include carbodiimides, such as DCC, EDC, DMAPEC, DPPA, PPA, and the BOP reagent, and
  • coupling reagents would additionally include organometallic reagents, such as lithium dimethyl copper which could be reacted with an acyl halide to form a ketone.
  • Typical bases include organic nitrogenous bases, such as triethylamine or diisopropylamine, or an alkali metal
  • Typical reactive groups which may require protection during synthesis are the hydroxyl, amino, carboxyl, carbonyl and mercapto groups. Conventional methods for protection and deprotection of reactive groups are disclosed in Greene,
  • R 4 may be introduced into the isosteres (II) or (III) as any desired amino acid side chain, with any reactive groups suitably protected.
  • R 2 may be introduced into the isostere and varied as
  • An ⁇ -substituted, ⁇ -amino-aldehyde, such as (IV), is prepared by conventional means.
  • a suitable ⁇ - amino acid may be reduced to its corresponding carboxaldehyde to initiate the preparation.
  • Suitable methods are disclosed Coppola et al . , Asymmetric Synthesis, Construction of Chiral Molecules Using Amino Acids, Wiley Interscience, New York, 1987.
  • the aldehyde (IV) is condensed with the enolate derived from methyl pyruvate (V) and a suitable base, such as
  • the enol lactone 1 is reduced with an appropriate reducing agent, such as hydrogen with a palladium catalyst to yield an ⁇ -hydroxy lactone as mixture of epimers at the hydroxyl position.
  • an appropriate reducing agent such as hydrogen with a palladium catalyst to yield an ⁇ -hydroxy lactone as mixture of epimers at the hydroxyl position.
  • the hydroxyl group is then protected with an appropriate protective group which is stable to strong base, such as a silyl ether.
  • Subsequent treatment of the lactone with a strong base such as lithium alkyl or
  • hydroxyl group at the 4 position can be protected, or the hydroxyl group at the 4 position can be selectively protected.
  • a silyl ether is the most useful group for this purpose.
  • the protected compound allows further modification of the carboxy terminus via amidation, esterification or coupling of an activated acyl intermediate, such as an acyl halide, with an organometallic reagent.
  • deprotecting the amino terminus allows further modification of the amine group by alkylation, acylation or sulfonylation.
  • Solution synthesis of the peptide bonds is accomplished using conventional methods for coupling the appropriate amino acid residues and optionally removing any protective groups. Suitable methods are disclosed, for instance, in Bodansky et al . , The Practice of Peptide Synthesis, Springer-Verlag, Berlin, 1984. Typically, a protected Boc-amino acid which has a free carboxyl group is coupled to a protected amino acid which has a free amino group using a suitable
  • carbodiimide coupling agent such as N, N' dicyclohexyl carbodiimide (DCC) or 1-(3-dimethylaminopropyl)-3- ethylcarbodiimide hydrochloride (DMAPEC), optionally in the presence of catalysts such as 1-hydroxybenzotriazole (HOBT) and dimethylamino pyridine (DMAP).
  • DCC N, N' dicyclohexyl carbodiimide
  • DMAPEC 1-(3-dimethylaminopropyl)-3- ethylcarbodiimide hydrochloride
  • catalysts such as 1-hydroxybenzotriazole (HOBT) and dimethylamino pyridine (DMAP).
  • HOBT 1-hydroxybenzotriazole
  • DMAP dimethylamino pyridine
  • a protected Boc-amino acid or peptide is treated in an anhydrous solvent, such as methylene chloride or tetrahydrofuran (THF), in the presence of a base, such as N-methyl morpholine, DMAP or a trialkyl amine, with isobutyl chloroformate to form the "activated anhydride", which is subsequently reacted with the free amine of a second protected amino acid or peptide.
  • anhydrous solvent such as methylene chloride or tetrahydrofuran (THF)
  • a base such as N-methyl morpholine, DMAP or a trialkyl amine
  • Esters are often used to protect the terminal carboxyl group of peptides in solution synthesis. They may be
  • the acids may be converted to other esters via an activated acyl intermediate as previously described.
  • the amides and substituted amides of this invention are prepared from carboxylic acids of the peptides in much the same manner.
  • ammonia or a substituted amine may be reacted with an activated acyl intermediate to produce the amide.
  • Use of coupling reagents, such as DCC or DMAPEC, is convenient for forming substituted amides from the carboxylic acid itself and a suitable amine.
  • methyl esters of this invention may be converted to the amides, or substituted-amides, directly by treatment with ammonia, or a substituted amine, in methanol solution.
  • a methanol solution of the methyl ester of the peptide is saturated with ammonia and stirred in a
  • an acid addition salt may be
  • Acid addition salts of the peptides are prepared in a standard manner in a suitable solvent from the parent compound and an excess of an acid, such as hydrochloric, hydrobromic, sulfuric, phosphoric, acetic, maleic, succinic or methanesulfonic .
  • the acetate salt form is especially useful.
  • cationic salts may be prepared.
  • the parent compound is treated with an excess of an alkaline reagent, such as a hydroxide, carbonate or alkoxide, containing the appropriate cation.
  • Cations such as Na + , K + , Ca ++ and NH 4 + are examples of cations present in pharmaceutically
  • Certain of the compounds form inner salts or zwitterions which may also be acceptable.
  • the compounds of formula (I) are used to induce antiviral activity in patients which are infected with
  • the this invention is a method of treating disease resulting from infection by retroviruses by
  • this invention is a method of treating Acquired Immune Deficiency Syndrome (AIDS).
  • the method of treatment comprises the administration orally, parenterally, buccally, trans-dermally, rectally or by insufflation, of an effective quantity of the chosen
  • Dosage units of the active ingredient are generally selected from the range of 0.01 to 25 mg/kg, but will be readily determined by one skilled in the art depending upon the route of administration, age and condition of the patient. These dosage units may be administered one to ten times daily for acute or chronic infection.
  • protease inhibiting properties of the peptides of this invention are demonstrated by their ability to inhibit the hydrolysis of a peptide substrate by rHIV protease in the range of about 20 nM to about 60 ⁇ M.
  • the following table is representative of the inhibition constants of the compounds of this invention.
  • compositions of the peptides of this invention, or derivatives thereof, may be formulated as solutions or lyophilized powders for parenteral
  • Powders may be reconstituted by addition of a suitable diluent or other pharmaceutically acceptable carrier prior to use.
  • the liquid formulation is generally a buffered, isotonic, aqueous solution.
  • suitable diluents are normal isotonic saline solution, standard 5% dextrose in water or buffered sodium or ammonium acetate solution.
  • Such formulation is especially suitable for parenteral administration, but may also be used for oral administration or contained in a metered dose inhaler or nebulizer for insufflation. It may be desirable to add excipients such as polyvinylpyrrolidone, gelatin, hydroxy cellulose, acacia, polyethylene glycol, mannitol, sodium chloride or sodium citrate.
  • these peptides may be encapsulated, tableted or prepared in a emulsion or syrup for oral
  • compositions may be added to enhance or stabilize the
  • Liquid carriers include syrup, peanut oil, olive oil, glycerin, saline and water.
  • Solid carriers include starch, lactose, calcium sulfate dihydrate, terra alba, magnesium stearate or stearic acid, talc, pectin, acacia, agar or gelatin.
  • the carrier may also include a sustained release material such as glyceryl monostearate or glyceryl
  • the amount of solid carrier varies but, preferably, will be between about 20 mg to about 1 g per dosage unit.
  • the pharmaceutical preparations are made following the conventional techniques of pharmacy involving milling, mixing, granulating, and compressing, when necessary, for tablet forms; or milling, mixing and filling for hard gelatin capsule forms .
  • a liquid carrier When a liquid carrier is used, the preparation will be in the form of a syrup, elixir, emulsion or an aqueous or non-aqueous suspension. Such a liquid formulation may be administered directly p.o. or filled into a soft gelatin capsule.
  • a pulverized powder of the peptides of this invention may be combined with excipients such as cocoa butter, glycerin, gelatin or polyethylene glycols and molded into a suppository.
  • excipients such as cocoa butter, glycerin, gelatin or polyethylene glycols
  • the pulverized powders may also be compounded with an oily preparation, gel, cream or emulsion, buffered or unbuffered, and administered through a transdermal patch.
  • Beneficial effects may be realized by co-administering, individually or in combination, other anti-viral agents with the protease inhibiting compounds of this invention.
  • anti-viral agents examples include nucleoside analogues, phosphonoformate, rifabutin, ribaviran, phosphonothioate oligodeoxynucleotides, castanospermine, dextran sulfate, alpha interferon and ampligen.
  • Nucleoside analogues which include 2',3'-dideoxycytidine(ddC), 2',3'-dideoxyadenine(ddA) and 3'-azido-2',3'-dideoxythymide(AZT), are especially useful.
  • AZT is one preferred agent.
  • pharmaceutical compositions comprise an anti-viral agent, a protease
  • the enzyme used to assay the peptide of this invention was produced in this manner and purified from the cell pellet as follows.
  • the E. coli cell pellet was resuspended in a buffer consisting of 50 mM Tris- HCl, pH 7.5; 1.0 mM each DTT, EDTA and PMSF
  • the column was equilibrated in the same buffer at a flow rate of 4 ml/min.
  • the effluent of the column was monitored at 280 nm and 1 min. fractions collected.
  • the rHIVPRT recombinant HIV protease
  • the protease was 85-95% pure.
  • By immunoblot analysis >90% of the immunoreactive material was precipitated at the ammonium sulfate step.
  • activity assay the highest peak of activity was found in the fractions collected at 45 and 46 minutes.
  • the overall yield of rHIVPRT was N1 mg from a 50 gm E. coli cell pellet.
  • MENDT buffer 50 mM Mes (pH 6.0; 2-(N-morpholino)ethanesulfonic acid), 1 mM
  • reaction was initiated with purified 0.01-1 mg HIV protease.
  • Reaction mixtures 37°C
  • reaction mixtures 37°C
  • Ultrasphere ® and Ultrasphere ® ODS are silica gel and octadecylsilane chromatographic supports
  • Microsorb ® is a silica gel chromatographic support manufactured by Rainin Instruments Co., Woburn, Mass. Bakerbond ® is a silica gel chromatographic support
  • Celite ® is filter aid composed of acid washed diatomaceous silica manufactured by Mansville Corp., Denver, Colorado.
  • steps 1(b) and 1(c) except substituting the enantiomerically pure (5S)-5-((1S)-1-t-butyloxycarbonylamino-2-phenyl)ethyl-4H,5H-dihydrofuran-2,3-dione 1a in step 1(b), (5S)-5-((1S)-1-t-butyloxycarbonylamino-2-phenyl)ethyl-3-trimethysilyloxy-tetrahydrofuran-2-one (3a) was prepared.
  • reaction mixture was poured into 0.05 M HCl, and separated.
  • the aqueous layer was extracted ethyl acetate (3X), the combined organic layers were washed with 0.05 M HCl, brine, dried over MgSO 4 , and concentrated to a yellow oil.
  • steps 1 (b) through 1(d) except using the enantiomerically pure (5S)-5-((1S)-1-t-butyloxycarbonylamino- 2-phenyl)ethyl-4H,5H-dihydrofuran-2,3-dione, 1a, in step 1(b), (3S,5S)-5-((1S)-1-t-butyloxycarbonylamino-2-phenyl)ethyl-3-trimethysilyloxy-3-benzyl-tetrahydrofuran-2- one (4a) was prepared.
  • n-butyllithium (1.6M solution in hexane, 1.25 mL, 1.98 mmol). The reaction mixture was allowed to stir for 20 min. To this mixture was added a solution of 4 (240 mg, 0.495 mmol) in THF (10 mL) via cannula. The reaction mixture was allowed to stir for 1 h, poured into 0.05 M HCl, and extracted with ethyl acetate (3X). The organic layers were washed with 0.05 M HCl, brine, dried over MgSO 4 , and concentrated to a white solid. The solid was recrystallized from ethyl acetate, to afford the product (220 mg,86%):
  • reaction mixture was cooled to 0°C and Cbz-D-Ala (34 mg, 0.15 mmol), 1-hydroxybenzotriazole (HOBT) (21 mg, 0.15 mmol), and 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide
  • reaction mixture was cooled to 0°C and Cbz-L-Ala (40 mg, 0.18 mmol), HOBT (24 mg, 0.18 mmol), and DMAPEC (35 mg, 0.18 mmol) were added.
  • the reaction mixture was allowed to stir at 0°C for 1 h, and the ice bath was removed.
  • the mixture was allowed to warm and stir to room temperature for 16 h.
  • the mixture was diluted with methylene chloride, washed with 10% NaHCO 3 , 0.05 M HCl, brine, dried over MgSO 4 , and
  • reaction mixture was poured into 0.05 M HCl, and separated.
  • the aqueous layer was extracted ethyl acetate (3X), the combined organic layers were washed with 0.05 M HCl, brine, dried over MgS ⁇ 4 , and concentrated to a yellow oil. This was
  • Phosphatidylcholine 1.4 g
  • phosphatidylglycerol 0.6g
  • polycarbonate filter five times to produce a uniform size distribution. If necessary the suspension can be dialysed or ultracentrifuged to remove non-encapsulated peptide.
  • a preparation which contains 250 mg of the compound of Example 5 is prepared as follows:
  • Example 15 distilled water. The solution is filtered under sterile conditions in to a multi-dose ampoule and lyophilized. The powder is reconstituted by addition of 200 mL of 5% dextrose in water (D5W) for intravenous injection. The dosage is thereby determined by the injection volume. This solution is also suitable for use in other methods for administration, such as in a bottle or bag for IV drip infusion.
  • Example 15
  • a capsule for oral administration is prepared by mixing and milling 350 mg of the compound of Example 5 with 750 mg of lactose and 50 mg of magnesium stearate. The resulting powder is screened and filled into a hard gelatin capsule.

