AU712120B2 - HIV protease inhibitors useful for the treatment of AIDS - Google Patents

HIV protease inhibitors useful for the treatment of AIDS Download PDF

Info

Publication number
AU712120B2
AU712120B2 AU27473/97A AU2747397A AU712120B2 AU 712120 B2 AU712120 B2 AU 712120B2 AU 27473/97 A AU27473/97 A AU 27473/97A AU 2747397 A AU2747397 A AU 2747397A AU 712120 B2 AU712120 B2 AU 712120B2
Authority
AU
Australia
Prior art keywords
compound
hiv
combination
substituted
aids
Prior art date
Legal status (The legal status 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 status listed.)
Ceased
Application number
AU27473/97A
Other versions
AU2747397A (en
Inventor
Craig A Coburn
Mark E. Fraley
M Katharine Holloway
Randall W. Hungate
Kristine Prendergast
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Merck and Co Inc
Original Assignee
Merck and Co Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from GBGB9613487.9A external-priority patent/GB9613487D0/en
Application filed by Merck and Co Inc filed Critical Merck and Co Inc
Publication of AU2747397A publication Critical patent/AU2747397A/en
Application granted granted Critical
Publication of AU712120B2 publication Critical patent/AU712120B2/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D221/00Heterocyclic compounds containing six-membered rings having one nitrogen atom as the only ring hetero atom, not provided for by groups C07D211/00 - C07D219/00
    • C07D221/02Heterocyclic compounds containing six-membered rings having one nitrogen atom as the only ring hetero atom, not provided for by groups C07D211/00 - C07D219/00 condensed with carbocyclic rings or ring systems
    • C07D221/22Bridged ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/10Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/06Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms

Landscapes

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

Description

WO 97/40833 PCT/US97/07131 -1- TITLE OF THE INVENTION HIV PROTEASE INHIBITORS USEFUL FOR THE TREATMENT OF AIDS This application is related to Merck Case 19531PV.
The present invention is concerned with compounds which inhibit the protease encoded by human immunodeficiency virus (HIV) or pharmaceutically acceptable salts thereof and are of value in the prevention of infection by HIV, the treatment of infection by HIV and the treatment of the resulting acquired immune deficiency syndrome (AIDS). It also relates to pharmaceutical compositions containing the compounds and to a method of use of the present compounds and other agents for the treatment of AIDS and viral infection by HIV.
BACKGROUND OF THE INVENTION A retrovirus designated human immunodeficiency virus (HIV) is the etiological agent of the complex disease that includes progressive destruction of the immune system (acquired immune deficiency syndrome; AIDS) and degeneration of the central and peripheral nervous system. This virus was previously known as LAV, HTLV-III, or ARV. A common feature of retrovirus replication is the extensive post-translational processing of precursor polyproteins by a virally encoded protease to generate mature viral proteins required for virus assembly and function. Inhibition of this processing prevents the production of normally infectious virus. For example, Kohl, N.E. et al., Proc. Nat'l Acad. Sci., 85, 4686 (1988) demonstrated that genetic inactivation of the HIV encoded protease resulted in the production of immature, non-infectious virus particles. These results indicate that inhibition of the HIV protease represents a viable method for the treatment of AIDS and the prevention or treatment of infection by HIV.
The nucleotide sequence of HIV shows the presence of a pol gene in one open reading frame [Ratner, L. et al., Nature, 313, 277(1985)]. Amino acid sequence homology provides evidence that the pol sequence encodes reverse transcriptase, an endonuclease and an HIV protease [Tho, H. et al., EMBO 4, 1267 (1985); Power, M.D. et al., Science, 231, 1567 (1986); Pearl, L.H. et al., Nature, 329, 351 (1987)]. Applicants demonstrate that the compounds of this invention are inhibitors of HIV protease.
Brief Description of the Invention Compounds of Formula I, as herein defined, are disclosed. These compounds are useful in the inhibition of HIV protease, the prevention of infection by HIV, the treatment of infection by HIV and in the treatment of AIDS, either as compounds, pharmaceutically acceptable salts, pharmaceutical composition ingredients, whether or not in combination with other antivirals, immunomodulators, antibiotics or vaccines. Methods of treating AIDS, methods of preventing infection by HIV, and methods of treating infection by HIV are also disclosed.
Detailed Description of the Invention and Preferred Embodiments This invention is concerned with compounds of Formula I, combinations thereof, or pharmaceutically acceptable salts thereof, in the inhibition of HIV protease, the S 15 prevention or treatment of infection by HIV and in the treatment of the resulting acquired immune deficiency syndrome (AIDS). Compounds of Formula I are defined as follows:
Z
R2 Ri /R3
Y
wherein X is -NR 4 or Y is O, or forms, with the carbon to which it is attached, [n:\ibc]00082:tab j WO 97/40833 WO 9740833PCTIUS97/07131 -3-
,OH
/C\
Z is or forms, with the carbon to which it is attached,
\OH
RI is a) H; b) Ci1-4 alkyl; c) C3-7 cycloalkyl; d) aryl, unsubstituted or substituted one or more times with hydroxy; e) CH2R 5 or f) 5-7 membered heterocycle; and 'I WO 97/40833 WO 9740833PCT/US97/07131 -4-
R
2 is a) C 1-4 alkyl; b) aryl, unsubstituted or substituted with aryl; c) CH2R 6 or d) hetero cycle; and
R
3 is a) CH(OH)R 7 or b) CH(NI-2)R 7 and
R
4 is a) C 1-4 alkyl; b) C3-6 cycloalkyl; c) aryl unsubstituted or substituted with halo or with Ci1 -4 alkyl unsubstituted or substituted one or more times with hydroxy; d) CH2RI; or e) 5-7 membered heterocycle; and
R
5 is a) C 1-4 alkyl; or b) aryl; and
R
6 is a) C 1-4 alkyl; b) aryl unsubstituted or substituted with halo or with Ci1 -4 alkyl unsubstituted or substituted one or more times with hydroxy; or c) 5-7 membered heterocycle; and
R
7 is a) H; b) C 1-4 alkyl; c) aryl unsubstituted or substituted with amino; 11WO 97/40833 PCT/US97/07131 d) C1-3 alkylaryl unsubstituted or substituted with amino; or e) 5-7 membered heterocycle; or pharmaceutically acceptable salt thereof.
One preferred embodiment is a compound of the formula 0 R
NR
4
HO
HO R7 wherein
R
2 is C1-4 alkylene-aryl; and
R
4 is C1-4 alkyl, unsubstituted or substituted with aryl, C3-6 cycloalkyl, or 5-7 membered heterocycle;
R
7 is H, benzyl unsubstituted or substituted with amino; or pharmaceutically acceptable salt thereof.
Preferred compounds of this invention are shown below.
Compound A: I WO 97/40833 WO 9740833PCTIUS97/07131 -6or pharmaceutically acceptable salts thereof; and )WO 97/40833 PCT/US97/07131 -7- Compound B:
HO
H 0c
HO
or pharmaceutically acceptable salts thereof.
The compounds of the present invention, may have asymmetric centers and occur as racemates, racemic mixtures and as individual diastereomers, or enantiomers with all isomeric forms being included in the present invention.
When any variable aryl, heterocycle, R 1
R
2 X, Y, or Z, etc.) occurs more than one time in any constituent or in Formula I, its definition on each occurrence is independent of its definition at every other occurrence. Also, combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
As used herein except where noted, "alkyl" is intended to include both branched- and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms (Me is methyl, Et is ethyl, Pr is propyl, Bu is butyl).
As used herein, with exceptions as noted, "aryl" is intended to mean phenyl (Ph) or naphthyl.
The term heterocycle or heterocyclic, as used herein except where noted, represents a stable 5- to 7-membered mono- or bicyclic or stable 7- to 10-membered bicyclic heterocyclic ring system, any ring of which may be saturated or unsaturated, and which consists of carbon atoms and from one to three heteroatoms selected from the group WO 97/40833 PCT/US97/07131 -8consisting of N, O and S, and wherein the nitrogen and sulfur heteroatoms may optionally be oxidized, and the nitrogen heteroatom may optionally be quaternized, and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring. The heterocyclic ring may be attached at any heteroatom or carbon atom which results in the creation of a stable structure.
Examples of such heterocyclic elements include piperidinyl, piperazinyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolodinyl, 2 -oxoazepinyl, azepinyl, pyrrolyl, 4-piperidonyl, pyrrolidinyl, pyrazolyl, pyrazolidinyl, imidazolyl, imidazolinyl, imidazolidinyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, oxazolyl, oxazolidinyl, isoxazolyl, isoxazolidinyl, morpholinyl, thiazolyl, thiazolidinyl, isothiazolyl, quinuclidinyl, isothiazolidinyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, thiadiazoyl, benzopyranyl, benzothiazolyl, benzoxazolyl, furyl, tetrahydrofuryl, tetrahydropyranyl, thienyl, benzothienyl, thiamorpholinyl, thiamorpholinyl sulfoxide, thiamorpholinyl sulfone, and oxadiazolyl.
The pharmaceutically-acceptable salts of the compounds of Formula I (in the form of water- or oil-soluble or dispersible products) include the conventional non-toxic salts or the quatemary ammonium salts which are formed, from inorganic or organic acids or bases.
Examples of such acid addition salts include acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, lactate, maleate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, oxalate, pamoate, pectinate, persulfate, 3-phenylpropionate, picrate, pivalate, propionate, succinate, tartrate, thiocyanate, tosylate, and undecanoate. Base salts include ammonium salts, alkali metal salts such as sodium and potassium salts, alkaline earth metal salts such as calcium and magnesium salts, salts with organic bases such as dicyclohexylamine salts, N-methyl-Dglucamine, and salts with amino acids such as arginine, lysine, and so WO 97/40833 PCT/US97/07131 -9forth. Also, the basic nitrogen-containing groups may be quatemized with such agents as lower alkyl halides, such as methyl, ethyl, propyl, and butyl chloride, bromides and iodides; dialkyl sulfates like dimethyl, diethyl, dibutyl; and diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl bromides and others. Other pharmaceutically acceptable salts include the sulfate salt ethanolate and sulfate salts.
Schemes I and II for preparing the novel compounds of this invention are presented below. Tables I and II which follow the schemes illustrate the compounds that can be synthesized by Schemes I and II, but Schemes I and II are not limited by the compounds in the tables nor by any particular substituents employed in the schemes for illustrative purposes. The examples specifically illustrate the application of the following schemes to specific compounds.
Additional related information on synthetic background is contained in EPO 0337714.
One method for producing Formula I compounds is provided by Scheme I.
0 0 'OMe 0/
O
K
2 00 3 /R2X' acetone/reflux MeONa[THF 0 0 00/
OCH
3
IV
CH
2 0/MeOH
R
4
NH
2 /reflux 1) LiAIH 4
/THF
2) [1 3 O+/acetone Alkylation of ester I by reaction with R 2 X' (wherein X' is halo) in base gives II.
Reaction with MeONa rearranges 11 to afford III> Cyclisation of R 4
NH
2 gives the azabicyclic 1) nonane core precursor IV which, after reduction and acid hydrolysis, provides V. Scheme I is illustrated as one embodiment in Example I.
LLj
(NT)
[n:\libc]00082: tab WO 97/40833 PCT/US97/07131 -11- SCHEME II 1) S0 3 :pyr
DMSO/TEA