Abstract

The invention relates to HIV protease inhibitors wherein the compounds have a 2,4-dihydroxy isostere wherein the hydroxyl group in the 2-position is further substituted to create a tertiary alcohol. Also disclosed are pharmaceutically acceptable salts thereof, which inhibit HIV-I protease and are useful in the treatment of AIDS.

Description

TITLE
HIV PROTEASE INHIBITORS
BACKGRO UND
Retroviruses, that is, viruses within the family of Retroviridae, are a class of viruses which transport their genetic material as ribonucleic acid rather than
deoxyribonucleic acid. Also known as RNA-tumor viruses, their presence has been associated with a wide range of diseases in humans and animals. They are believed to be the causative agents in pathological states associated with infection by Rous sarcoma virus (RSV), murine leukemia virus (MLV), mouse mammary tumor virus (MMTV), feline leukemia virus (FeLV), bovine leukemia virus (BLV), Mason-Pfizer monkey virus (MPMV) , simian sarcoma virus (SSV), simian acquired immunodeficiency syndrome (SAIDS), human T-lymphotropic virus (HTLV-I, -II) and human immunodeficiency virus (HIV-1, HIV-2), which is the etiologic agent of AIDS (acquired immunodeficiency syndrome) and AIDS related
complexes, and many others. Although the pathogens have, in many of these cases, been isolated, no effective method for treating this type of infection has been developed. Among these viruses, the HTLV and HIV have been especially well characterized.
Although diverse in detail, all retroviruses are rather similar in overall structure. The extracellular virus particle is composed of an outer membrane studded with viral glycoproteins, a core of structural proteins, and a genome of single stranded ribonucleic acid. The retroviral genome has a distinctive regional organization, referred to as the 5'- gag-pol-env-3' structure, wherein the gag region encodes the core structural proteins, the pol region encodes certain critical viral enzymes such as reverse transcriptase, integrase and protease, and the env region encodes the envelope glycoproteins. Viral replication occurs only within host cells and is dependent upon host cellular functions.
Critical to this replication is the production of functional viral proteins. Protein synthesis is accomplished by
translation of the open reading frames into polyprotein constructs, corresponding to the gag, pol and env reading frames, which are processed, at least in part, by a viral protease into the functional proteins. The proteolytic activity provided by the viral protease in processing the polyproteins cannot be provided by the host and is essential to the life cycle of the retrovirus. In fact, it has been demonstrated that retroviruses which lack the protease or contain a mutated form of it, lack infectivity. See Katoh et al., Virology, 145, 280-92(1985), Crawford, et al., J.
Virol . , 53, 899-907(1985) and Debouck, et al., Proc. Natl . Acad. Sci . USA, 84, 8903-6(1987). Inhibition of retroviral protease, therefore, presents a method of therapy for
retroviral disease.
Methods to express retroviral proteases in E. coli have been disclosed by Debouck, et al., Proc. Natl . Acad. Sci .
USA, 8903-06(1987) and Graves, et al., Proc. Natl . Acad. Sci . USA, 85, 2449-53(1988) for the HIV-1 virus. The crystal structure of an HIV-1 protease has been disclosed by Miller et al . , Science, 246, 1149 (1989).
The method of isosteric replacement has been disclosedas a strategy for the development of protease inhibitors for HIV-1. Published European Patent applications EP-A 337 714, EP-A 352 000 and EP-A 357 332, EP-A 346 847, EP-A 342 541 and EP-A 393 445 are representative. Similar strategies have also been reported for inhibition of renin in U.S. Patents 4,713,445 and 4,661,473. There remains a need for protease- inhibiting compounds which have a favorable balance of potency and pharmacokinetics properties. The compounds of this invention have a unique 2,4 dihydroxy isostere wherein the hydroxyl group in the 2 position is further substituted to create a tertiary alcohol. The compounds are potent inhibitors of retroviral protease and are able to penetrate affected cells.
Dihydroxy isosteres have been reported as intermediates in the preparation of inhibitors of renin in Metternich et al . , Tet . Lett . , 29, 3923 (1988), Dellaria et al . , WO
87/04349, and Dellaria et. al . , J. Med. Chem. , 30, 1978, (1987).
SUMMARY OF THE INVENTION
This invention comprises compounds, hereinafter
described, of the formula (I), which inhibit the retroviral protease of HIV-1, and are useful for treating Acquired
Immunodeficiency Syndrome (AIDS).
This invention is also a pharmaceutical composition, which comprises a compound of formula (I) and a
pharmaceutically acceptable carrier.
This invention further constitutes a method for treating retroviral disease, which comprises administering to a mammal in need thereof an effective amount of a compound of formula
(I).
DETAILED DESCRIPTION OF THE INVENTION The peptides of this invention are illustrated by formula (I):
Figure imgf000006_0001
wherein
R1 is A, A-B or A-B-D;
A is R11, R11CO, R11- (CHR11 ' ) n-CO, R1 1OCO, R1 1OCH (R11 ' ) CO, R11NHCH ( R11 ' ) CO, R11SCH ( R11 ' ) CO, R1 1SO2, R1 1SO or R1 1C (O) CO;
B and D are
Figure imgf000006_0002
R2 and R4 are C1-6alkyl, optionally substituted with 1-5 fluorine atoms, C3-6alkenyl, C1-6alkyl-O-CH2 or (CH2)n-T, wherein T is phenyl, naphthyl, C5-6cycloalkyl or indolyl, optionally substituted with nitro, halogen, C1-4alkyl,
C1-4alkoxy, C1-4alkylthio, provided that R2 is not C1-6alkyl when R4 is cyclohexylmethyl;
R5 is H, C1-6alkyl, C1-6alkyl-O-CH2 or J-CH2(CH2)n, wherein J is CONHR', CO2R', NHR', SR', or phenyl, naphthyl, C5-6cycloalkyl or indolyl, optionally substituted with nitro, halogen, hydroxy, C1-4alkyl, C1-4alkoxy, C1-4alkylthio or trifluoromethyl;
R3 is G, X-G or X-Y-G; X and Y are
Figure imgf000006_0003
G is R10, NR'R10, NR'NR'-A, OR10 or SR10;
R10 is (CR11R11')n-W;
R11 and R11' are H, Alk, Ar, Het, Ar-C1-5alkyl,
Het-C1-5alkyl, C3-7cycloalkyl, Ar-C3-7cycloalkyl;
W is H, CH2OR", COR", OR11, OCOR11, NR'R11, NR'COR11, SR11, Ar or Het;
Alk is C1-6alkyl or Cι-6alkyl substituted by one or two hydroxy, nitro or C1-4alkoxy groups, or one to five fluoro atoms;
R' is H, C1-6alkyl or (CH2)n-Ar;
R" is H, C1-6alkyl or (CH2)n-Ph;
n is 0, 1 or 2; and q is 0 or 1;
or a pharmaceutically acceptable salt thereof.
Suitably R1 is A or A-B.
Suitably R2 is C1-6alkyl or (CH2)n-phenyl.
Suitably R3 is G. Typically R3 is NHCH(R11)-W, wherein R11 is H or Alk and W is CH2OH, CONH2 or phenyl.
Preferably R1 is A. More suitably R1 is C1-6alkoxycarbonyl. Preferably R2 is benzyl.
Preferably R3 is NHCH(i-pr)CH2OH.
Preferably R4 is benzyl.
Also included in this invention are pharmaceutically acceptable addition salts, complexes or prodrugs of the compounds of this invention. Prodrugs are considered to be any covalently bonded carriers which release the active parent drug according to formula (I) in vivo.
Ar indicates aryl, Ar is phenyl or naphthyl, or phenyl or naphthyl substituted by one to three C1-4alkyl, C1-4alkoxy, C1-4alkylthio, trifluoromethyl, nitro, mercapto, hydroxy, halogen, cyano, CO2R" or CON(R")2. Het indicates an
optionally substituted heterocyclic ring, and is a saturated or unsaturated, five or six membered ring or nine or ten-membered bicyclic ring, containing one to three heteroatoms chosen from the group of nitrogen, oxygen and sulfur, which are stable and available by conventional chemical synthesis. Illustrative heterocycles are pyridyl, furyl, thienyl, morpholinyl, pyrrolinyl, pyrrolidinyl, pyrazolyl,
pyrazolinyl, pyrazolidinyl, imidazolyl, imidazolinyl,
imidazolidininlyl, piperidinyl, pyrazinyl, piperazinyl, pyrimidinyl, pyridazinyl, tetrazolyl, oxazolyl, oxazolidinyl, isoxazolyl, isoxazolidinyl, indolyl, quinolinyl, benzofuryl, benzisoxazolyl and benzothienyl. Het is optionally
substituted with C1-4alkyl, C1-4alkoxy, C1-4alkylthio,
trifluoromethyl, nitro, mercapto, hydroxy, halogen, cyano, CO2R" or CON(R")2. Any accessible combination of up to three substituents on the phenyl, naphthyl or Het ring which is available by chemical synthesis and is stable is within the scope of this invention. Abbreviations and symbols commonly used in the peptide and chemical arts are used herein to describe the compounds of this invention. In general, the amino acid abbreviations follow the IUPAC-IUB Joint Commission on Biochemical
Nomenclature as described in Eur. J. Biochem. 158, 9 (1984).
3 1 3 1
Amino Acid letter letter Amino Acid letter letter
code code code code
Alanine Ala A Leucine Leu L
Arginine Arg R Lysine Lys K
Asparagine Asn N Methionine Met M
Aspartic AcidAsp D Phenylalanine Phe F
Cysteine Cys C Proline Pro P
Glutamine Gln Q Serine Ser S
Glutamic AcidGlu E Threonine Thr T
Glycine Gly G Tryptophan Trp W
Histidine His H Tyrosine Tyr y
Isoleucine Ile I Valine Val V
Asparagine or Aspartic Acid Asx B
Glutamine or Glutamic Acid Glx Z
In accordance with conventional representation, the amino terminus is on the left and the carboxy terminus is on the right. Unless specified otherwise, all chiral natural amino acids (AA) are assumed to be of the L absolute
configuration. Boc refers to the t-butyloxycarbonyl radical, Z and Cbz refer to the carbobenzyloxy radical, BrZ refers to the o-bromobenzyloxy-carbonyl radical, C1Z is the
p-chlorocarbobenzyloxy radical, C12Z refers to the 2,4-dichlorocarbobenzyloxy radical, Bn refers to the benzyl radical, Ac refers to acetyl, Ph refers to phenyl, BrMgDA refers to bromomagnesium diisopropylamide, DCC refers to dicyclohexyl-carbodiimide, DMAP refers to dimethylaminopyridine, HOBT refers to 1-hydroxybenzotriazole, OSU is