2) Et 3 SiCl/pyr 0
C
1) PhCH 2 MgCI/THF 2) H30 Et 3 Si--, Scheme II outlines another general synthetic method.
Alcohol oxidation by treatment of P with S03*pyridine complex in DMSO, followed by silylation, gives Q. Alkylation with the appropriate Grignard reagent, followed thereafter with acid treatment, affords R. Scheme II is also illustrated in one embodiment in Example 3.
WO 97/40833 PCTIUS97/07131 12 The compounds of this invention are also illustrated by Tables 1-11, which follow.
TABLE 1 Table 11 0e 0* 0 0 WO 97/40833 PCT/US97/07131 14- TABLE II (Cont'd) 22 \N
CH
2 "J 84 100 10.4 23 0. 0 69 100 29 The compounds of this invention are useful in the preparation and execution of screening assays for antiviral compounds.
For example, the compounds of this invention are useful for isolating enzyme mutants, which are excellent screening tools for more powerful antiviral compounds. Furthermore, the compounds of this invention are useful in establishing or determining the binding site of other antivirals to HIV protease, by competitive inhibition. Thus the compounds of this invention are commercial products to be sold for these purposes.
The compounds of the present invention are useful in the inhibition of HIV protease the prevention or treatment of infection by the human immunodeficiency virus (HIV) and the treatment of, and delaying of the onset of consequent pathological conditions such as AIDS. Treating AIDS or preventing or treating infection by. HV is defined as including, but not limited to, treating a wide range of states of HIV infection: AIDS, ARC (AIDS related complex), both symptomatic and asymptomatic, and actual or potential exposure to HIV. For example, the compounds of this invention are useful in treating infection by HIV after suspected past exposure to HIV by, e.g., blood transfusion, organ transplant, exchange of body fluids, bites, accidental needle stick, or exposure to patient blood during surgery.
For these purposes, the compounds of the present invention may be administered orally, parenterally (including subcutaneous injections, intravenous, intramuscular, intrasternal injection or infusion techniques), by inhalation spray, or rectally, in dosage unit formulations containing conventional non-toxic pharmaceutically-acceptable carriers, adjuvants and vehicles.
WO 97/40833 PCT/US97/07131 Thus, in accordance with the present invention there is further provided a method of treating and a pharmaceutical composition for treating HIV infection and AIDS. The treatment involves administering to a patient in need of such treatment a pharmaceutical composition comprising a pharmaceutical carrier and a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt thereof.
These pharmaceutical compositions may be in the form of orally-administrable suspensions or tablets; nasal sprays; sterile injectable preparations, for example, as sterile injectable aqueous or oleagenous suspensions or suppositories.
When administered orally as a suspension, these compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may contain microcrystalline cellulose for imparting bulk, alginic acid or sodium alginate as a suspending agent, methylcellulose as a viscosity enhancer, and sweetners/flavoring agents known in the art. As immediate release tablets, these compositions may contain microcrystalline cellulose, dicalcium phosphate, starch, magnesium stearate and lactose and/or other excipients, binders, extenders, disintegrants, diluents and lubricants known in the art.
When administered by nasal aerosol or inhalation, these compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizing or dispersing agents known in the art.
The injectable solutions or suspensions may be formulated according to known art, using suitable non-toxic, parenterallyacceptable diluents or solvents, such as mannitol, 1,3-butanediol, water, Ringer's solution or isotonic sodium chloride solution, or suitable dispersing or wetting and suspending agents, such as sterile, bland, fixed oils, including synthetic mono- or diglycerides, and fatty acids, including oleic acid.
M
WO 97/40833 PCT/US97/07131 -16- When rectally administered in the form of suppositories, these compositions may be prepared by mixing the drug with a suitable non-irritating excipient, such as cocoa butter, synthetic glyceride esters or polyethylene glycols, which are solid at ordinary temperatures, but liquidify and/or dissolve in the rectal cavity to release the drug.
Dosage levels of the order of 0.02 to 5.0 or 10.0 gramsper-day are useful in the treatment or prevention of the above-indicated conditions, with oral doses two-to-five times higher. For example, infection by HIV is effectively treated by the administration of from to 50 milligrams of the compound per kilogram of body weight from one to four times per day. In one preferred regimen, dosages of 100- 400 mg every six hours are administered orally to each patient. It will be understood, however, that the specific dose level and frequency of dosage for any particular patient may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the host undergoing therapy.
The present invention is also directed to combinations of the HIV protease inhibitory compounds with one or more agents useful in the treatment of AIDS. For example, the compounds of this invention may be effectively administered, whether at periods of preexposure and/or post-exposure, in combination with effective amounts of the AIDS antivirals, immunomodulators, anti-infectives, or vaccines known to those of ordinary skill in the art.
WO 97/40833 PCTIUS97/07131 17- TABLE C
ANTIVIRALS
Drug Name AL-721 Recombinant Human Interferon Beta Acemannan Manufacturer Ethigen (Los Angeles, CA) Triton Biosciences (Almeda, CA) Carrington Labs (Irving, TX) Cytovene Syntex Indication ARC, PGL HIV positive, AIDS AIDS, Kaposi's sarcoma, ARC
ARC
(See also immunomodulators) sight threatening
CMV
Indication peripheral CMV retinitis AIDS, ARC AIDS, ARC Drug Name Ganciclovir d4T Didehydrodeoxythymidine ddl Dideoxyinosine Manufacturer (Palo Alto, CA) Bristol-Myers (New York, NY) Bristol-Myers (New York, NY) Elan Corp, PLC (Gainesville, GA) HIV infection (See also immunomodulators) WO 97/40833 PCT/US97/07131 -18- Drug Name Trisodium Phosphonoformate Dideoxycytidine; ddC Novapren Manufacturer Astra Pharm.
Products, Inc (Westborough,
MA)
Hoffman-La Roche (Nutley, NJ) Novaferon Labs, Inc.
(Akron, OH) Diapren, Inc.
(Roseville,
MN,
marketer) Indication CMV retinitis, HIV infection, other CMV infections AIDS, ARC HIV inhibitor Peptide T Octapeptide Sequence Peninsula Labs (Belmont, CA)
AIDS
Zidovudine; AZT Burroughs Wellcome (Rsch. Triangle Park,
NC)
AIDS, adv, ARC pediatric AIDS, Kaposi's sarcoma, asymptomatic HIV infection, less severe HIV disease, neurological involvement, in combination with other therapies.
Ansamycin LM 427 Adria Laboratories (Dublin, OH) Erbamont (Stamford, CT)
ARC
WO 97/40833 PCT/US97/07131 19- Drug Name Dextran Sulfate Virazole Ribavirin Alpha Interferon Acyclovir Antibody which neutralizes pH labile alpha aberrant Interferon in an immuno-adsorption column L-743,726 Manufacturer Ueno Fine Chem.
Ind. Ltd.
(Osaka, Japan) Viratek/ICN (Costa Mesa, CA) Burroughs Wellcome (Rsch. Triangle Park, NC) Burroughs Wellcome Advanced Biotherapy Concepts (Rockville, MD) Merck (Rahway, NJ) Indication AIDS, ARC, HIV positive asymptomatic asymptomatic HIV positive, LAS, ARC Kaposi's sarcoma, HIV in combination w/Retrovir AIDS, ARC, asymptomatic HIV positive, in combination with
AZT.
AIDS, ARC AIDS, ARC, asymptomatic HIV positive, also in combination with
AZT.
WO 97/40833 PCT/US97/07131
IMMUNO-MODULATORS
Drug Name AS-101 Bropirimine Acemannan CL246,738 Manufacturer Wyeth-Ayerst Labs.
(Philadelphia, PA) Upjohn (Kalamazoo, MI) Carrington Labs, Inc.
(Irving, TX) American Cyanamid (Pearl River, NY) Lederle Labs (Wayne, NJ) Indication
AIDS
advanced AIDS AIDS, ARC (See also anti-virals) AIDS, Kaposi's sarcoma ELIO Elan Corp, PLC (Gainesville, GA) Gamma Interferon HIV infection (See also anti-virals) ARC, in combination w/TNF (tumor necrosis factor) Granulocyte Macrophage Colony Stimulating Factor Granulocyte Macrophage Colony Stimulating Factor Genentech San Francisco,
CA)
Genetics Institute (Cambridge,
MA)
Sandoz (East Hanover, NJ) Hoeschst-Roussel (Sommerville, NJ) Immunex (Seattle, WA)
AIDS
AIDS
WO 97/40833 PCT/US97/07131 -21 Drug Name Granulocyte Macrophage Colony Stimulating Factor Manufacturer Schering-Plough (Madison, NJ) Indication
AIDS
HIV Core Particle Immunostimulant IL-2 Interleukin-2 IL-2 Interleukin-2 Immune Globulin Intravenous (human) IMREG-1 IMREG-2 Imuthiol Diethyl Dithio Carbamate Alpha-2 Interferon Rorer (Ft. Washington, PA) Cetus (Emeryville, CA) Hoffman-La Roche (Nutley, NJ) Immunex Cutter Biological (Berkeley, CA) Imreg (New Orleans, LA) Imreg (New Orleans, LA) Merieux Institute (Miami, FL) Schering Plough (Madison, NJ) AIDS, in combination w/AZT seropositive HIV AIDS, in combination w/AZT AIDS, ARC, HIV, in combination w/AZT pediatric AIDS, in combination w/AZT AIDS, Kaposi's sarcoma, ARC, PGL AIDS, Kaposi's sarcoma, ARC, PGL AIDS, ARC Kaposi's sarcoma w/AZT: AIDS WO 97/40833 PCT/US97/07131 -22- Drug Name Methionine- Enkephalin
MTP-PE
Muramyl- Tripeptide Granulocyte Colony Stimulating Factor rCD4 Recombinant Soluble Human CD4 Manufacturer TNI Pharmaceutical (Chicago, IL) Ciba-Geigy Corp.
(Summit, NJ) Amgen (Thousand Oaks, CA) Genentech San Francisco,CA) Indication AIDS, ARC Kaposi's sarcoma AIDS, in combination w/AZT AIDS, ARC rCD4-IgG hybrids AIDS, ARC Recombinant Soluble Human CD4 Interferon Alfa 2a SK&F106528 Soluble T4 Biogen (Cambridge,
MA)
Hoffman-La Roche (Nutley, NJ) Smith, Kline French Laboratories (Philadelphia, PA) AIDS, ARC Kaposi's sarcoma AIDS, ARC, in combination w/AZT HIV infection WO 97/40833 WO 9740833PCTIUS97/07131 23 Drug Name Thymopentin Tumor Necrosis Factor; TNF Manufacturer LInmunobiology Research Institute (Annandale, NJ) Genentech San Francisco,
CA)
ANTI- INFECTIVES Indication HIV infection ARC, in combination w/gamma Interferon Drug Name Clindamycin with Primaquine Fluconazole Pastille Nystatin Pastille Manufacturer Upjohn (Kalamazoo, MI) Pfizer (New York, NY) Squibb Corp.
(Princeton, NJ) Indication
PCP
cryptococcal meningitis, candidiasis prevention of oral candidiasis Omnidyl Eflomnithine Merrell Dow (Cincinnati, OH) LyphoMed (Rosemont, IL)
PCP
Pentamidine Isethionate (IM IV) PCP treatment Trimethoprim antibacterial antibacterial Trimethoprim/sulfa .WO 97/40833 PCT/US97/07131 -24- Drug Name Piritrexim Manufacturer Indication Burroughs Wellcome PCP treatment (Rsch. Triangle Park, NC) Pentamidine isethionate for inhalation Spiramycin Intraconazole- R51211 Fisons Corporation (Bedford, MA) Rhone-Poulenc Pharmaceuticals (Princeton, NJ) Janssen Pharm.
(Piscataway, NJ) PCP prophylaxis cryptosporidial diarrhea histoplasmosis; cryptococcal meningitis Trimetrexate Warner-Lambert
PCP
OTHER
Drug Name Recombinant Human Erythropoietin Megestrol Acetate Total Enteral Nutrition Manufacturer Ortho Pharm. Corp.
(Raritan, NJ) Bristol-Myers (New York, NY) Norwich Eaton Pharmaceuticals (Norwich, NY) Indication severe anemia assoc. with AZT therapy treatment of anorexia assoc.
w/AIDS diarrhea and malabsorption related to AIDS WO 97/40833 PCT/US97/07131 It will be understood that the scope of combinations of the compounds of this invention with AIDS antivirals, immunomodulators, anti-infectives or vaccines is not limited to the list in the above Table, but includes in principle any combination with any pharmaceutical composition useful for the treatment of AIDS.
Certain compounds of Table S are the following: L-743,726 is 6-chloro-4(S)-cyclopropylethynyl-4(S)trifluoromethyl-1,4-dihydro-2H-3, -benzoxazin-2-one, and is synthesized according to EP 0 582,455.
The synthesis of ddC, ddl and AZT are also described in EPO 484071.
Preferred combinations are simultaneous or alternating treatments of an inhibitor of HIV protease and a non-nucleoside inhibitor of HIV reverse transcriptase. An optional third component in the combination is a nucleoside inhibitor of HIV reverse transcriptase, such as AZT, ddC or ddl. A preferred inhibitor of HIV protease is L-735,524 (Compound disclosed and synthesized according to U. S.
Patent No. 5,413,999. Preferred non-nucleoside inhibitors of HIV reverse transcriptase include L-743,726. These combinations may have synergistic effects on limiting the spread of HIV. Preferred combinations include the following L-735,524, with L-743,726, and, optionally, AZT or ddl or ddC; L-735,524, and any of AZT or ddl or ddC.
Assay for Inhibition of Microbial Expressed HIV Protease Inhibition studies of the reaction of the protease expressed in Eschericia coli with a peptide substrate [Val-Ser-Gln-Asn- (betanapthyl)Ala-Pro-Ile-Val, 0.5 mg/mL at the time the reaction is initiated] were in 50 mM Na acetate, pH 5.5, at 30 0 C for 1 hour.
Various concentrations of inhibitor in 1.0 ul DMSO were added to gl of the peptide solution in water. The reaction is initiated by the addition of 15 gl of 0.33 nM protease (0.11 ng) in a solution of 0.133 M Na acetate pH 5.5 and 0.1% bovine serum albumin. The reaction was -26quenched with 160 ptl of 5% phosphoric acid. Products of the reaction were separated by HPLC (VYDAC wide pore 5 cm C-18 reverse phase, acetonitrile gradient, 0.1% phosphoric acid). The extent of inhibition of the reaction was determined from the peak heights of the products.
HPLC of the products, independently synthesized, proved quantitation standards and confirmation of the product composition. Tables I and II provide results for a variety of compounds.
EXAMPLE I
HO
H
OH
5(RS)-((4')-2"-furanyl)methylpheny-9(RS)-hydroxy- (RS)-hydroxymethyl-3-(2'-methylpropyl-3-azabicyclo[3.3.1]nonan-7-one (Compound 10. Table 1) Step 1: Compound Y S. 'YNal/reflux
B
O Br OBr Br -27- To a solution of 2 -carbomethoxy-4-ethylenedioxycyclohexanone I, (3.0 g, 14.0 mmol, Fuchs, P. L. et al., Syn. Comm, 13(3), 243, 1983) in 100 mL of acetone was added 4-bromobenzyl bromide (3.67 g, 14.7 mmol), K2C03 (9.69 g, 70.1 mmol) and Nal (210 mg, 1.4 mmol). The heterogenous reaction was heated at reflux for 16 h. The reaction mixture was cooled and filtered through Celite. The filtrate was diluted with 250 mL of Et20 and the organics were washed with water (2 x 20 mL) then brine (50 mL) and dried over MgSO4.
Evaporation of the solvent and flash chromatography (SiO2; 4:1 Hexane/EtOAc) gave 4.8 g of Y.
1 H NMR (CDC13) 8 7.40 J 7.8 Hz, 2H), 7.05 J 7.8 Hz, 2H), 3.98 5H), 3.60 3H), 3.00 3H), 2.50 2H), 1.95 2H).
Step 2: Compound z 0 0 0 0
OCH
3 NaOMe/THF iOCH 3 reflux Br 0 0 0Br 0 0 Br Y
Z
To a slurry of NaH (375 mg, 15.6 mmol) in THF (15 mL) 20 at 0°C was added MeOH (0.76 mL, 30.7 mmol). After stirring for min, keto ester A (4.8 g, 12.5 mmol) in THF (30 mL) was added dropwise and the solution was warmed to room temperature and stirred for 16 h. The reaction mixture was diluted with 50 mL of EtOAc, then excess NaOMe was quenched with 10 mL of saturated NH4C1. The organic phase was separated, washed with brine and dried over MgSO4.
Evaporation of the solvent and flash chromatography (SiO2; 4; 1 Hexane/EtOAc) gave 4.5 g of Z.
i -28- 11H NMR (400 MHz, CDCI3) 8 12.6 1H), 7.40 J 7.8 Hz, 2H), 7.05 J 7.8 Hz, 2H), 3.98 4H), 3.75 3H), 3.25 1H), 2.99 1H), 2.78 1H), 2.4-1.8 4H).
Step 3: Compound C 00 B 0- Br^ 0
OCH
3 Br 0 CH 2 0/HOAc/MeOH O O NH 2
CH
2
CH(CH
3 2 0 OCH 3 z
C
To a solution of keto ester (2.9 g, 7.57 mmol) z in MeOH (45 mL) and an aqueous solution of formaldehyde 5.6 mL, 75.7 mmol) was added isobutyl amine (0.9 mL, 9.1 mmol) and HOAc (0.52 mL, 9.1 mmol). The whole was heated at reflux for 16 h. The reaction was cooled to room temperature and the solvent was removed. The residue was dissolved in EtOAc (100 mL) and the resulting solution was washed with sat'd NaHCO3 (2 x 20 mL), water (2 x 20 mL) then brine mL) and dried over MgSO4. Evaporation of the solvent and flash chromatography (SiO2 gradient; 4:1, 2:1, 1:1 Hexane/EtOAc gave 2.5 g of C. m.p. 142-144 0
C.
20 1 H NMR (400 MHz, CDC13) 8 7.40 J 7.8 Hz, 2H), 7.05 J 7.8 Hz, 2H), 3.98 4H), 3.80 3H), 3.65 1H), 3.00 (dd, J 11.0 Hz, 1H), 2.85 J 14.1 Hz, 1H), 2.70 4H), 2.56 J 13.2 Hz, 1H), 2.40 J 13.2 Hz, 1H), 2.20 3H), 1.70 1H), 0.90 m, 6H).
0* r 08 WO 97/40833 PCT/US97/7131 -29- Step 4: Compound D Br O
A
0
OCH
3 0 Br 0 0-
HO
NaBH 4 /EtOH THF 0OC H iOCH 3 0 To a solution of ketone (1.8 g, 3.75 mml) C in 18 mL of 1:1:1 EtOH, CH2C12 and H20 at 0°C was added NaBH4 (142 mg, 3.75 mmol). The solution was stirred for 30 min, then excess NaBH4 was quenched with 5 mL of acetone. The solution was diluted with EtOAc and washed with water (4 x 10 mL) then brine (10 mL). Evaporation of the solvent and column chromatography (Si02; 65:35 Hexane/EtOAc) gave 964 mg of D.
1 H NMR (400 MHz, CDC13) 5 7.40 J 7.8 Hz, 2H), 7.05 J 7.8 Hz, 2H), 4.50 J 11 Hz, 1H), 4.20-3.90 4H), 3.75 3H), 3.40 J 11.2 Hz, 1H), 2.90 J 13.5 Hz, 1H), 2.80 J 12.2 Hz, 1H), 2.60 2H), 2.40 J 10.6 Hz, 1H), 2.20 J 10.4 Hz, 1H), 2.00-1.80 5H), 1.70 1H), 0.90 m, 6H).
Step 5: Compound E LiEt 3 BH/THF 0°C WO 97/40833 PCT/US97/07131 To a solution of ester (964 mg, 2.0 mmol) of D in THF mL) at 0°C was added LiEt3BH (6.09 mL, 6.0 mmol). The solution was warmed to room temperature and stirred for 4 hours. Excess LiEt3BH was quenched with 5 mL of saturated NaHCO3. The solution was diluted with Et20 (50 mL) and washed with saturated NaHCO3 (3 x mL), water (4 x 10 mL) and brine (10 mL). Evaporation of the solvent left 800 mg of crude diol E which was used directly in the next step without purification. m.p. 136-138 0
C.
1H NMR (400 MHz, CDC13) 8 7.39 J 8.2 Hz, 2H), 7.13 J 8.2 Hz, 2H), 4.74 J 11.9 Hz, 1H), 4.05 4H), 3.57 J 10.8 Hz, 1H), 3.39 J 10.82 Hz, 1H), 3.-6 J 11.9 Hz, 1H), 2.96 J 13.4 Hz, 2H), 2.50 J 13.4 Hz, 1H), 2.34 J 10.6 Hz, 1H), 2.27 J 10.6 Hz, 1H), 2.09 J 11.7 Hz, 2H), 1.95 J 17.3 Hz, 2H), 1.85 J 14.3 Hz, 1H), 1.77 1H), 1.63 J 11.1 Hz, 3H), 0.90 J 12.4 Hz, 6H).
Step 6: Compound F (L-770.274) 0- Br 0 Br 0 IH O OH H OH HCI/acetone H E
F
To a solution of ketal (453 mg, 1.0 mmol) E in acetone (8 mL) at 0°C was added 8 mL of 50% HCI in water. The solution was heated at reflux for 16 h, then cooled to 0°C. Saturated NaHCO3 solution was added to quench excess HC1. The solution was then washed with EtOAc (3 x 10 mL) and the combined organic extracts were dried over MgSO4. Evaporation of the solvent and trituration of the resulting white solid with Et20 gave 300 mg of F. m.p. 155-156 C WO 97/40833 PCT/US97/07131 -31 1 H NMR (400 MHz, CDC13) 5 7.40 J 7.8 Hz, 2H), 7.05 J 7.8 Hz, 2H), 3.95 1H), 3.65 (dd, J 4.4, 10.4 Hz, 1H), 3.49 1H), 3.45 (dd, J 4.6, 10.4 Hz, 1H), 2.80 2H), 2.60 J 14.2 Hz, 2H), 2.52 J 3.7 Hz, 1H), 2.43 J 11.4 Hz, 1H), 2.35 J 11.1 Hz, 1H), 2.00 (m 4H), 1.83 J 11.2 Hz, 1H), 1.69 J 11.2 Hz, 1H), 1.60 1H), 0.76 6H).
Anal calc'd for C20H28NO3Br: C, 58.54; H, 6.88; N, 3.41.
Found: C, 58.91; H, 6.88; N, 3.51.
Ste 7: Compound 10. Table I Br 0_
HON
H OH PdCI 2 (PPh 3 2 /DMF HO
SOH
F 0 O Sn(n-Bu) 4 To a solution of the aryl bromide (41 mg, 0.10 mmol) in DMF (0.4 mL) was added 2 -(tri-n-butylstannyl) furan (53.5 mg, 0.15 mmol) and PdCl2(PPh3)2 (1.5 mg, 0.0020 mmol). The resulting yellow-brown solution was stirred at 95C for 4 h. The reaction mixture was cooled, diluted with ether and filtered through Celite. The filtrate was washed with water (7 x 2 mL), brine (2 mL) and dried over MgSO4. The yellow oil was subjected to flash chromatography (SiO2; 95:5:0.5 CHC13/IPA/NH40H) to afford 25 mg of the title compound as a foam.