1-hydroxysuccinimide NMM is N-methylmorpholine, DTT is dithiothreitol, EDTA is ethylenediamine tetraacetic acid, DIEA is diisopropyl ethylamine, DBU is 1,8 diazobicyclo [5.4.0]undec-7-ene, DMSO is dimethylsulfoxide, DMF is dimethyl formamide, HMPA is hexamethylphosphoramide, DMAPEC is 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide
hydrochloride, DCC refers to dicyclohexylcarbodiimide, EDC refers to N-ethyl-N'(dimethylaminopropyl)-carbodiimide, PPA refers to 1-propanephosphonic acid cyclic anhydride, DPPA refers to diphenylphosphoryl azide, BOP refers to
benzotriazol-1-yloxy-tris(dimethylamino)phosphonium
hexafluorophosphate, TMS-Imid is N-trimethysilylimidazole, valinol is (2S)-2-amino-3-methyl-butanol and THF is
tetrahydrofuran. HF refers to hydrofluoric acid and TFA refers to trifluoroacetic acid. C1-6alkyl as applied herein is meant to include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tertiary butyl, n-pentyl, isopentyl arid hexyl. As used herein in the compounds of this invention.
When administered to an animal infected or potentially infected with a virus, which is dependent upon a virally encoded protease for processing of viral polyproteins, viral replication is inhibited, hence, disease progression is retarded.
Representative compounds of this invention are:
(2S,4S,5S) and (2R,4R,5R)-5-(t-butyloxycarbonylamino)-6-phenyl-2,4-dihydroxy-2-benzyl-1-oxo-hexyl-(benzyl)amide, (2S,4S,5S) and (2R,4R,5R)-5-(benzyloxycarbonyl-D-alanyl)amino-6-phenyl-2,4-dihydroxy-2-benzyl-1-oxo-hexyl- (benzyl)amide,
(2S, 4S, 5S) and (2R, 4R, 5R) -5- (benzyloxycarbonyl-L-alanyl) amino-6-phenyl-2 , 4-dihydroxy-2-benzyl-1-oxo-hexyl- (benzyl) amide,
(2S, 4S, 5S) and (2R, 4R, 5R) -5- (t-butyloxycarbonylamino) -6-phenyl-2, 4-dihydroxy-2-benzyl-1-oxo-hexyl- ( (2S) -2- (3-methyl-butanol))amide,
(2S,4S,5S) and (2R,4R,5R)-5-(t-butyloxycarbonylamino)-6-phenyl-2,4-dihydroxy-2-methyl-1-oxo-hexyl-((2S)-2-(3-methyl-butanol))amide,
(2S,4S,5S)-5-(t-butyloxycarbonylamino)-6-phenyl-2,4-dihydroxy-2-benzyl-1-oxo-hexyl-valinamide, (2S,4S,5S)-5-(t-butyloxycarbonylamino)-6-phenyl-2,4- dihydroxy-2-benzyl-1-oxo-hexyl-(2-(propan-1,3-diol))amide, N-((2S,4S,5S)-5-(t-butyloxycarbonylamino)-6-phenyl-2,4- dihydroxy-2-benzyl-1-oxo-hexyl)-N'-(benzyloxycarbonyl)- hydrazine,
N-((2S,4S,5S)-5-(t-butyloxycarbonylamino)-6-phenyl-2,4- dihydroxy-2-benzyl-1-oxo-hexyl)-hydrazine, and
(2S,4S,5S)-5-(benzoylformyl)amino-6-pheny1-2,4-dihydroxy-2- benzyl-1-oxo-hexyl-(benzyl)amide.
Preferred compounds of this invention are:
(2S,4S,5S)-5-(t-butyloxycarbonylamino)-6-phenyl-2,4- dihydroxy-2-benzyl-1-oxo-hexyl-(benzyl)amide,
(2S,4S,5S)-5-(t-butyloxycarbonylamino)-6-phenyl-2,4- dihydroxy-2-benzyl-1-oxo-hexyl-((2S)-2-(3-methyl- butanol))amide, and
(2S,4S,5S)-5-(t-butyloxycarbonylamino)-6-phenyl-2,4- dihydroxy-2-benzyl-1-oxo-hexyl-valinamide.
The chiral centers of the compounds of the invention may be racemic or nonracemic. Racemic mixtures, mixtures of diastereomers, as well as single diastereomers and
enantiomers of the compounds of the invention are included in the present invention. The terms "S" and "R" are used to denote the configuration of asymmetric carbons, and are used as defined by the IUPAC 1974 Recommendations for Section E, Fundamental Stereochemistry, Pure Appl. Chem., 45, 13 (1976). Where double bonds may be present in the compounds, these bonds may be of the Z (cis) or E (trans) configuration. When any variable substituent occurs more than once in formula (I), its definition in each occurrence is independent of its definition at every other occurrence. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
The peptides of this invention are prepared by
conventional methods of peptide synthesis from the compounds (II) and (III):
Figure imgf000011_0001
Figure imgf000011_0002
wherein Pr1 is an amino protecting group, Pr2 is a carboxy protecting group and Pr3 and Pr5 are H or a hydroxy
protecting group. These intermediate compounds are prepared by methods well known in the art, similar to those disclosed by Metternich et al . , Tet . Lett . , 29, 3923 (1988) and in WO 87/94349, which are incorporated fully herein by reference, and illustrated in Scheme 1. Acetyl, Boc or Cbz are
representative protecting groups for the amino group, and acetyl or trialkylsilyl are representative protecting groups for the hydroxyl group. Pr2 is generally OH, O-alkyl, O-benzyl or O-phenyl. Methods of general synthetic utility are found in U.S. Patents 4,661,473 and 4,713,445, published European Patent applications EP-A 352 000 and 337 714,
Holladay et al . , J. Med. Chem. , 30, 374 (1987) and Kempf, D., J. Org. Chem . , 51, 21, 3921 (1986), all of which are
incorporated fully herein by reference.
The compounds of formula (I) are prepared by:
1) (i) reacting a compound of the formula:
Figure imgf000011_0003
wherein Pr3 is H or a hydroxy-protecting group, and A,
B, D, R4 and R2 are as defined for formula (I), with any reactive groups protected,
with a coupling agent and a compound of the formula:
H-X-Y-G
wherein G, X and Y are as defined for formula (I), with any reactive groups protected, and
(ii) if necessary, removing any protecting groups; or
2) (i) reacting a compound of the formula: H-X-Y-G
wherein G, X and Y are as defined for formula (I), with any reactive groups protected,
with a base and a compound of the formula:
Figure imgf000012_0002
wherein Pr5 is H or a hydroxy-protecting group, and A, B, D, R4 and R2 are as defined for formula (I), with any reactive groups protected, and
(ii) if necessary, removing any protecting groups; or
3) (i) reacting a compound of the formula:
Figure imgf000012_0001
wherein Pr3 is H or a hydroxy-protecting group, and X, Y, G, R2 and R4 are as defined in formula (I), with any reactive groups protected,
with a coupling agent and a compound of the formula:
A-B-D-OH
wherein G, X, Y, A and R10 are as defined for formula (I), with any reactive groups protected, and
(ii) if necessary, removing any protecting groups.
Typical coupling agents include carbodiimides, such as DCC, EDC, DMAPEC, DPPA, PPA, and the BOP reagent, and
reagents which may be used to form activated esters,
anhydrides and acid halides, such as oxalyl chloride, thionyl chloride, N-hydroxysuccinimide and 4-nitrophenol. Many other such reagents are well know in the art. When G is R10, coupling reagents would additionally include organometallic reagents, such as lithium dimethyl copper which could be reacted with an acyl halide to form a ketone. Typical bases include organic nitrogenous bases, such as triethylamine or diisopropylamine, or an alkali metal
alkoxide. It will also be understood that when the reagent H-X-Y-G contains an amino group, it may act as the base itself and an additional base is not required.
Typical reactive groups which may require protection during synthesis are the hydroxyl, amino, carboxyl, carbonyl and mercapto groups. Conventional methods for protection and deprotection of reactive groups are disclosed in Greene,
Protective Groups in Organic Chemistry, John Wiley and Sons, New York, 1981.
Representative methods for making the compounds of this invention are illustrated in Scheme 1 hereinafter.
Figure imgf000014_0001
It will be apparent that, by starting the synthetic sequence with an appropriately substituted amino acid, such as (IV), the substituent R4 may be introduced into the isosteres (II) or (III) as any desired amino acid side chain, with any reactive groups suitably protected. Similarly, by choosing an appropriate electrophile, such as R2-X wherein X is an appropriate displaceable group (i.e., Cl, Br or I), R2 may be introduced into the isostere and varied as
appropriate.
An α-substituted, α-amino-aldehyde, such as (IV), is prepared by conventional means. For instance, a suitable α- amino acid may be reduced to its corresponding carboxaldehyde to initiate the preparation. Suitable methods are disclosed Coppola et al . , Asymmetric Synthesis, Construction of Chiral Molecules Using Amino Acids, Wiley Interscience, New York, 1987.
The aldehyde (IV) is condensed with the enolate derived from methyl pyruvate (V) and a suitable base, such as
bromomagnesium diisopropylamide or lithium diisopropylamide, to form an aldol adduct which undergoes intramolecular cyclization in situ to yield the dihydrofuran dione 1. This lactone exists exclusively as the enol lactone, as depicted in Scheme 1. It has been found that by using lithium
diisopropylamide as the base, the stereogenic integrity of the aldehyde is sometimes lost and the resulting enol lactone cannot reproducibly be obtained in enantiomerically pure form. The use of bromomagnesium diisopropylamide instead of lithium diisopropylamide affords lactone enaήtiomeri'cally pure. The stereochemistry of the two centers agrees with that described by Metternich et al . , Tet . Lett . , 29, 3923
(1988), although little or no undesired isomeric lactone is detected.
The enol lactone 1 is reduced with an appropriate reducing agent, such as hydrogen with a palladium catalyst to yield an α-hydroxy lactone as mixture of epimers at the hydroxyl position. The hydroxyl group is then protected with an appropriate protective group which is stable to strong base, such as a silyl ether. Subsequent treatment of the lactone with a strong base, such as lithium alkyl or
dialkylamide, and treatment with an electrophile R2-X, wherein X is a leaving group, such as chloride, bromide, iodide, or sulfonate, yields the appropriately α-R2
substituted lactone 4 diastereomerically pure. Reaction of the lactone with an appropriate nucleophile, such as an amine or hydroxide, provides the 2,4-dihydroxy-isostere.
Hydrolysis of the lactone yields the stable 2,4-dihydroxy carboxylic acid. The hydroxyl groups in the 2 and 4
positions can be protected, or the hydroxyl group at the 4 position can be selectively protected. A silyl ether is the most useful group for this purpose. The protected compound allows further modification of the carboxy terminus via amidation, esterification or coupling of an activated acyl intermediate, such as an acyl halide, with an organometallic reagent. Likewise, deprotecting the amino terminus allows further modification of the amine group by alkylation, acylation or sulfonylation.
Solution synthesis of the peptide bonds is accomplished using conventional methods for coupling the appropriate amino acid residues and optionally removing any protective groups. Suitable methods are disclosed, for instance, in Bodansky et al . , The Practice of Peptide Synthesis, Springer-Verlag, Berlin, 1984. Typically, a protected Boc-amino acid which has a free carboxyl group is coupled to a protected amino acid which has a free amino group using a suitable
carbodiimide coupling agent, such as N, N' dicyclohexyl carbodiimide (DCC) or 1-(3-dimethylaminopropyl)-3- ethylcarbodiimide hydrochloride (DMAPEC), optionally in the presence of catalysts such as 1-hydroxybenzotriazole (HOBT) and dimethylamino pyridine (DMAP). Suitable protective groups for amino acids and intermediates are disclosed in Greene, Protective Groups in Organic Chemistry, John Wiley and Sons, New York, 1981. Other methods for forming peptide bonds, such as the formation of activated esters, anhydrides or acid halides, of the free carboxyl of a protected Boc-amino acid, and subsequent reaction with the free amine of a protected amino acid, optionally in the presence of a base, are also suitable. For example, a protected Boc-amino acid or peptide is treated in an anhydrous solvent, such as methylene chloride or tetrahydrofuran (THF), in the presence of a base, such as N-methyl morpholine, DMAP or a trialkyl amine, with isobutyl chloroformate to form the "activated anhydride", which is subsequently reacted with the free amine of a second protected amino acid or peptide. The peptide formed by these methods may be deprotected selectively, using conventional techniques, at the amino or carboxy terminus and coupled to other peptides or amino acids using similar techniques. After the peptide has been completed, the protecting groups may be removed as hereinbefore described, such as by hydrogenation in the presence of a palladium or platinum catalyst, treatment with sodium in liquid ammonia, hydrofluoric acid or alkali.
Esters are often used to protect the terminal carboxyl group of peptides in solution synthesis. They may be
converted to carboxylic acids by treatment with an alkali metal hydroxide or carbonate, such as potassium hydroxide or sodium carbonate, in an aqueous alcoholic solution. The acids may be converted to other esters via an activated acyl intermediate as previously described.
The amides and substituted amides of this invention are prepared from carboxylic acids of the peptides in much the same manner. Thus, ammonia or a substituted amine may be reacted with an activated acyl intermediate to produce the amide. Use of coupling reagents, such as DCC or DMAPEC, is convenient for forming substituted amides from the carboxylic acid itself and a suitable amine.
In addition, the methyl esters of this invention may be converted to the amides, or substituted-amides, directly by treatment with ammonia, or a substituted amine, in methanol solution. A methanol solution of the methyl ester of the peptide is saturated with ammonia and stirred in a
pressurized reactor to yield the simple carboxamide of the peptides. Carboxamides and alcohols are preferred
embodiments of this invention due their enhanced stability relative to esters. If the final peptide, after it has been deprotected, contains a basic group, an acid addition salt may be
prepared. Acid addition salts of the peptides are prepared in a standard manner in a suitable solvent from the parent compound and an excess of an acid, such as hydrochloric, hydrobromic, sulfuric, phosphoric, acetic, maleic, succinic or methanesulfonic . The acetate salt form is especially useful. If the final peptide contains an acidic group, cationic salts may be prepared. Typically the parent compound is treated with an excess of an alkaline reagent, such as a hydroxide, carbonate or alkoxide, containing the appropriate cation. Cations such as Na+, K+, Ca++ and NH4 + are examples of cations present in pharmaceutically
acceptable salts. Certain of the compounds form inner salts or zwitterions which may also be acceptable.
The compounds of formula (I) are used to induce antiviral activity in patients which are infected with
susceptible viruses and require such treatment. In
particular, the this invention is a method of treating disease resulting from infection by retroviruses by
administering a compound of this invention. The Human
Immunodeficiencey Virus, type 1 (HIV-1) has been show to be susceptible to inhibition by the compounds of this invention. Thus, in one preferred embodiment this invention is a method of treating Acquired Immune Deficiency Syndrome (AIDS). The method of treatment comprises the administration orally, parenterally, buccally, trans-dermally, rectally or by insufflation, of an effective quantity of the chosen
compound, preferably dispersed in a pharmaceutical carrier. Dosage units of the active ingredient are generally selected from the range of 0.01 to 25 mg/kg, but will be readily determined by one skilled in the art depending upon the route of administration, age and condition of the patient. These dosage units may be administered one to ten times daily for acute or chronic infection.
The protease inhibiting properties of the peptides of this invention, are demonstrated by their ability to inhibit the hydrolysis of a peptide substrate by rHIV protease in the range of about 20 nM to about 60 μM. The following table is representative of the inhibition constants of the compounds of this invention.
Compd # R1 R2 R3 Ki (μM)
5 Boc CH2Ph NH-CH2Ph 0.3
6 Cbz-D-Ala CH2Ph NH-CH2Ph 7.1
7 Cbz-L-Ala CH2Ph NH-CH2Ph 0.53
9 Boc CH2Ph NH-CH(i-pr)CH2OH 0.05
12 Boc Me NH-CH(i-pr)CH2OH 53
13 Boc CH2Ph Val-NH2 0.022
14 Boc CH2Ph NH-CH(CH2OH)2 2.1
15 Boc CH2Ph NHNH-Cbz 37
16 Boc CH2Ph NHNH2 15.6
17 PhCOCO CH2Ph NHBzl 4.1
Compounds of this invention in which R1 is A, and R2 and R4 are benzyl, and have notably superior activity.
Pharmaceutical compositions of the peptides of this invention, or derivatives thereof, may be formulated as solutions or lyophilized powders for parenteral
administration. Powders may be reconstituted by addition of a suitable diluent or other pharmaceutically acceptable carrier prior to use. The liquid formulation is generally a buffered, isotonic, aqueous solution. Examples of suitable diluents are normal isotonic saline solution, standard 5% dextrose in water or buffered sodium or ammonium acetate solution. Such formulation is especially suitable for parenteral administration, but may also be used for oral administration or contained in a metered dose inhaler or nebulizer for insufflation. It may be desirable to add excipients such as polyvinylpyrrolidone, gelatin, hydroxy cellulose, acacia, polyethylene glycol, mannitol, sodium chloride or sodium citrate.
Alternately, these peptides may be encapsulated, tableted or prepared in a emulsion or syrup for oral
administration. Pharmaceutically acceptable solid or liquid carriers may be added to enhance or stabilize the
composition, or to facilitate preparation of the composition. Liquid carriers include syrup, peanut oil, olive oil, glycerin, saline and water. Solid carriers include starch, lactose, calcium sulfate dihydrate, terra alba, magnesium stearate or stearic acid, talc, pectin, acacia, agar or gelatin. The carrier may also include a sustained release material such as glyceryl monostearate or glyceryl
distearate, alone or with a wax. The amount of solid carrier varies but, preferably, will be between about 20 mg to about 1 g per dosage unit. The pharmaceutical preparations are made following the conventional techniques of pharmacy involving milling, mixing, granulating, and compressing, when necessary, for tablet forms; or milling, mixing and filling for hard gelatin capsule forms . When a liquid carrier is used, the preparation will be in the form of a syrup, elixir, emulsion or an aqueous or non-aqueous suspension. Such a liquid formulation may be administered directly p.o. or filled into a soft gelatin capsule.
For rectal administration, a pulverized powder of the peptides of this invention may be combined with excipients such as cocoa butter, glycerin, gelatin or polyethylene glycols and molded into a suppository. The pulverized powders may also be compounded with an oily preparation, gel, cream or emulsion, buffered or unbuffered, and administered through a transdermal patch.
Beneficial effects may be realized by co-administering, individually or in combination, other anti-viral agents with the protease inhibiting compounds of this invention.
Examples of anti-viral agents include nucleoside analogues, phosphonoformate, rifabutin, ribaviran, phosphonothioate oligodeoxynucleotides, castanospermine, dextran sulfate, alpha interferon and ampligen. Nucleoside analogues, which include 2',3'-dideoxycytidine(ddC), 2',3'-dideoxyadenine(ddA) and 3'-azido-2',3'-dideoxythymide(AZT), are especially useful. AZT is one preferred agent. Suitably pharmaceutical compositions comprise an anti-viral agent, a protease