1 H NMR (400 MHz, CDC13) 5 7.60 J 7.8 Hz, 2H), 7.45 J 0.8 Hz, 1H), 7.21 J 7.8 Hz, 2H), 6.62 J 4 .2 Hz, 1H), 6.44 (dd, J 4.2, 0.8 Hz, 1H), 3.81 1H), 3.63 3H), 3.50 2H), 2.85 WO 97/40833 PCTUS97/07131 32 (in, 2H), 2.60 (in, 4H), 2.45 J 14 Hz, 1H), 2.35 J 14 Hz, 1H), 1.8-2.1 (in, 6H), 1.7 J 14 Hz, lH), 1.6 (in, 2H), 0.77 J 8 Hz, 6H).
Anal calc'd for C24H31IN04-0.8 C, 69.97; H, 7.98; N, 3.40.
Pound: C, 69.95; H, 7.70; N, 3.52.
EXAMPLE 2 5(RS)-inethylphenyl-9(RS)-hydroxy- I 1'-hydroxy)-2'-phenyl)ethyl-3-(2"-methyl)propyl.3 -azabicyclo nonan-7-one (Compound 16). Table HI
HO.
Step 1: Compound G
K
2 C0 3 benzyl bromide ,OMe cat. Nal acetone A mixture of 1 (12.0 g, 56.0 inmol), benzyl bromide (10.1 g, 7.0 mL, 58.8 minol), potassium carbonate (48.4 g, 350 mmol), and sodium iodide (250 mg, 1.7 mmol) in acetone (200 mL) was heated at reflux for 16 h. The heterogeneous mixture was then poured into water (150 mL). The aqueous mixture was extracted with ethyl acetate (2 x 200 ml). The combined organic layers were dried over Na2SO4 and W-O 97/40833 PCT/US97/07131 -33concentrated to give G as a colorless oil (18.7 Rf 0.16 EtOAc/Hexane)] which was used without further purification.
1 H NMR (400 MHz, CDC13) 8 7.17-7.26 5H), 3.91-4.03 4H), 3.63 3H), 3.15 1H, J 13.6 Hz), 3.03 1H, J 13.6 Hz), 2.97 (ddd, 1H, J 15.0, 8.1, 12.3 Hz), 2.58 (dd, 1H, J 14.1, 2.9 Hz), 2.49 (ddd, 1H, J 15.0, 4.8, 3.7 Hz), 1.91-1.95 2H), 1.78 1H, J 13.9 Hz).
Step 2: Compound H 0 0 0 0 ANaOMe OMe THF j^OMe Ph O O O O 0 0 0 0 \-i G
H
Sodium hydride (2.40 g, 61 mmol, 60 wt% in mineral oil) was washed with hexane to remove mineral oil and then was suspended in tetrahydrofuran (80 mL) at 0°C. Anhydrous methanol (2.15 g, 2.72 mL, 67.1 mmol) was added dropwise over 3 min., followed by warming to 23 0 C and stirred for 30 min. The resulting suspension was cooled to 0°C and G was added via dropping funnel in tetrahydrofuran (40 mL) over 30 min. The reaction mixture was allowed to warm to 23°C over 1 h and then was stirred at that temperature for 16 h.
Aqueous acetic acid 10 mL) was carefully added and the mixture was poured into saturated NaHCO3 (100 mL), washed with EtOAc (2 x 150 mL), dried (Na2SO4) and concentrated to give a brown oil.
Recrystallization from MeOH afforded H as white prisms (11.2 The mother liquor was concentrated and purified by flash chromatography EtOAc/Hexane) to give a colorless oil which was further purified by recrystallization from Et20 to give H as white prisms (11.4 g WO 97/40833 PCT/US97/07131 -34overall, 67% yield for two steps), Rf 0.32 (30% EtOAc/Hexane), mp 110-115 0
C.
IH NMR (CDC13) 5 7.13-7.29 5H), 3.90-4.00 4H), 3.81 (dd, 1H, J 13.9, 5.7 Hz), 3.77 3H), 3.22 (dd, 1H, J 14.1, 5.0 Hz), 2.97-3.05 1H), 2.45 (dd, 1H, J 14.1, 8.6 Hz), 2.35 1H, J 13.6 Hz), 2.18 (ddd, 1H, J 13.4, 5.7, 3.8 Hz), 1.98 (ddd, IH, J 13.2, 5.9, 3.8 Hz), 1.76 1H, J 13.4 Hz).
Step 3: Compound I 0 0 isobutyllamine 0\
CH
2 0 OMe HOAc 0 MeOH Ph 0 0 0 COCH 3
H
Isobutylamine (4.07 mL, 40.9 mmol), glacial acetic acid (2.28 mL, 39.8 mmol), aqueous formaldehyde 25.0 mL, 373 mmol), and 3 (10.38 g, 34.1 mmol) were heated at reflux in MeOH (200 mL) for 48 h. The reaction was then concentrated, diluted with EtOAc (125 mL), and poured into saturated NaHCO3 (100 mL). The biphasic system was partitioned and the aqueous layer was washed with additional EtOAc (2 x 100 mL). The combined organics were dried (Na2SO4), concentrated, eluting with Et20 (50 mL), and then filtered through silica gel washing with Et20 (500 mL). The filtrate was concentrated to give 4 as a colorless oil [13.2 g, 96%, Rf=0.27 EtOAc/Hexane)]. This material was used without further purification.
1 H NMR (CDC13) 5 7.15-7.29 5H), 3.87-4.03 4H), 3.80 (s, 3H), 3.02 (dd, 1H, J 11.0, 3.1 Hz), 2.90 1H, J 13.9 Hz), 2.87 (d, WO 97/40833 PCT/US97/07131 1H, J 14.1 Hz), 2.75 2H, J 11.0 Hz), 2.68 (dd, 1H, J 13.2, Hz), 2.54 1H, J 13.0 Hz), 2.38 1H, J 13.2 Hz), 2.14-2.26 (m, 3H), 2.11 (dd, 1H, J 13.2, 3.5 1.57-1.74 1H), 0.91 3H, J 6.6 Hz), 0.89 3H, J 6.6 Hz).
Step 4: Compound J O NaBH 4 O Ph
CH
2
CI
2
/H
2 0/EtOH Ph
HO
SCCO
2
CH
3 J equatorial isomer K axial isomer Ethanol was added to a suspension of I in dichloromethane: water:ethanol (200 mL, 1:2:1) until the solution became homogeneous.
Sodium borohydride was added in one gram portions until TLC indicated that I had been consumed (7 x 1.0 The reaction mixture was cooled to 0°C and acetone was slowly added until it no longer provoked gas evolution. The resulting mixture was then poured into saturated NaCI (150 mL) and washed with EtOAc (2 x 250 mL). The organic layer was dried (Na2SO4), concentrated, and purified by flash chromatography (30% EtOAc/Hexane) to give a mixture of J and K as a colorless oil (9.49 g, 72% yield), Rf 0.23 (30% EtOAc/Hexane).
Compound J: 1H NMR (400 MHz, CDC13) 8 7.22-7.30 5H), 4.55 1H, J 11.2 Hz), 3.95-4.15 4H), 3.73 3H), 3.47 IH, J 11.3 Hz), 2.96 1H, J 13.4 Hz), 2.83 (dd, 1H, J 14.6, 2.1 Hz), 2.68 1H, J 13.4 Hz), 2.53 (dd, 1H, J 10.6, 2.1 Hz), 2.42 (dd, 1H, J 10.8, 2.2 Hz), 2.19 IH, J 10.4 Hz), 1.91-2.05 5H), 1.85 (d, 1H, J 10.7 Hz), 1.58-1.67 1H), 0.85 6H, J 7.4 Hz).
WO 97/40833 PCT/US97/07131 -36- Compound K: 1 H NMR (400 MHz, CDC13) 8 7.19-7.29 5H), 4.24 1H), 3.70-4.08 4H), 3.70 3H), 2.89 IH, J 13.2 Hz), 2.84 IH, J 10.0 Hz), 2.62 1H, J 10.8 Hz), 2.49 1H, J 13.4 Hz), 2.46 IH, J 11.9 Hz), 2.26 IH, J 10.8 Hz), 2.20 1H, J 14.1 Hz), 2.14 2H, J 7.3 Hz), 1.96 IH, J 14.5 Hz), 1.90 (d, 1H, J 14.0 Hz), 1.79 1H, J 14.1 Hz), 1.76-1.81 1H), 0.89 (d, 3H, J 3.9 Hz), 0.88 3H, J 4.0 Hz).
Step Compound L: TESCI 0 pyridine
O
J K cat. DMAP Ph S, CO 2
CH
3
L
A mixture of J and K (2.16 g, 5.3 mmol, 1:1) in pyridine mL) at 0°C was treated with chlorotriethylsilane (4.03 g, 4.48 mL, 26.7 mmol). 4-Dimethylaminopyridine (2 mg) was added and the reaction mixture was heated at 60 0 C for 4 h. The reaction mixture was then concentrated, and the residue was partitioned between Et20 (100 mL) and saturated NaHCO3 (100 mL). The organic layer was washed with saturated NaCI solution, dried (Na2SO4), and concentrated. The resulting oil was purified by flash chromatography EtOAc/Hexane) to give L as a colorless oil (1.14 g, 41 yield of desired isomer), Rf= 0.33 (10% EtOAc/Hexane).
1H NMR (400 MHz, CDC13) 5 7.14-7.31 5H), 4.04 IH), 3.76- 3.83 4H), 3.71 3H), 2.83 (dd, 1H, J 11.3, 1.5 Hz), 2.78 2H), 2.62 (dd, IH, J 11.6, 1.5 Hz), 2.40 (dd, IH, J 11.8, 3.3 Hz), 2.08 (dd, 2H, J 7.3, 1.5 Hz), 2.00 2H, J 11.6 Hz), 1.88 IH, J 11.6 Hz), 1.82 (dd, 1H, J 11.5, 3.4 Hz), 1.71 1H, J 11.4 Hz), WO 97/40833 PCT/US97/07131 -37- 1.65-1.70 1H), 0.97 9H, J 8.0 Hz), 0.83 3H, J 7.2 Hz), 0.85 3H, J 6.7 Hz), 0.63 6H, J 7.9 Hz).
Step 6: Compound M L
M
Sr 0 Si 0
CO
2
CH
3 CHO L
M
Diisobutylaluminum hydride (1.0 M in toluene, 2.91 mL, 2.91 mmol) was cooled to -78 0 C and added via cannula to a solution of L (753 mg, 1.45 mmol) in toluene (10 mL) at -78 0 C. The reaction mixture was stirred for 20 min and then acetone (5 mL) at -78 0 C was added via cannula to destroy excess reagent. The mixture was allowed to warm to 23 C, poured into saturated sodium potassium tartrate (100 mL), and the resulting suspension was washed with EtOAc (2 x 100 mL). The organic layer was dried (Na2SO4) and concentrated to give a mixture of L and M as a colorless oil which was azeotropically dried with toluene (2 x 40 mL) and used without further purification (720 mg, 3:1 mixture of M:L, 75% yield of Rf 0.41 EtOAc/Hexane)].
1H NMR (400 MHz, CDC13) 8 9.66 1H), 7.13-7.32 5H), 3.71- 3.82 5H), 2.78 2H, J 3.4 Hz), 2.74 (dd, 1H, J 11.8, 2.0 Hz), 2.66 (dd, 1H, J 11.8, 2.0 Hz), 2.21 (dd, 1H, J 11.9, 2.6 Hz), 2.10 (dd, 2H, J 7.5, 1.6 Hz), 1.93 1H, J 1.8 Hz), 1.92 1H, J 11.9 Hz), 1.83-1.87 3H), 1.65-1.73 1H), 0.97 9H, J 8.0 Hz), 0.85 3H, J 6.7 Hz), 0.83 3H, J 7.2 Hz), 0.63 6H, J 7.9 Hz).
WO 97/40833 PCT/US97/07131 -38- Step 7: Compound N 0 BenzylMgCI Ph M+ L THF Si K HO
N
A mixture of M and L (955 mg, 3:1, 1.45 mmol of L) in anhydrous THF (10 mL) at -78 0 C was treated with benzylmagnesium chloride (4.75 mL, 9.80 mmol, 2.06 M in THF) over 3 min. The reaction mixture was warmed to 0°C and was kept at that temperature for 1 h. Saturated NH4C1 was added (5 mL), and the heterogeneous mixture was poured into saturated NaHCO3 (100 mL) and was washed with EtOAc 2 x 100 mL). The organic layer was dried (Na2SO4), concentrated and purified by flash chromatography EtOAc/ Hexane), to isolate the desired isomer N as a colorless oil (374 mg, Rf= 0.23 (10% EtOAc/Hexane).
1 H NMR (400 MHz, CDC13) 5 7.13-7.35 10H), 3.74-3.84 6H), 2.88 1H, J 13.7 Hz), 2.80 1H, J 13.4 Hz), 2.77 1H, J 11.4 Hz), 2.75 1H, J 13.4 Hz), 2.62 1H, J 11.4 Hz), 2.47 (dd, 1H, J 13.7, 10.9 Hz), 2.10 (dd, J 7.2, 4.9 Hz), 1.99 IH, J 11.4 Hz), 1.90 1H, J 11.5 Hz), 1.85 1H, J 11.4 Hz), 1.85 IH), 1.70-1.78 4H), 0.99 9H, J 8.0 Hz), 0.86 3H, J 6.2 Hz), 0.85 3H, J 6.2 Hz), 0.65 6H, J 8.0 Hz).
WO 97/40833 WO 9740833PCTIUS97/07131 39 StQp8: Comnpouind16 Ph0 Ph Ph 3 NHCI HO si~~o Acetone HJ.
N 16 Aqueous HCI (3 N, 10 mL) was added to a solution of N (374 mg, 0.64 mmol) in acetone (10 mL). The mixture was heated at for 16 h. After cooling to 23'C, the reaction mixture was slowly poured into saturated NaHCO3 (75 mL). The biphasic system was extracted with EtOAc (2 x 150 mL), and the combined organic layers were dried (Na2SO4), and concentrated to give 16 as a white solid (265 mg, Rf 0.13 (30% EtOAc/Hexane), mp, 138-141'C.
IH NMR (CDCl3) 8 7.16-7.37 (in, 1OH), 3.88 1H), 3.83 1H), 3.66 I H, J 11.2 Hz), 2.98 I1H, J 15.9 Hz), 2.90 1 H, J 13.4 Hz), 2.69 (mn, 1H), 2.81 2H), 2.65 2H, J =13.9 Hz), 2.48 (d, 2H, J= 11.4 Hz), 2.00-2.09 5H), 1.91 1H, J 11.5 Hz), 1.53- 1.66 (in, I1H), 0.79 d, 3H, J 6.4 Hz), 0.78 3H, J =6.6 Hz).
Anal. Calcd for C27H35NO3-0.30 C, 75.95; H, 8.40; N, 3.28.
Found: C, 75.91; H, 8.30; N, 3.55.
HRMS calcd for C27H35N03 422.2695, found 422.2693.