inhibiting peptide of this invention and a pharmaceutically acceptable carrier. The Examples which follow serve to further illustrate this invention. The Examples are intended to in no way limit the scope of this invention, but are provided to show how to make and use the novel compounds of this invention.
Purification of Recombinant HIV Protease
Methods for expressing recombinant HIV protease in E. coli have been described by Debouck, et al., Proc. Natl.
Acad. Sci. USA, 84, 8903-6(1987). The enzyme used to assay the peptide of this invention was produced in this manner and purified from the cell pellet as follows. The E. coli cell pellet was resuspended in a buffer consisting of 50 mM Tris- HCl, pH 7.5; 1.0 mM each DTT, EDTA and PMSF
(phenylmethylsulfonyl fluoride). The cells were lysed by sonication and insoluble material was removed by
centrifugation at 15,000 x g av, for 15 min. The clarified supernatant was then brought to 40% of saturation with ammonium sulfate. This suspension was stirred at room temperature for 30 min. and then centrifuged as above. The resulting precipitate was redissolved/resuspended in a minimal volume of 20 mM Tris-HCl, pH 7.5; 200 mM NaCl; 0.1 mM each DTT and EDTA. The sample was centrifuged again before application (in 5 ml aliquots) to a Beckman TSK G2000SW preparative HPLC gel filtration column (2.1 cm x 60 cm.).
The column was equilibrated in the same buffer at a flow rate of 4 ml/min. The effluent of the column was monitored at 280 nm and 1 min. fractions collected. Typically, the rHIVPRT (recombinant HIV protease) eluted 45-46 min. into the run. At this stage, the protease was 85-95% pure. By immunoblot analysis >90% of the immunoreactive material was precipitated at the ammonium sulfate step. By activity assay, the highest peak of activity was found in the fractions collected at 45 and 46 minutes. Analysis of the TSK column fractions by RP-HPLC and SDS-PAGE indicated that the majority of the 11,000 Mr protein is also found in fractions 45 and 46. The
activity itself cannot be used to obtain reliable recovery data as it is influenced by high salt, i.e., with increasing salt, increasing levels of activity were obtained. Thus, with each step in the purification, more total activity was recovered than was started with. The overall yield of rHIVPRT was N1 mg from a 50 gm E. coli cell pellet.
Inhibition of HIV protease activity
Assays for inhibition of HIV-1 protease activity are well known in the art and are disclosed for instance in Moore et al . , Biochem. Biophys . Res. Comm. , 159, 420 (1989) and
Kohl et al . , Proc. Natl . Acad. Sci . USA, 85, 4686 (1988). A typical assay contained 10 mL MENDT buffer (50 mM Mes (pH 6.0; 2-(N-morpholino)ethanesulfonic acid), 1 mM EDTA, 1 mM dithiothreitol, 200 mM NaCl, 0.1% Triton X-100); 2, 3, or 6 mM N-acetyl-L-arginyl-L-alanyl-L-seryl-L-glutaminyl-L- asparaginyl-L-tyrosyl-L-prolyl-L-valyl-L-valinamide (Ac-Arg- Ala-Ser-Gln-Asn-Tyr-Pro-Val-Val-NH2; Km=7 mM); and micromolar and sub-micromolar concentrations of synthetic compounds.
Following incubation at 37°C for several .minutes, the
reaction was initiated with purified 0.01-1 mg HIV protease. Reaction mixtures (37°C) were quenched after 10-20 minutes with an equal volume of cold 0.6 N trichloroacetic acid, and, following centrifugation to remove precipitated material, peptidolysis products were analyzed by reverse phase HPLC (Beckman Ultrasphere® ODS, 4.5 mm × 25 mm; mobile phase: 5- 20% acetonitrile/H2O-0.1% TFA (15 min), 20% acetonitrile/H2O- 0.1% TFA (5 min) at 1.5 mL/min, detection at 220 nm. The elution positions of Ac-Arg-Ala-Ser-Gln-Asn-Tyr-Pro-Val-Val-NH2 (17-18 min) and Ac-Arg-Ala-Ser-Gln-Asn-Tyr (10-11 min) were confirmed with authentic material. Initial rates of Ac- Arg-Ala-Ser-Gln-Asn-Tyr formation were determined from integration of these peaks, and typically, the inhibitory properties of the synthetic compounds were determined from slope/intercept analysis of a plot of 1/v vs. [inhibitor] (Dixon analysis). Ki values resulting from this type of primary analysis are accurate for competitive inhibitors only, and under conditions in which the Michaelis constant of the substrate used is well-determined. The Examples which follow serve to illustrate this invention. The Examples are intended to in no way limit the scope of this invention, but are provided to show how to make and use the compounds of this invention.
In the Examples, all temperatures are in degrees
Centigrade. Ultrasphere® and Ultrasphere® ODS are silica gel and octadecylsilane chromatographic supports,
respectively, manufactured by Beckman Instruments Inc.,
Fullerton, CA. Microsorb® is a silica gel chromatographic support manufactured by Rainin Instruments Co., Woburn, Mass. Bakerbond® is a silica gel chromatographic support
manufactured by J.T. Baker Chemical Co., Phillipsburg, N.J. Celite® is filter aid composed of acid washed diatomaceous silica manufactured by Mansville Corp., Denver, Colorado.
FAB mass spectra were performed upon a VG Zab mass
spectrometer using fast atom bombardment.. NMR were recorded at 250 MHz using a Bruker AM 250 spectrometer.
Multiplicities indicated are: s=singlet, d=doublet,
t=triplet, q=quartet, m=multiplet and br indicates a broad signal.
Example 1 Preparation of (2S,4S,5S)-5-(t-butyloxycarbonylamino)-6-phenyl-2,4-dihydroxy-2-benzyl-1-oxo-hexyl-(benzyl)amide, and (2R,4R,5R)-5-(t-butyloxycarbonylamino)-6-phenyl-2,4-dihydroxy-2-benzyl-1-oxo-hexyl-(benzyl)amide (5) a) (5R)-5-((1R)-1-t-butyloxycarbonylamino-2-phenyl)ethyl-4H,5H-dihydrofuran-2,3-dione, and
(5S)-5-((1S)-1-t-butyloxycarbonylamino-2-phenyl)ethyl-4H,5H-dihydrofuran-2,3-dione (1).
A solution of Boc-(JC)-phenylalanine methyl ester (9.86 g, 0.0353 mol) in toluene (150 mL) under an argon atmosphere was cooled to -78° C. Diisobutyllithium aluminum hydride (1.0 M solution in hexane, 67 mL, 0.067 mol) was added dropwise by addition funnel over 2 h. After complete addition, the reaction mixture was quenched with methanol, and poured into a 10% solution of sodium potassium tartrate. The two layers were allowed to stir for 2 h and separated. The aqueous layer was extracted 3 X with ethyl acetate. The combined organic layers were washed twice with 0.05 M HCl, brine, dried over MgSO4 and concentrated to a colorless oil, Boc-phenylalanine-carboxaldehyde. A solution of lithium diisopropylamide (2.5 M in hexane, 42.4 mL, 0.106 mol) was diluted with THF (600 mL) and cooled to -78° C. To this mixture was added a solution of methyl pyruvate (10.8 g,
0.106 mol), HMPA (19 g, 0.106 mol) in THF (200 mL) over a 2 h period by addition funnel. After complete addition, the reaction mixture was allowed to stir an additional 30 min. To this reaction mixture was added a solution of Boc- phenylalanine-carboxaldehyde in THF (150 mL) via cannula. The reaction mixture was allowed to warm and stir to room temperature over 20 h. The mixture was poured into ice cold 1 N HCl. The aqueous layer was extracted 3 X with ethyl acetate. The combined organic layers were washed with cold 1N HCl, brine, dried over MgSO4 and concentrated to an orange oil. The product was crystallized from methylene
chloride/hexane, filtered and washed with hexane/Et2O (2:1), to afford a fine white solid. Performing the same procedure, except using
bromomagnesium diisopropylamide in place of lithium
diisopropylamide as the base, yielded the enantiomerically pure product la, which was:
(5S)-5-((1S)-1-t-butyloxycarbonylamino-2-phenyl)ethyl-4H,5H-dihydrofuran-2,3-dione (1a).
The product was determined to be enantiomerically pure by HPLC (Bakerbond® OB, 3% ethanol/hexane, 0.8 mL/min): 1H NMR (CDCI3, 250 MHz) 51.38 (s, 9H), 3.02 (m, 2H), 4.19 (m,
1H), 4.89 (bs, 1H), 6.15 (bs, 1H), 7.3 (m, 5H) b) (5S)-5-((lS)-1-t-butyloxycarbonylamino-2-phenyl)ethyl-3-hydroxy-tetrahydrofuran-2-one, and
(5R)-5-((1R)-1-t-butyloxycarbonylamino-2-phenyl)ethyl-3-hydroxy-tetrahydrofuran-2-one (2)
To a solution of lactone 1 (10 g, 0.031 mol) in ethyl acetate (250 mL) was added 10% Pd/C (8 g). The reaction mixture was degassed and one atmosphere of H2 was applied using a balloon. The reaction was allowed to stir at room temperature for 24 h, purged with Ar, and filtered through Celite® washing with ethyl acetate. The organic layer was concentrated to a white solid (10 g, 99%). The product was determined to be a 10:1 mixture of isomers at the alcohol position by NMR: 1H NMR (CDCI3, 250 MHz, major isomer) δ 1.40
(s, 9H), 2,12 (m, 1H), 2.49 (m, 1H), 2.95 (m, 2H), 4.03 (m, 1H), 4.36 (m, 1H), 4.49 (m, 1H), 4.75 (m, 1H), 7.3 (m, 5H) c) (5S)-5-((1S)-1-t-butyloxycarbonylamino-2-phenyl)ethyl-3-trimethysilyloxy-tetrahydrofuran-2-one, and
(5R)-5-((1R)-1-t-butyloxycarbonylamino-2-phenyl)ethyl-3-trimethysilyloxy-tetrahydrofuran-2-one (3)
A solution of (±)-hydroxylactone 2 (1 g, 3.12 mmol) in methylene chloride (25 mL) was treated with N-trimethylsilylimidazole (TMSImid) (0.873 g, 6 mmol). The reaction mixture was allowed to stir at room temperature for 24 h. The mixture was diluted with methylene chloride, washed with 0.05 M HCl, brine, dried over MgS04, and
concentrated to yield a white solid (1.2 g, 99%).
1H NMR (CDCI3, 250 MHz) δ 0.19 (s, 9H), 1.41 (s, 9H), 2.10
(m, 1H), 2.38 (m, 1H), 2.95 (m, 2H), 3.99 (m, 1H), 4.28 (m, 1H), 4.45 (m, 1H), 7.3 (m, 5H)
Performing steps 1(b) and 1(c), except substituting the enantiomerically pure (5S)-5-((1S)-1-t-butyloxycarbonylamino-2-phenyl)ethyl-4H,5H-dihydrofuran-2,3-dione 1a in step 1(b), (5S)-5-((1S)-1-t-butyloxycarbonylamino-2-phenyl)ethyl-3-trimethysilyloxy-tetrahydrofuran-2-one (3a) was prepared. d) (3S,5S)-5-((1S)-1-t-butyloxycarbonylamino-2-phenyl)ethyl- 3-trimethysilyloxy-3-benzyl-tetrahydrofuran-2-one, and
(3R,5R)-5-((1R)-1-t-butyloxycarbonylamino-2-phenyl)ethyl-3- trimethysilyloxy-3-benzyl-tetrahydrofuran-2-one (4)
A solution of lithium diisopropylamide (1.5 M solution in cyclohexane, 7.44 mL, 11.16 mmol) in THF (50 mL) was cooled to -78° C under an argon atmosphere. To this was added a solution of 3 (1.1 g, 2.79 mmol) in THF (25 mL) via cannula. The reaction mixture was allowed to stir 15 min. HMPA (2 g, 11.16 mmol) was added via syringe and the reaction mixture was allowed to stir for 15 min. Benzyl bromide (1.9 g, 11.16 mmol) was added via syringe and the reaction mixture was allowed to stir for 3 h allowing the mixture to warm to -50°C. The reaction mixture was poured into 0.05 M HCl, and separated. The aqueous layer was extracted ethyl acetate (3X), the combined organic layers were washed with 0.05 M HCl, brine, dried over MgSO4, and concentrated to a yellow oil. This was chromatographed (silica gel, 10:1 hexane:ethyl acetate) to afford a white crystalline solid (620 mg,46%): 1H NMR (CDC13, 250 MHz) δ 0.18 (s, 9H), 1.38 (s, 9H) , 2.13 (m, 1H), 2.30 (m, 1H), 2.70 (m, 2H), 2.98 (m, 2H), 3.53 (m, 1H), 3.82 (m, 1H), 4.60 (m, 1H), 7.2 (m, 10H)
Performing steps 1 (b) through 1(d), except using the enantiomerically pure (5S)-5-((1S)-1-t-butyloxycarbonylamino- 2-phenyl)ethyl-4H,5H-dihydrofuran-2,3-dione, 1a, in step 1(b), (3S,5S)-5-((1S)-1-t-butyloxycarbonylamino-2-phenyl)ethyl-3-trimethysilyloxy-3-benzyl-tetrahydrofuran-2- one (4a) was prepared. e) (2S,4S,5S)-5-(t-butyloxycarbonylamino)-6-phenyl-2,4-dihydroxy-2-benzyl-1-oxo-hexyl-(benzyl)amide, and
(2R,4R,5R)-5-(t-butyloxycarbonylamino)-6-phenyl-2,4-dihydroxy-2-benzyl-1-oxo-hexyl-(benzyl)amide (5)
To a solution of benzylamine (212 mg, 1.98 mmol) in THF (20 mL) at -78° C under an argon atmosphere was added
n-butyllithium (1.6M solution in hexane, 1.25 mL, 1.98 mmol). The reaction mixture was allowed to stir for 20 min. To this mixture was added a solution of 4 (240 mg, 0.495 mmol) in THF (10 mL) via cannula. The reaction mixture was allowed to stir for 1 h, poured into 0.05 M HCl, and extracted with ethyl acetate (3X). The organic layers were washed with 0.05 M HCl, brine, dried over MgSO4, and concentrated to a white solid. The solid was recrystallized from ethyl acetate, to afford the product (220 mg,86%):
1H NMR (CDCl3, 250 MHz) δ 1.39 (s, 9H), 1.80 (m, 1H), 2.23
(m, 1H), 2.7-3.2 (m, 4H), 3.39 (m, 1H), 3.90 (m, 1H), 4.32 (m, 2H), 4.76 (m, 1H), 5.02 (m, 1H), 7.2 (m, 15H); MS DCI/NH3 m/e 519[M+H]+
Performing steps 1(b) through 1(e), except using
enantiomerically pure (5S)-5-((1S)-1-t-butyloxycarbonylamino-2-phenyl)ethyl-4H,5H-dihydrofuran-2,3-dione, 1a, in step 1(b), (2S,4S,5S)-5-(t-butyloxycarbonylamino)-6-phenyl-2,4-dihydroxy-2-benzyl-1-oxo-hexyl-(benzyl)amide (5a) was prepared. Example 2
Preparation of (2S,4S.5S)-5-(benzyloxycarbonyl-D-alanvl)amino-6-phenyl-2,4-dihydroxy-2-benzyl-1-oxo-hexyl- (benzyl)amide, and
(2R,4R,5R)-5-(benzyloxγcarbonyl-D-alanγl)amino-6-pheny1-2,4-dihydroxy-2-benzyl-1-oxo-hexyl-(benzyl)amide (6)
To a solution of 5 (73 mg, 0.141 mmol) in methylene chloride (3 mL) was added trifluoroacetic acid (3 mL). The reaction mixture was allowed to stir for 1 h at room
temperature. The mixture was concentrated and dried under high vacuum. The residue was diluted with methylene chloride (5 mL) and treated with triethylamine (16 mg, 0.15 mmol).
The reaction mixture was cooled to 0°C and Cbz-D-Ala (34 mg, 0.15 mmol), 1-hydroxybenzotriazole (HOBT) (21 mg, 0.15 mmol), and 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide
hydrochloride (DMAPEC) (29 mg, 0.15 mmol) were added. The reaction mixture was allowed to stir at 0°C for 1 h, and the UBSTITUTE SHEET ice bath removed. The mixture was allowed to warm and stir to room temperature for 16 h. The mixture was diluted with methylene chloride, washed with 10% NaHCO3, 0.05 M HCl, brine, dried over MgSO4, and concentrated to an oily solid. The product was precipitated from methylene chloride/pentane, to afford a white powder (27 mg, 31%). NMR shows that this is a mixture of two diastereomers. Partial resolution of the isomers can be obtained by HPLC (Rainin Microsorb® Si02/ 3%
2-propanol/methylene chloride):
1H NMR (mixture of diastereomers, DMSO, 250 MHz) δ 0.92 (d,
1.5H, J=6Hz), 1.12 (d, 1.5H, J=6Hz), 1.65 (m, 1H), 1.94 (m, 1H), 2.55-3.1 (m, 4H), 3.77 (m, 2H), 4.02 (m, 2H), 4.2 (m, 1H), 5.00 (2, 2H), 5.60 (m, 1H), 7.2 (m, 20H), 7.6 (m, 1H), 8. 13 (m, 1H)
Example 3
Preparation of (2S,4S, 5S) -5- (benzyloxycarbonyl-L- alanyl) amino-6-pheny1-2 , 4-dihydroxy-2-benzyl-1-oxo-hexyl- (benzyl)amide, and
(2R,4R,5R)-5-(benzvloxycarbonyl-L-alanyl)amino-6-phenyl-2,4- d.hydroxy-2-benzyl-1-oxo-hexyl-(benzyl)amide (7)
To a solution of 5 (82 mg, 0.164 mmol) in methylene chloride (3 mL) was added trifluoroacetic acid (3 mL). The reaction mixture was allowed to stir for 1 h at room
temperature. The mixture was concentrated and dried under high vacuum. The residue was diluted with methylene chloride (5 mL) and treated with triethylamine (18 mg, 0.18 mmol).
The reaction mixture was cooled to 0°C and Cbz-L-Ala (40 mg, 0.18 mmol), HOBT (24 mg, 0.18 mmol), and DMAPEC (35 mg, 0.18 mmol) were added. The reaction mixture was allowed to stir at 0°C for 1 h, and the ice bath was removed. The mixture was allowed to warm and stir to room temperature for 16 h. The mixture was diluted with methylene chloride, washed with 10% NaHCO3, 0.05 M HCl, brine, dried over MgSO4, and
concentrated to an oily solid. The product was precipitated from methylene chloride/pentane, to afford a white powder (23 mg, 23%) NMR shows that this is a mixture of two
diastereomers. Partial resolution of the isomers can be obtained by HPLC (Ranin Microsorb® SiO2, 3% 2-propanol/methylene chloride): 1H NMR (mixture of diastereomers, DMSO, 250 MHz) δ 0.92 (d, 3H, J=6Hz), 1.12 (d, 1.5H, J=6Hz), 1.65
(m, 1H), 1.94 (m, 1H), 2.55-3.1 (m, 4H), 3.77 (m, 2H), 4.02 (m, 2H), 4.2 (m, 1H), 5.00 (2, 2H), 5.60 (m, 1H), 7.2 (m, 20H), 7.6 (m, 1H), 8. 13 (m, 1H); MS DCI/NH3 m/e 624[M+H]+ Example 4
Preparation of (2S,4S,5S)-5-(t-butyloxycarbonylamino)-6-phenyl-2,4-dihydroxy-2-benzyl-1-oxo-hexyl-((2S)-2-(3-methyl-butanol))amide, and (2R,4R,5R)-5-(t-butyloxycarbonylamino)-6-phenyl-2.4-dihydroxy-2-benzyl-1-oxo-hexyl-((2S)-2-(3-methyl-butanol))amide (9) a) (2S,4S,5S)-5-(t-butyloxycarbonylamino)-6-phenyl-4-trimethylsilyloxy-2-hydroxy-2-benzyl-hexanoic acid, and
(2R,4R,5R)-5-(t-butyloxycarbonylamino)-6-phenyl-4-trimethylsilyloxy-2-hydroxy-2-benzyl-hexanoic acid (8)
A solution of 4 (310 mg, 0.64 mmol) in THF/Water (1/1, 20 mL) was treated with 2.5 N NaOH (0.540 mL, 1.34 mmol) and allowed to stir at room temperature for 30 min. The mixture was concentrated to a white solid, diluted with 0.05N HCl, and extracted with ethyl acetate (3X). The organic layers were washed with brine, dried over MgS04 and concentrated to a white solid. The solid was dissolved in TMSImid (3 mL) and was heated to 80°C for 16 h. The reaction mixture was cooled, diluted with ethyl acetate, washed with 0.05 N HCl (3X), brine, dried over MgSO4, and concentrated to an oil. The acid was precipitated from the oil with pentane to afford a white solid (278 mg, 87%):
1H NMR (CDCI3, 250 MHz) δ 0.21 (s, 8H), 1.38 (bs, 9H), 2.00 (m, 1H), 2.22 (m, 1H), 2.7-3.1 (m, 4H), 3.9-4.2 (m, 2H), 4.72 (m, 1H), 7.2 (m, 10H) Performing the same reaction, except using the
enantiomerically pure 4a, the compound (2S,4S,5S)-5-(t- butyloxycarbonylamino)-6-phenyl-4-trimethylsilyloxy-2- hydroxy-2-benzyl-hexanoic acid (8a) was prepared. b) (2S,4S,5S)-5-(t-butyloxycarbonylamino)-6-phenyl-2,4- dihydroxy-2-benzyl-1-oxo-hexyl-((2S)-2-(3-methyl- butanol))amide, and
(2R,4R,5R)-5-(t-butyloxycarbonylamino)-6-phenyl-2,4- dihydroxy-2-benzyl-1-oxo-hexyl-((2S)-2-(3-methyl- butanol))amide (9)
To a solution of 8 (110 mg, 0.22 mmol) in methylene chloride (10 mL) at 0°C was added, HOBT (32 mg, 0.24 mmol), DMAPEC (46 mg, 0.24 mmol), and valinol (25 mg, 0.24 mmol).. The reaction was allowed to stir at 0°C for 1 h, and allowed to warm to room temperature and stir for 16 h. The reaction mixture was diluted with methylene chloride, washed with 0.05 N HCl, brine, dried over MgSO4, and concentrated to a yellow oil. Crystallization from methylene chloride/pentane affords Isomer A (7 mg, 6%). The mother liquor was concentrated and chromatographed (1:1 hexane:ethyl acetate). The major fractions were the hydroxylactone, 4 (50 mg, 45%) and Isomer B. Isomer B was recrystallized from methylene
chloride/pentane to afford a white solid (12 mg, 11%).
Isomer A
1H NMR (CDCI3, 250 MHz) δ 0.70 (dd, 6H, J=7Hz, 28Hz), 1.12
(m, 1H), 1.39 (s, 9H), 1.62 (m, 1H), 1.85 (m, 1H), 2.17 (sm, 1H), 2.7-3.1 (m, 3H), 3.50 (m, 4H), 3.90 (m, 1H), 4.77 (m, 2H), 5.12 (m, 1H), 6.7-7.4 (m, 10H)
Isomer B
1H NMR (CDCI3, 250 MHz) δ 0.80 (dd, 6H, J=7Hz, 22Hz), 1.41
(s, 9H), 1.5-1.9 (m, 3H), 2.27 (m, 1H), 2.7-3.1 (m, 3H), 3.40
(m, 4H), 3.85 (m, 1H), 4.82 (m, 2H), 5.57 (m, 1H), 6.7-7.4
(m, 10H); MS DCI/NH3 m/e 515[M+H]+ Example 5
Preparation of (2S,4S,5S)-5-(t-butvloxycarbonylamino)-6- phenyl-2,4-dihydroxy-2-benzyl-1-oxo-hexyl-((2S)-2-(3-methyl- butanol))amide (9)
To a solution of (2S,4S,5S)-5-(t-butyloxycarbonylamino)- 6-phenyl-4-trimethylsilyloxy-2-hydroxy-2-benzyl-hexanoic acid 8a (320 mg, 0.64 mmol) in methylene chloride (10 mL) at 0°C was added, N-hydroxysuccinimide (80 mg, 0.7 mmol), DMAPEC (135 mg, 0.7 mmol), and valinol (72 mg, 0.7 mmol). The reaction was allowed to stir at 0°C for 1 h, and allowed to warm to room temperature and stir for 16 h. The reaction mixture was diluted with methylene chloride, washed with 0.05N HCl, brine, dried over MgSO4, and concentrated to a yellow oil. Crystallization from methylene chloride/pentane affords a white solid (15 mg, 5%). The mother liquor was concentrated and chromatographed (1/1 hexane/ethyl acetate) to afford an additional 75 mg of product (27%) and 140 mg of 4a (52%): 1H NMR (CDCI3, 250 MHz) δ 0.80 (dd, 6H, J=7Hz,