WO 97/40833 PCTIUS97/07131 EXAMPLE 3 5(RS)-methylpheny-9(RS)-hydroxy- '-hydroxy)-2'-phenyl)ethvl-3-benzvl-3-azabicvclo[3.3.1 nonan-7-one (Compound 18). Table II
OHO
7 N Ph
HO
H H Ph Step 1: Compound S 0 1) S0 3 :pyr/DMSO O HO NN Ph TEA S Ph 2) Et 3 SiCI/Pyr Et 3 Si H H OH H O 0
S
A mixture of diols (162 mg, 0.396 mmol), and S03*pyridine complex (189 mg, 1.19 mmol) were dissolved in dry DMSO (4 mL). Triethylamine (0.34 mL) was then added dropwise via syringe. The reaction was allowed to proceed for 1.5 hours at which point it was poured into saturated NH4C1 solution. The aqueous phase was extracted with EtOAc (2 x 50 mL). The organics were combined and washed with H20, NaCI and dried (Na2SO4). The extracts were filtered, concentrated to an oil (147 mg) that was used in the next step without purifcation. The crude mixture of hydroxyaldehydes were dissolved in pyridine (6 mL) and treated with triethylsilyl chloride (0.34 mL, 20 mmol) and catalytic amounts of 4 -dimethylaminopyridine mg). The reaction was allowed to proceed at 60 0 C for 16 hours.
WO 97/40833 PCT/US97/07131 -41 The reaction was cooled to room temperature and the volatiles were removed via rotorevaporater. The residue was diluted with Et20 mL) and washed succesively with NaHCO3, H20 and NaC1. The organics were dried over Na2SO4 and concentrated. Flash chromatography Hexanes/EtOAc) gave the desired compound
(S)
as an oil (62 mg, 28%).
1 H NMR (400 MHz, CDC13) 5 9.54 1H), 7.14-7.31 10H), 3.82 5H), 3.54 (AB, JAB 13Hz, 2H 2.78 (AB, JAB 13.5Hz, 2H), 2.71 2H), 2.19 (dd, J 11.7Hz, 3.1Hz, IH), 1.96 4H), 0.99 J 8Hz, 9H), 0.65 J 8Hz, 6H).
Step 2: Compound 18 O 0 0 HO N Ph Ph
HO
Et3Si' 1) PhCH 2 MgClTHF
H
H 0
H
HO Ph 2) H30O/acetone 18 A solution of benzyl magnesium chloride in THF (2.06M, 0.2 mL) was added to a solution of the aldehyde S from step 1 (61 mg, 0.109 mmol) in dry THF (1 mL) at -78 0 C. The reaction was stirred at -78 0 C for 1.5 hours then slowly warmed to room temperature and excess Grignard was quenched with saturated NH4CI solution. The reaction was diluted with EtOAc (60 mL) and washed with NH4CI, NaCl and dried over Na2SO4. The material was immediately hydrolyzed in THF/1N HCI 2.5mL). The desired compound was purified via flash chromatography 1:1 EtOAc/Hexanes and crystallized from Et20/Hexanes. m. p. 145.5-147°C.
I
WO 97/40833 PCTALJS97/07131 42 1H NMR (400 MHz, CDCI3) 5 7.17-7.34 (in, 15H), 3.90 J =9.9 Hz, 3.58 (in, 3.35 J 13.4 Hz, I1H), 3.00 J =15.7 Hz, I H), 2.43-2.84 (in, 5H), 2.81 (AB, JAB =13.5 Hz, 2H), 2.0-2.12 (in, 3H).
Low resolution FAB Mass spec 1) m/z 456.
Anal calc'd for C30H33N03-0.65 C, 77.10; H, 7.40, N, Found: C, 71.15; H, 7.24; N, 3. EXAMPLE 4 )-inethylpheny-9(RS)-hydroxy- I 1 -hydroxy)-2'- (tetra-.
hydro-1I,2-thiazine 1,1 -dioxide))-ethyl-3-benzyl-3azabicyclo[3 nonan-7-one (Compound Table II 0,,P Step1: Compound T Me 3 S(0) I/NaH Ph DMF OC
S
WO 97/40833 PCT/US97/07131 -43- A solution of trimethylsulfoxonium iodide (298 mg, 1.35 mmol), NaH (32 mg, 1.35 mmol) and DMF (4 mL) were stirred at 0 C for 30 minutes. A solution of aldehyde S from above (152 mg, 0.2709 mmol) in DMF (0.5 mL) was added via syringe. The transfer was completed with two washings of DMF (2 x 0.25 mL). The reaction was stirred at 0°C for 1 hour and quenched with a saturated solution of NH4CI. The reaction was poured into NaCI and extracted with Et20 (3 x 35 mL). The organics were combined and washed with H20, NaCI, and dried over Na2SO4. Flash chromatography using 8:1 Hexane/EtOAc gave 68 mg of one diastereomer and 25 mg of a second diastereomer (both oils).
Major more polar isomer 1 H NMR (400 MHz, CDC13), 8 7.14-7.3 (m, 3.81 4H), 3.60 1H), 3.50 (AB, JAB 13.2 Hz, 2H), 2.67 (br t, J 3 Hz, 1H), 2.78 (AB, JAB 13.5 Hz, 2H), 2.72 J 11.4 Hz, 1H), 2.66 2H), 2.60 J 11.4 Hz, 1H), 1.96 J 11.4 Hz, 1H), 1.89 J 11.5 Hz, 1H), 1.82 (dd, J 11.5 Hz, 2.9 Hz, 1H), 1.76 (dd, J 11.4 Hz, 9.4 Hz, 1H), 1.53 (dd, J 11.9 Hz, 3.1 Hz, 1H), 1.00 J 7.5 Hz, 9H), 0.65 J 7.5 Hz, 6H).
Minor less polar isomer 1 H NMR (400 MHz, CDC13), 8 7.10-7.32 (m, 3.79 4H), 3.50 (AB, JAB 13.2 Hz, 2H), 3.23 1H), 2.62- 2.77 7H), 1.80-1.96 4H), 1.70 (dd, J 11.5 Hz, 2.5 Hz, 1H), 0.98 J 7.7 Hz, 9H), 0.64 J 7.7 Hz, 6H).
Step 2: Compound 23 0 H NH Ph H -N Ph 4 H
HO
Et 3 SiO Ho
S
H 0 1)NaHTHF S2 H ",OH T 2)H 3
O
/acetone
SO
WO 97/40833 PCT/US97/07131 -44- NaH (8 mg, 60 wt% in mineral oil, 200 pmol) was added to a solution of tetrahydro-l,2-thiazine-1,1-dioxide (34 mg, 250 pmol) in N,N-dimethylformamide (1 mL) and the resulting mixture was stirred at 23°C for 30 min. A solution of T (27 mg, 50 pmol) in N,Ndimethylformamide (1 mL) was added and the mixture was heated at 0 C for 16 h. The reaction was cooled to room temperature and NH4CI solution (2 mL) was added. The reaction mixture was poured into water (50 mL) and the resulting aqueous mixture was extracted with Et20 (2 x 50 mL). The combined organic extracts were washed with water (25 mL), dried (Na2SO4), and concentrated to give a mixture of starting material and desired product which was used without further purification.
A solution containing the crude reaction mixture from above (30 mg) in acetone (3 mL) was treated with aqueous hydrochloric acid (3 N, 3 mL) and the colorless solution was heated at 65 0 C for 12 h.
The reaction mixture was cooled to room temperature and slowly poured into saturated NaHCO3 (25 mL). The resulting suspension was washed with EtOAc (30 mL) and the organic layer was dried (Na2SO4), and concentrated to give a crude oil which was purified by flash chromatography (70% EtOAc/Hexane) to give 23 as a white solid mg, 58% for 2 steps). Rf= 0.13 (70% EtOAc/Hexane), mp 163-164
°C.
IH NMR (400 MHz, CDC13) 5 7.13-7.33 10H), 3.80 (bs, 1H), 3.63 (bd, 1H, J 10.1 Hz), 3.59 (bs, 1H), 3.51 1H, J 13.4 Hz), 3.21- 3.39 4H), 3.01-3.06 3H), 2.86 1H, J 15.6 Hz), 2.79 1H, J 13.4 Hz), 2.73 1H, J 13.4 Hz), 2.66 1H, J 15.6 Hz), 2.53 (dd, 1H, J 11.3, 1.8 Hz), 2.37 (dd, 1H, J 10.9, 1.9 Hz), 2.20 (quintet, 2H, J 6.0 Hz), 1.93-2.09 4H), .1.64-1.66 2H).
Anal calcd. for C28H36N2SO5.0.80 C, 63.81; H, 7.19; N, 5.31.
Found: C, 63.81; H, 7.02; N, 5.18; W0 97/40833 PCTUS97/07131 45 HRMS calcd for C28H36N2S05 513.2423, found 513.2437.
EXAMPLE 5(RS)-methylpheny-9(RS)-hydroxy- I 1I'hydroxy)-2-(2I -amino)phenyl)-ethyl-3-benzyw3-azabicyclo[3 .3.1 ]nonan-7-one (Compound Table 11
HO
H.
HO"
H
2
N/
THF, -40 P0
TESO
CHO
WO 97/40833 PCT/US97/07131 -46- A solution of tert-butyllithium in pentane (1.7 M, 3.00 mL, 5.10 mmol, 9.64 equiv) was added over 1 min to a solution of tert-butyl 2-methylcarbanilate (515 mg, 2.48 mmol, 4.69 equiv) in THF (3.5 mL) at -40 The resulting bright yellow mixture was stirred at -40 °C for 15 min, then a solution of the aldehyde, U, (300 mg, 0.529 mmol, 1 equiv) in THF (4 mL) was added. The resulting mixture was warmed to 0°C and was held at that temperature for 15 min. The product solution was diluted with pH 7 phosphate buffer solution (100 mL), and the resulting aqueous mixture was extracted with EtOAc 2 x 75 mL).
The combined organc layers were dried over Na2SO4 and were concentrated. The residue was purified by flash chromatography EtOAc in hexanes initially, grading to 20% ethyl acetate in hexanes) to provide the desired alcohol as a colorless oil (85 mg, 21%) as well as the undesired diastereomeric alcohol as a colorless oil (81 mg, 1 H NMR (400 MHz, CDC13), 5 7.78 (br s, 1H, NH), 7.59 (br d, 1H, J 7.9 Hz, ArH), 7.32-7.07 7H, PhH and ArH), 7.00 (br t, 1H, J 7.3 Hz, ArH), 3.81 2H, OCH2CH20), 3.75 2H, OCH2CH20), 3.67 1H, (CH3CH2)3SiOCH), 3.11 (br d, 1H, J 5.1 Hz, HOCH), 2.77- 2.56 6H, PhCH2, ArCH2 and NCH2), 2.10 2H, NCH2CH(CH3) 2 and NCH2 or CH2), 2.03 1H, J 11.2 Hz, NCH2 or CH2), 1.94 (d, 1H, J 11.4 Hz, NCH2 or CH2), 1.84 2H, NCH2CH(CH3)2 and NCH2 or CH2), 1.71 3H, NCH2 and/or CH2 and NCH2CH(CH3)2, 1.52 9H, OC(CH3)3), 0.98 9H, J 7.9 Hz, (CH3CH2)3Si), 0.86 3H, J 6.4 Hz, CH(CH3)2), 0.85 3H, J 6.4 Hz, CH(CH3)2), 0.64 6H, J 7.9 Hz, (CH3CH2)3Si).
TLC (20% EtOAc-hexanes), Rf: desired alcohol: 0.36 undesired alcohol: 0.44 (UV) WO 97/40833 PCT/US97/07131 -47- TESO 3M HCI HO N
H
HO
HO
BocHN
H
2
N
V A solution of the ketal (80 mg, 0.11 mmol) in a mixture of aqueous 3 M HCI solution (10 mL) and acetone (10 mL) was heated at 0 C for 16 h. After cooling to 23 0 C, the product solution was carefully diluted with aqueous saturated NaHCO3 solution (100 mL).
The resulting aqueous mixture was extracted with EtOAc (2 x 50 mL).
The combined organic layers were dried over Na2SO4 and were concentrated. The residue was purified by flash chromatography EtOAc in Hexanes initially, then 40% Hexanes in EtOAc) to afford the product ketone as a colorless oil (31 mg, The product oil was triturated with Et20 to produce a white crystalline solid (mp 164- 165 0
C).
1 H NMR (400 MHz, CDC13) 8 7.32-7.12 5H, PhH), 7.08 (td, 1H, J 7.6, 1.4 Hz, ArH), 7.00 (dd, 1H, J 7.5, 1.3 Hz, ArH), 6.80 (td, 1H, J 7.5, 1.1 Hz, ArH), 6.71 (dd, 1H, J 7.9, 0.7 Hz, ArH), 3.98 (br s, 1H, OH), 3.88 (br s, 1H, HOCH), 3.74 (br s, 2H, NH), 3.68 (dd, 1H, J 10.4, 1.7 Hz, HOCH), 2.87 1H, J 15.9 Hz, NCH2), 2.77 3H, PhCH2 and ArCH2), 2.65 (br d, 1H, J 14.7 Hz, ArCH2), 2.62 1H, J 16.1 Hz, NCH2), 2.46 2H, NCH2CH(CH3) 2 2.04 NCH2 and CH2), 1.91 IH, J 11.5 Hz, NCH2 or CH2), 1.61 1H, NCH2CH(CH 3 2 0.77 3H, J 6.6 Hz, CH(CH3)2), 0.76 3H, J 6.4 Hz, CH(CH3)2).
I
M
WO 97/40833 PCT/US97/07131 -48- High-Res MS (FAB): Calcd for C27H36N203 437.2804 Found: 437.2813 Calcd for C27H36N20 3 C, 74.28; H, 8.31; N, 6.42.
Found: C, 74.28; H, 8.34; N, 6.52 TLC (40% EtOAc-hexanes), Rf: 0.06 While the foregoing specification teaches the principles of the present invention, with examples provided for the purpose of illustration, it will be understood that the practice of the invention emcompasses all of the usual variations, adaptations, or modifications, as come within the scope of the following claims and its equivalents.
I

Claims (11)

  1. 4. .4* 9*4* 4* *4 9 9*
  2. 9. 9 0* S S 9. 9* 9* 4. 9. *4 9 9 9 wherein X is -NR 4 or Y is 0, or forms, with the carbon to which it is attached, OH C \H Z is 0, or forms, with the carbon to which it is attached, OH C H 10 R 1 is a) b) C) d) 15 e) f) H, C 1 4 alkyl; C 3 7 cycloalkyl; aryl, unsubstituted or substituted one or more times with hydroxy; CH 2 R 5 or 5-7 membered heterocycle; and [n:\Iibc]00082:tab WO 97/40833 PCT/US97/07131 50 R 2 is a) CI1-4 alkyl; b) aryl, unsubstituted or substituted with aryl; c) CH2R 6 or d) heterocycle; and R 3 is a) CH(OH)R 7 or b) CH(NI-12)R 7 and R 4 is a) Ci1-4 alkyl; b) C3-6 cycloalkyl; c) aryl unsubstituted or substituted with halo or with C 1 -4 alkyl unsubstituted or substituted one or more times with hydroxy; d) CH2R 1 ;or e) 5-7 membered heterocycle; and R 5 is a) C 1-4 alkyl; or b) aryl; and R 6 is a) C 1-4 alkyl; b) aryl unsubstituted or substituted with halo or with Ci1 -4 alkyl unsubstituted or substituted one or more times with hydroxy; or c) 5-7 membered heterocycle; and R 7 is a) H;, b) C 1-4 alkyl; c) aryl unsubstituted or substituted with amino; d) C1- 3 alkylaryl unsubstituted or substituted with amino; or e) 5-7 membered heterocycle; or pharmaceutically acceptable salt thereof. 2. A compound of the formula 0 R 2 R 4 HO HO R wherein R 2 is Ci-4 alkylene-aryl; and R 4 is C1-4 alkyl, unsubstituted or substituted with aryl, C3-6 cycloalkyl, or 5-7 membered heterocycle; R 7 is H, benzyl unsubstituted or substituted with amino; or pharmaceutically accetpable sat thereof. 3. The compound A 0 HoC o HO H. A HO H 2 N or pharmaceutically acceptable salts thereof. 15 4. The compound B *o HO B HO or pharmaceutically acceptable salts thereof. 5. A 3-aza-bicyclo[3.3.1]nonane-7,9-dione, 7-hydroxy-3-aza-bicyclo[3.3.1]nonan-9-one, 9-hydroxy-3-aza-bicyclo[3.3.1]nonan-7-one or 3-aza-bicyclo[3.3.1]nonane-7,9-diol derivative, S substantially as hereinbefore described with reference to any one of the examples. [R\LIBAA]5018 TAB 6. The combination of a compound of any one of claims 1 to 5 and one or more of 6-chloro-4(S)-cyclopropylethynyl-4(S)-trifluoromethyl-1 ,4-dihydro-2H-3, 1-benzoxazin-2-one, AZT, dd I or ddC. 7. A combination of compounds which is 6-chloro-4(S)-cyclopropyiethynyl-4(S)- trifluoromethy-1 ,4-dihydro-2H-3, 1-benzoxazin-2-one with compound A HO A H; HO H2NQ and, optionally, one or more of AZT or ddl or ddC. 8. A combination of compounds which is compound A 0 0 HO A H; HO H 2 N and one or more of compound B, H B H C) AZT, ddlIor ddC. A combination of compouds which is ()6-chloro-4(S)-cycloprpylethynyl-4(S)- trifluoromethyl-1 ,4-dihydro-2H-3, 1-benzoxazin-2-one with compound B [R:\LIBAA]5018:TAB 0 0 N, HO B HO and, optionally, one or more of AZT or ddl or ddC. A pharmaceutical composition including or consisting of an effective amount of at least one compound according to any one of claims 1 to 5 or of a combination according to any one of claims 6 to 9, together with a pharmaceutically acceptable carrier, diluent or adjuvant therefor.
  3. 11. A method for treating or delaying the onset of AIDS in a mammal, which method includes or consists of administering to said mammal an effective amount of a compound according to any one of claims 1 to 5, a combination according to any one of claims 6 to 9, or a composition according to claim
  4. 12. A compound according to any one of claims 1 to 5, a combination according to any one of claims 6 to 9 or a composition according to claim 10 when used for treating or delaying the onset of AIDS.
  5. 13. The use of a compound according to any one of claims 1 to 5 or a combination according to any one of claims 6 to 9 for the manufacture of a medicament for treating or delaying the onset of AIDS.
  6. 14. A method for preventing infection by HIV in a mammal, which method includes or consists of administering to said mammal an effective amount of a compound according to any one of claims 1 to 5, a combination according to any one of claims 6 to 9, or a composition according to claim o 20 15. A compound according to any one of claims 1 to 5, a combination according to any one of claims 6 to 9 or a composition according to claim 10 when used for preventing infection by HIV.
  7. 16. The use of a compound according to any one of claims 1 to 5 or a combination according to any one of claims 6 to 9 for the manufacture of a medicament for preventing infection by HIV.
  8. 17. A method for treating infection by HIV in a mammal, which method includes or consists of administering to said mammal an effective amount of a compound according to any one of claims 1 to 5, a combination according to any one of claims 6 to 9, or a composition according to claim S18. A compound according to any one of claims 1 to 5, a combination according to any one of claims 6 to 9 or a composition according to claim 10 when used for treating infection by HIV. [R:\LIBAA]5018TAB
  9. 19. The use of a compound according to any one of claims 1 to 5 or a combination according to any one of claims 6 to 9 for the manufacture of a medicament for treating infection by HIV. A method for inhibiting HIV protease in a mammal, which method includes or consists of administering to said mammal an effective amount of a compound according to any one of claims 1 to a combination according to any one of claims 6 to 9, or a composition according to claim
  10. 21. A compound according to any one of claims 1 to 5, a combination according to any one of claims 6 to 9 or a composition according to claim 10 when used for inhibiting HIV protease.
  11. 22. The use of a compound according to any one of claims 1 to 5 or a combination according to any one of claims 6 to 9 for the manufacture of a medicament for inhibiting HIV protease. Dated 2 September 1999 MERCK CO., INC. Patent Attorneys for the Applicant/Nominated Person SPRUSON&FERGUSON S** S S S S g o** *o oa*° **o *o [R:\LIBAA15018:TAB
AU27473/97A 1996-05-02 1997-04-29 HIV protease inhibitors useful for the treatment of AIDS Ceased AU712120B2 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US1669596P 1996-05-02 1996-05-02
US60/016695 1996-05-02
GB9613487 1996-06-27
GBGB9613487.9A GB9613487D0 (en) 1996-06-27 1996-06-27 HIV protease inhibitors useful for the treatment of aids
PCT/US1997/007131 WO1997040833A1 (en) 1996-05-02 1997-04-29 Hiv protease inhibitors useful for the treatment of aids