22Hz), 1.41 (s, 9H), 1.5-1.9 (m, 3H), 2.27 (m, 1H), 2.7-3.1 (m, 3H), 3.40 (m, 4H), 3.85 (m, 1H), 4.82 (m, 2H), 5.57 (m, 1H), 6.7-7.4 (m, 10H) Example 6
Preparation of (2S,4S,5S)-5-(t-butyloxycarbonylamino)-6-phenyl-2,4-dihydroxy-2-methyl-1-oxo-hexyl-((2S)-2-(3-methyl-bntanol))amide (12) a) (3S,5S)-5-((1S)-1-t-butyloxycarbonylamino-2-phenyl)ethyl-3-trimethysilyloxy-3-methyl-tetrahydrofuran-2-one (10)
A solution of lithium diisopropylamide (1.5 M solution in cyclohexane, 4.4 mL, 6.6 mmol) in THF (40 mL) was cooled to -78° C under an argon atmosphere. A solution of
(2S,4S,5S)-5-(benzyloxycarbonyl-L-alanyl)amino-6-phenyl-2,4-dihydroxy-2-benzyl-1-oxo-hexyl-(benzyl)amide 3a (0.65 g, 1.65 mmol) in THF (20 mL) was added via cannula. The reaction mixture was allowed to stir 15 min. HMPA (0.935 g, 6.6 mmol) was added via syringe and the reaction mixture was allowed to stir for 15 min. Methyliodide (1.2 g, 6.6 mmol) was added via syringe and the reaction mixture was allowed to stir for 3 h allowing the mixture to warm to -50°C. The reaction mixture was poured into 0.05 M HCl, and separated. The aqueous layer was extracted ethyl acetate (3X), the combined organic layers were washed with 0.05 M HCl, brine, dried over MgSθ4, and concentrated to a yellow oil. This was
chromatographed (silica gel, 7:1 hexane:ethyl acetate) to afford a white crystalline solid (280 mg, 41%) and 80 mg of a mixture of alkylated/unalkylated lactone: 1H NMR (CDCI3, 250 MHz) δ 0.18 (s, 9H), 1.38 (s, 9H), 1.43 (s, 3H), 2.2 (m,
2H),.2.95 (m, 2H), 3.98 (m, 1H), 4.25 (m, 1H), 4.68 (m, 1H), 7.25 (m, 5H) b) (2S,4S,5S)-5-(t-butyloxycarbonylamino)-6-phenyl-4- trimethylsilyloxy-2-hydroxy-2-methyl-hexanoic acid (11)
A solution of 10 (280 mg, 0.64 mmol) in THF/water (1/1, 6 mL) was treated with 2.5N NaOH (0.540 mL, 1.34 mmol) and allowed to stir at room temperature for 30 min. The mixture was concentrated to a white solid, diluted with 0.05N HCl, and extracted 3 X with ethyl acetate. The organic layers were washed with brine, dried over MgSO4 and concentrated to a white solid. The solid was dissolved in TMSImid (3 mL) and was heated to 80°C for 16 h. The reaction mixture was cooled, diluted with ethyl acetate, washed 3 X with 0.05N HCl, brine, dried over MgSO4, and concentrated to a yellow oil (225 mg, 77%). NMR of the oil showed that this to be correct product although further attempts to purify it were unsuccessful. c) (2S,4S,5S)-5-(t-butyloxycarbonylamino)-6-phenyl-2,4-dihydroxy-2-methyl-1-oxo-hexyl-((2S)-2-(3-methyl-butanol))amide (12)
To a solution of crude 10 (115 mg, 0.26 mmol) in
methylene chloride (10 mL) at 0°C was added,
hydroxysuccinimide (30 mg, 0.26 mmol), DMAPEC (50 mg, 0.26 mmol), and valinol (29 mg, 0.26 mmol). The reaction was allowed to stir at 0°C for 1 h, and allowed to warm to room temperature and stir for 16 h. The reaction mixture was diluted with methylene chloride, washed with 0.05N HCl, brine, dried over MgSθ4, and concentrated to a yellow oil.
Chromatography (silica gel, ethyl acetate) affords a white solid (6 mg, 5%): 1H NMR (CDCl3, 250 MHz) δ 0.92 (dd, 6H,
J1=7Hz, J2=10Hz), 1.4 (s, 3H), 1.41 (s, 9H), 1.65-2.1 (m,
3H), 2.62 (m, 1H), 2.95 (m, 2H), 3.44 (m, 1H), 3.61 (m, 2H), 3.75 (m, 1H), 4.83 (m, 1H), 5.42 (m, 1H), 7.2 (m, 5H);
MS (FAB) m/e 439 [M+H]+
Example 7 Preparation of (2S,4S,5S)-5-(t-butyloxycarbonylamino)-6-phenγl-2,4-dihydroxy-2-benzyl-1-oxo-hexyl-valinamide (13)
To a solution of crude 4a (0.021 g, 0.043 mmol) in toluene (5 mL), excess valineamide and sodium cyanide (0.001 g) were added. The reaction was heated to reflux for 36 h, cooled and concentrated. The residue was filtered through a short column of silica gel eluting with 10%
methanol/methylene chloride followed by column chromatography
(silica gel, 5% methanol/methylene chloride) to afford the title compound (5 mg) : 1H NMR (CDCI3, 250 mhz) δ0.78 (d, J=6
Hz, 3H), 0.85 (d, J=6 Hz, 3H), 1.46 (s, 9H), 1.7-2.4 (m, 3H), 2.8-3.5 (m, 4H), 3.8-4.2 (m, 3H), 4.7 (m, 1H), 7.2-7.4 (m, 10H); MS DCI/NH3 m/e 528 {M+H]+. Example 8
Preparation of (2S,4S,5S)-5-(t-butyloxycarbonvlamino)-6-phenyl-2,4-dihydroxy-2-benzyl-2-oxo-hexyl-((2)-propane-(1.3)-d_iol)amide (14)
To a solution of 8a (0.02g, 0.041 mmol) in methylene chloride (10 mL) at 0°C, was added hydroxysuccmimide (0.005g, 0.04 mmol), DMAPEC (0.008g, 0.04 mmol), and serinόl (0.005g, 0.04 mmol). The reaction was allowed to stir at 0°C for 1 h, and allowed to warm to room temperature and stir for 16 h. The reaction mixture was diluted with methylene chloride, washed with 0.05N HCl, brine, dried over MgSO4, and concentrated to a yellow oil. Crystallization from methylene chloride/pentane affords the title compound as a white solid (10 mg, 48%): 1H NMR (CDCl3, 250 Mhz) δ 1.38 (s, 9H) , 2.24 (m, 2H), 2.6-3.1 (m,
4H), 3.6-3.9 (m, 6H), 4.7 (m, 1H) 7-7.4 (m, 10H). Example 9
Preparation of N-((2S,4S,5S)-5-(t-butγloxγoarbonyl- amino)-6-phenyl-2,4-dihydroxγ-2-methyl-1-oxo-hexy])-N'- (benzyloxycarbonyl)-hydrazine (15)
To solid 4a (0.039g, 0.08 mmol) was added 5 mL of
hydrazine hydrate and enough methanol to dissolve all
the solid (~5 mL). The reaction was stirred for 1 h at
room temperature. The reaction was diluted with water,
extracted with methylene chloride and concentrated to an oil. The residue was dissolved in THF/water (2:1, 10
mL), K2CO3 (0.022g, 0.167 mmol) and benzylchloroformate
(0.014 g, 0.08 mmol) was added. The reaction mixture
was stirred for 2 h at room temperature. The mixture
was diluted with methylene chloride, washed with water
and brine, dried and concentrated. The residue was
chromatographed (1:1 hexane:ethyl acetate) to afford the title compound (0.01 gm, 22%): 1H NMR (CDCI3, 250 MHz) δ
1.36 (s, 9H) 1.85 (m, 1H), 2.1 (m, 1H), 2.6-3.2 (m, 5H),
3.67 (m, 1H), 3.97 (m, 1H), 5.18 (m, 2H), 7-7.5 (m,
15H); MS DCI/NH3 m/e 577 M+ Example 10
Preparation of N-((2S,4S,5S)-5-(t-butyloxycarbonylamino)- 6-phenyl-2,4-dihydroxy-2-benzyl-1-oxo-hexyl)-hydrazine (16)
To solid 4a (0.048 g, 0.1 mmol) was added 5 mL of hydrazine hydrate and enough methanol to dissolve the solid (~5 mL). The reaction mixture was stirred for 1 h at room temperature. The reaction was diluted with
water, extracted with methylene chloride and
concentrated to an oil. The residue was dissolved in
0.05N HCl and extracted with methylene chloride. The aqueous phase was brought to basic pH with 2N sodium hydroxide, extracted with methylene chloride, dried with K2CO3, and concentrated to an oily solid. The solid was dissolved in methylene chloride and pentane was added to precipitate the title compound as off white solid (0.018 mg, 43%): 1H NMR (CDCI3, 250 MHz) δ1.36 (s, 9H), 1.67
(m, 2H) 2.7-3.1 (m, 4H), 3.46 (m, 1H), 3.7-3.9 (m, 2H), 4.79 (m, 1H), 7.1-7.4 (m, 10H)
Example 11
Preparation of (2S,4S,5R)-5-(benzoylformyl)amino-6-phenyl-2,4-dihydroxy-2-benzyl-1-oxo-hexyl-(benzyl)amide (17)
To a solution of 5 (0.023 g, 0.045 mmol) in
methylene chloride (2mL) was added TFA (2mL) and the
reaction mixture was stirred for 15 min. The mixture was concentrated in vacuo and the residue dissolved in methylene chloride. To this solution was added
hydroxysuccinimide (1 mg, 0.01 mmol), DMAPEC (9 mg, 0.05 mmol), and benzoylformic acid (5 mg, 0.04 mmol). The reaction mixture was stirred overnight at room
temperature, diluted with methylene chloride, washed with NaHCO3, HCl and brine. The organic phase was dried over MgSO4 and concentrated to an oil. The oil was
purified by preparative thin layer chromatography (silica gel, 1:1 hexane:ethyl acetate) to yield the
title compound as a white solid, which was
recrystallized from methylene chloride/pentane: 1H NMR
(CDCI3, 250 Mhz) δ1.8 (m, 1H), 2.24 (m, 1H), 2.8-3.3 (m, 4H), 4.0 (m, 1H), 4.2-4.4 (m, 2H), 6.9-7.3 (m, 17H), 7.4 (m, 1H), 7.53 (m, 1H), 8.18 (m, 1H)
Example 12 Liposomal Dosage Unit Composition
Phosphatidylcholine (1.4 g) and phosphatidylglycerol (0.6g) are dissolved in 300 ml of a 20% methanol in
chloroform solvent and evaporated to dryness. A solution of the peptide (30 mg in 200 ml of phosphate buffered saline) is added to the dry phospholipid film which is allowed to equilibrate at room temperature for 1-2 hr. The liposome dispersion formed is then vortexed to insure uniform mixing. The resulting suspension is extruded through a 0.2μ
polycarbonate filter five times to produce a uniform size distribution. If necessary the suspension can be dialysed or ultracentrifuged to remove non-encapsulated peptide.
Example 13 Parenteral Dosage Unit Composition
A preparation which contains 250 mg of the compound of Example 5 is prepared as follows:
250 mg of the peptide is dissolved in 150 mL of
distilled water. The solution is filtered under sterile conditions in to a multi-dose ampoule and lyophilized. The powder is reconstituted by addition of 200 mL of 5% dextrose in water (D5W) for intravenous injection. The dosage is thereby determined by the injection volume. This solution is also suitable for use in other methods for administration, such as in a bottle or bag for IV drip infusion. Example 15
Oral Dosage Unit Composition
A capsule for oral administration is prepared by mixing and milling 350 mg of the compound of Example 5 with 750 mg of lactose and 50 mg of magnesium stearate. The resulting powder is screened and filled into a hard gelatin capsule.
The above description fully discloses how to make and use this invention. This invention, however, is not limited to the precise embodiments described herein, but encompasses all modifications within the scope of the claims which follow.