Publications (2)

Publication Number Publication Date
AU2747397A AU2747397A (en) 1997-11-19
AU712120B2 true AU712120B2 (en) 1999-10-28

Family

ID=26309579

Family Applications (1)

Application Number Title Priority Date Filing Date
AU27473/97A Ceased AU712120B2 (en) 1996-05-02 1997-04-29 HIV protease inhibitors useful for the treatment of AIDS

Country Status (5)

Country Link
EP (1) EP0914125A4 (en)
JP (1) JP2000509392A (en)
AU (1) AU712120B2 (en)
CA (1) CA2252915A1 (en)
WO (1) WO1997040833A1 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP4626896B2 (en) * 1998-02-13 2011-02-09 ダイセル化学工業株式会社 Acylating agent, acylation method using the same, and adamantane derivative
ZA99981B (en) 1998-02-17 2000-08-08 Du Pont Pharm Co Oral liquid formulations of benzoxazinones HIV reverse transcriptase inhibitors.
JP2002518452A (en) * 1998-06-24 2002-06-25 エモリ ユニバーシティ Use of 3'-azido-2 ', 3'-dideoxyuridine in combination with other anti-HIV drugs for the manufacture of a therapeutic drug for HIV

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5504104A (en) * 1993-11-19 1996-04-02 Warner-Lambert Company Tricyclic pyrone derivatives as protease inhibitors and antiviral agents
US5510375A (en) * 1993-11-19 1996-04-23 Warner-Lambert Company Coumarin derivatives as protease inhibitors and antiviral agents
US5480887A (en) * 1994-02-02 1996-01-02 Eli Lilly And Company Protease inhibitors

Also Published As

Publication number Publication date
EP0914125A1 (en) 1999-05-12
WO1997040833A1 (en) 1997-11-06
EP0914125A4 (en) 2000-04-05
JP2000509392A (en) 2000-07-25
AU2747397A (en) 1997-11-19
CA2252915A1 (en) 1997-11-06

Similar Documents

Publication Publication Date Title
EP0766674B1 (en) New hiv protease inhibitors
JP4982482B2 (en) HIV integrase inhibitor
AU676563B2 (en) HIV protease inhibitors useful for the treatment of AIDS
US5308854A (en) Inhibitors of HIV reverse transcriptase
EP0617968B1 (en) HIV protease inhibitors in pharmaceutical combinations for the treatment of AIDS
US5846978A (en) HIV protease inhibitors useful for the treatment of AIDS
WO1993004047A1 (en) Quinazoline derivatives as inhibitors of hiv reverse transcriptase
IL103613A (en) Hiv protease inhibitors process and intermediats for their preparation and pharmaceutical compositions containing them
AU692509B2 (en) HIV protease inhibitors
PL175615B1 (en) Benzoxasinones as inhibitors of reverse hiv transcriptase
EP3452045A1 (en) Methods of treatment and combination therapies using gcase activator heterobicyclic and related compounds
US9416143B2 (en) Griseofulvin derivatives
JPH05148205A (en) Hiv protease inhibitor with symmetrical structure
US5650412A (en) HIV protease inhibitors useful for the treatment of AIDS
AU712120B2 (en) HIV protease inhibitors useful for the treatment of AIDS
WO1994026749A1 (en) Hiv protease inhibitors
US5747540A (en) HIV protease inhibitors useful for the treatment of AIDS
AU711713B2 (en) HIV protease inhibitors useful for the treatment of AIDS
US5811462A (en) HIV Protease inhibitors useful for the treatment of AIDS
GB2307683A (en) HIV protease inhibitors useful for the treatment of AIDS