Claims

What is claimed is:
1. A compound of the formula:
Figure imgf000038_0001
wherein
R1 is A, A-B or A-B-D;
A is R11, R11CO, R11-(CHR11')n-CO, R11OCO, R11OCH(R11')CO, R11NHCH(R11')CO, R11SCH(R11')CO, R11SO2, R11SO or R11C(O)CO;
B and D are
Figure imgf000038_0002
.
R2 and R4 are C1-6alkyl, optionally substituted with 1-5 fluorine atoms, C3-6alkenyl, C1-6alkyl-O-CH2 or (CH2)n _T, wherein T is phenyl, naphthyl, Cs-gcycloalkyl or indolyl, optionally substituted with nitro, halogen, Cι_4alkyl,
C1-4alkoxy, C1-4alkylthio, provided that R2 is not C1-6alkyl when R4 is cyclohexylmethyl;
R5 is H, C1-6alkyl, C1-6alkyl-O-CH2 or J-CH2(CH2)n, wherein J is CONHR1, CO2R', NHR', SR', or phenyl, naphthyl, Cs-gcycloalkyl or indolyl, optionally substituted with nitro, halogen, hydroxy, C1-4alkyl, C1-4alkoxy, C1-4alkylthio or trifluoromethyl;
R3 is G, X-G or X-Y-G;
X and Y are
Figure imgf000038_0003
;
G is R10, NR'R10, NR 'NR ' -A, OR10 or SR10;
R10 is (CR11R11 ' ) n-W ;
R11 and R11 ' are H, Alk, Ar, Het, Ar-Ci-salkyl,
Het-Ci-salkyl, C3-7cycloalkyl, Ar-C3-7cycloalkyl;
W is H, CH2OR", COR" , OR11, OCOR11, NR'R11, NR' COR11, SR11, Ar or Het;
Alk is C1-6alkyl or C1-6alkyl substituted by one or two hydroxy, nitro or C1-4alkoxy groups, or one to five fluoro atoms; R' is H, C1-6alkyl or (CH2)n-Ar;
R" is H, C1-6alkyl or (CH2)n-Ph;
n is 0, 1 or 2; and
q is 0 or 1;
or a pharmaceutically acceptable salt thereof.
2. A compound according to claim 1 in which R1 is A.
3. A compound according to claim 1 in which R2 is Ci-βalkyl or (CH2)n-phenyl.
4. A compound according to claim 1 in which R3 is
NHCH(R11)-W, wherein R11 is H or Alk and W is CH2OH, CONH2 or phenyl.
5. A compound according to claim 2 in which R3 is
NHCH(i-pr)CH2OH.
6 A compound according to claim 4 in which R2 and R4 are benzyl.
7. A compound according to claim 2 which is:
(2S,4S,5S)-5-(t-butyloxycarbonylamino)-6-phenyl-2,4-dihydroxy-2-benzyl-1-oxo-hexyl-(benzyl)amide,
(2R,4R,5R)-5-(t-butyloxycarbonylamino)-6-phenyl-2,4-dihydroxy-2-benzyl-1-oxo-hexyl-(benzyl)amide,
(2S,4S,5S)-5-(benzyloxycarbonyl-D-alanyl)amino-6-phenyl-2,4-dihydroxy-2-benzyl-1-oxo-hexyl-(benzyl)amide,
(2R,4R,5R)-5-(benzyloxycarbonyl-D-alanyl)amino-6-phenyl-2,4-dihydroxy-2-benzyl-1-oxo-hexyl-(benzyl)amide,
(2S,4S,5S)-5-(benzyloxycarbonyl-L-alanyl)amino-6-pheny1-2,4-dihydroxy-2-benzyl-1-oxo-hexyl-(benzyl)amide,
(2R,4R,5R)-5-(benzyloxycarbonyl-L-alanyl)amino-6-phenyl-2,4-dihydroxy-2-benzyl-1-oxo-hexyl-(benzyl)amide,
(2S,4S,5S)-5-(t-butyloxycarbonylamino)-6-phenyl-2,4-dihydroxy-2-benzyl-1-oxo-hexyl-((2S)-2-(3-methyl-butanol))amide, (2R,4R,5R)-5-(t-butyloxycarbonylamino)-6-phenyl-2,4- dihydroxy-2-benzyl-1-oxo-hexyl-((2S)-2-(3-methyl- butanol))amide,
(2S,4S,5S)-5-(t-butyloxycarbonylamino)-6-phenyl-2,4- dihydroxy-2-methyl-1-oxo-hexyl-((2S)-2-(3-methyl- butanol))amide,
(2R,4R,5R)-5-(t-butyloxycarbonylamino)-6-phenyl-2,4- dihydroxy-2-methyl-1-oxo-hexyl-((2S)-2-(3-methyl- butanol))amide,
(2S,4S,5S)-5-(t-butyloxycarbonylamino)-6-phenyl-2,4- dihydroxy-2-benzyl-1-oxo-hexyl-valinamide,
(2S,4S,5S)-5-(t-butyloxycarbonylamino)-6-phenyl-2,4- dihydroxy-2-benzyl-1-oxo-hexyl-(2-(propan-1,3-diol))amide, N-((2S,4S,5S)-5-(t-butyloxycarbonylamino)-6-phenyl-2,4- dihydroxy-2-benzyl-1-oxo-hexyl)-N'-(benzyloxycarbonyl)- hydrazine,
N-((2S,4S,5S)-5-(t-butyloxycarbonylamino)-6-phenyl-2,4- dihydroxy-2-benzyl-1-oxo-hexyl)-hydrazine, or
(2S,4S,5S)-5-(benzoylformyl)amino-6-phenyl-2,4-dihydroxy-2- benzyl-1-oxo-hexyl-(benzyl) amide.
8. A pharmaceutical composition which comprises a compound according to claim 1 and a pharmaceutically acceptable carrier.
9. A pharmaceutical composition which comprises a compound according to claim 7 and a pharmaceutically acceptable carrier.
10. A method for treating retroviral infection which comprises administering an effective amount of a compound according to claim 1.
11. A method according to claim 10 for treating infection by HIV-1 which comprises administering an effective amount of a compound according to claim 1.
12. A method according to claim 10 for treating AIDS which comprises administering an effective amount of a compound according to claim 1.
13. The use of a compound according to claim 1 in the manufacture of a medicament.
14. A process for preparing a compound of the formula (I):
Figure imgf000041_0001
wherein R1, R2, R3 and R4 are as defined in claim 1, which comprises:
1) (i) reacting a compound of the formula:
Figure imgf000041_0002
wherein Pr3 is H or a hydroxy-protecting group, and A,
B, D, R4 and R2 are as defined in claim 1, with any reactive groups protected,
with a coupling agent and a compound of the formula:
H-X-Y-G
wherein G, X and Y are as defined in claim 1, with any reactive groups protected, and
(ii) if necessary, removing any protecting groups; or
2) (i) reacting a compound of the formula:
H-X-Y-G
wherein G, X and Y are as defined in claim 1, with any reactive groups protected,
with a base and a compound of the formula:
Figure imgf000042_0001
wherein Pr5 is H or a hydroxy-protecting group, and A, B, D, R4 and R2 are as defined in claim 1, with any reactive groups protected, and
(ii) if necessary, removing any protecting groups; or
3) (i) reacting a compound of the formula:
Figure imgf000042_0002
wherein Pr3 is H or a hydroxy-protecting group, and X, Y, G, R2 and R4 are as defined in claim 1, with any reactive groups protected,
with a coupling agent and a compound of the formula:
A-B-D-OH
wherein G, X, Y, A and R10 are as defined for formula (I), with any reactive groups protected, and
(ii) if necessary, removing any protecting groups.
PCT/US1992/001738 1991-03-08 1992-03-07 Hiv protease inhibitors WO1992015319A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP4508263A JPH06505495A (en) 1991-03-08 1992-03-07 HIV protease inhibitor

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US66629291A 1991-03-08 1991-03-08
US666,292 1991-03-08

Publications (1)

Publication Number Publication Date
WO1992015319A1 true WO1992015319A1 (en) 1992-09-17

Family

ID=24673620

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1992/001738 WO1992015319A1 (en) 1991-03-08 1992-03-07 Hiv protease inhibitors

Country Status (3)

Country Link
EP (1) EP0575500A4 (en)
JP (1) JPH06505495A (en)
WO (1) WO1992015319A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995007269A1 (en) * 1993-09-10 1995-03-16 Narhex Limited Polar-substituted hydrocarbons
WO1995007696A1 (en) * 1993-09-13 1995-03-23 Abbott Laboratories Pharmaceutical composition of hiv-protease inhibitors
AU700942B2 (en) * 1993-09-13 1999-01-14 Abbvie Inc. Pharmaceutical compositions containing HIV protease inhibitors
US7807677B2 (en) * 2005-02-10 2010-10-05 Medivir Ab HIV protease inhibitors

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1987004349A1 (en) * 1986-01-16 1987-07-30 Joseph Dellaria Peptide analogs
EP0337714A2 (en) * 1988-04-12 1989-10-18 Merck & Co. Inc. HIV protease inhibitors useful for the treatment of aids
EP0352000A2 (en) * 1988-07-08 1990-01-24 Smithkline Beecham Corporation Retroviral protease binding peptides
EP0357332A2 (en) * 1988-08-24 1990-03-07 Merck & Co. Inc. Renin inhibitors useful for the treatment of aids by inhibition of HIV protease
US4994477A (en) * 1988-03-24 1991-02-19 Abbott Laboratories Heterocyclic renin inhibitors

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1987004349A1 (en) * 1986-01-16 1987-07-30 Joseph Dellaria Peptide analogs
US4994477A (en) * 1988-03-24 1991-02-19 Abbott Laboratories Heterocyclic renin inhibitors
EP0337714A2 (en) * 1988-04-12 1989-10-18 Merck & Co. Inc. HIV protease inhibitors useful for the treatment of aids
EP0352000A2 (en) * 1988-07-08 1990-01-24 Smithkline Beecham Corporation Retroviral protease binding peptides
EP0357332A2 (en) * 1988-08-24 1990-03-07 Merck & Co. Inc. Renin inhibitors useful for the treatment of aids by inhibition of HIV protease

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Journal of Medicinal Chemistry, Vol. 30, No. 11, issued 1987, KEMPF et al., "Renin Inhibitors Based on Novel Dipeptide Analogues - Incorporation of the Dehydrohydroxyethylene Isostere the Scissile Bond", pages 1978-1983, see entire document. *
See also references of EP0575500A4 *
Tetrahedron Letters, Vol. 29, No. 32, issued 1988, METTERNICH et al., "New Diastereoselective Synthesis of Novel Chiral Gamma-(aminoalkyl)-alpha-hydroxy-gamma-Lact ones and their Application for the Synthesis of Renin Inhibitors", pages 3923-3926. *

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995007269A1 (en) * 1993-09-10 1995-03-16 Narhex Limited Polar-substituted hydrocarbons
AP597A (en) * 1993-09-10 1997-07-15 Narhex Ltd Retroviral protease inhibitors.
WO1995007696A1 (en) * 1993-09-13 1995-03-23 Abbott Laboratories Pharmaceutical composition of hiv-protease inhibitors
US5725878A (en) * 1993-09-13 1998-03-10 Abbott Laboratories Pharmaceutical composition comprising HIV protease inhibiting compounds
AU700942B2 (en) * 1993-09-13 1999-01-14 Abbvie Inc. Pharmaceutical compositions containing HIV protease inhibitors
US5876749A (en) * 1993-09-13 1999-03-02 Abbott Laboratories Pharmaceutical composition
US5948436A (en) * 1993-09-13 1999-09-07 Abbott Laboratories Pharmaceutical composition
US7807677B2 (en) * 2005-02-10 2010-10-05 Medivir Ab HIV protease inhibitors

Also Published As

Publication number Publication date
EP0575500A4 (en) 1994-06-01
JPH06505495A (en) 1994-06-23
EP0575500A1 (en) 1993-12-29

Similar Documents

Publication Publication Date Title
JP2725690B2 (en) Carboxyalkyl peptide derivatives
KR910007884B1 (en) Process for their preparation 2 - azabicyclo (3,1,0) hexane - 3 - carboxylic acid derivatives
US5438118A (en) HIV protease inhibitors
JPH0381256A (en) Renin blocking agent
CZ280292A3 (en) Novel derivatives of 5-amino-4-hydroxyhexanoic acid which derivatives act as therapeutic agents
EP0538366A1 (en) Retroviral protease inhibitors
HU219915B (en) Process for producing pharmaceutically active hydrazine derivatives and pharmaceutical compositions containing them
JPH05230005A (en) Immunodeficiency virus protease inhibitor
NZ260675A (en) Phosphinyloxymethyl ketones containing 1-3 amino acids as enzyme inhibitors; medicaments
JPS6323897A (en) Taftsin analog, manufacture and medicinal composition
KR101264934B1 (en) - 1----- amidomethyl-substituted 1-carboxyalkyl-cyclopentylcarbonylamino-benzazepine-n-acetic acid derivatives process and intermediate products for their preparation and medicaments containing these compounds
CA2092414A1 (en) N-(2-alkyl-3-mercaptoglutaryl)-amino-diaza cycloalkanone derivatives and their use as collagenase inhibitors
US5643878A (en) 5-amino-4-hydroxyhexanoic acid derivatives
EP0374098A2 (en) Inhibitors of retroviral proteases
EP0538396A1 (en) Inhibitors of retroviral proteases
WO1992015319A1 (en) Hiv protease inhibitors
EP0594586A1 (en) Hiv protease inhibitors
JPH069628A (en) Hiv protease inhibitor containing hydroxyethylene equivalentweight type ditholanylglycine
JPH05508846A (en) Aspartic protease inhibitor
JPH04275263A (en) Stereoregulated preparation of hydroxy ester, hydroxy amide and lactone compound from chiral alpha-aminoaldehyde
SK30394A3 (en) Derivatives of 2,4-diamino-3-hydroxycarboxylic acid, method of their preparation, pharmaceutical compositions containing these derivatives and their use
EP0538383A1 (en) Inhibitors of aspartic proteases
EP0628035A1 (en) Retroviral protease inhibitors
CZ54194A3 (en) Derivatives of acylhexanoic acid, process of their preparation, pharmaceutical preparations in which they are comprised and their pharmaceutical use
JPH09512012A (en) Synthesis of N-Boc-L-propargylglycine by zinc-mediated addition of organic halides to glycine cation equivalents

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): JP US

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IT LU MC NL SE

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 1992908310

Country of ref document: EP

ENP Entry into the national phase

Ref country code: US

Ref document number: 1993 117007

Date of ref document: 19930908

Kind code of ref document: A

Format of ref document f/p: F

WWP Wipo information: published in national office

Ref document number: 1992908310

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1992908310

Country of ref document: EP