WO2000042060A1 - Bis-amino acid sulfonamides containing n-terminally a substituted benzyl group as hiv protease inhibitors - Google Patents

Bis-amino acid sulfonamides containing n-terminally a substituted benzyl group as hiv protease inhibitors Download PDF

Info

Publication number
WO2000042060A1
WO2000042060A1 PCT/US2000/000820 US0000820W WO0042060A1 WO 2000042060 A1 WO2000042060 A1 WO 2000042060A1 US 0000820 W US0000820 W US 0000820W WO 0042060 A1 WO0042060 A1 WO 0042060A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
compound according
mmol
formula
hiv
Prior art date
Application number
PCT/US2000/000820
Other languages
French (fr)
Inventor
Robert F. Kaltenbach
George L. Trainor
Original Assignee
Dupont Pharmaceuticals Company
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 to BR0007854-9A priority Critical patent/BR0007854A/en
Priority to IL14376300A priority patent/IL143763A0/en
Priority to KR1020017008803A priority patent/KR20010101478A/en
Priority to CA002353088A priority patent/CA2353088A1/en
Priority to SK952-2001A priority patent/SK9522001A3/en
Priority to HU0105235A priority patent/HUP0105235A3/en
Application filed by Dupont Pharmaceuticals Company filed Critical Dupont Pharmaceuticals Company
Priority to JP2000593627A priority patent/JP2002536298A/en
Priority to EP00905604A priority patent/EP1140983A1/en
Priority to PL00349774A priority patent/PL349774A1/en
Priority to AU27257/00A priority patent/AU2725700A/en
Publication of WO2000042060A1 publication Critical patent/WO2000042060A1/en
Priority to NO20013338A priority patent/NO20013338L/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • C07K5/06017Dipeptides with the first amino acid being neutral and aliphatic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • C07K5/06017Dipeptides with the first amino acid being neutral and aliphatic
    • C07K5/06026Dipeptides with the first amino acid being neutral and aliphatic the side chain containing 0 or 1 carbon atom, i.e. Gly or Ala
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06191Dipeptides containing heteroatoms different from O, S, or N
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • This invention relates generally to bis-amino acid sulfonamides containing substituted benzyl amines useful as HIN protease inhibitors, pharmaceutical compositions and diagnostic kits comprising the same, and methods of using the same for treating viral infection or as assay standards or reagents.
  • HIV seropositive individuals are initially asymptomatic but typically develop AIDS related complex (ARC) followed by AIDS. Affected individuals exhibit severe immunosuppression which predisposes them to debilitating and ultimately fatal opportunistic infections.
  • ARC AIDS related complex
  • the disease AIDS is the end result of an HIV-1 or HIV-2 virus following its own complex life cycle.
  • the virion life cycle begins with the virion attaching itself to the host human T-4 lymphocyte immune cell through the bonding of a glycoprotein on the surface of the virion's protective coat with the CD4 glycoprotein on the lymphocyte cell. Once attached, the virion sheds its glycoprotein coat, penetrates into the membrane of the host cell, and uncoats its RNA.
  • the virion enzyme, reverse transcriptase directs the process of transcribing the RNA into single-stranded DNA. The viral RNA is degraded and a second DNA strand is created. The now double-stranded DNA is integrated into the human cell's genes and those genes are used for virus reproduction.
  • RNA polymerase transcribes the integrated DNA into viral RNA.
  • the viral RNA is translated into the precursor gag-pol fusion polyprotein.
  • the polyprotein is then cleaved by the HIV protease enzyme to yield the mature viral proteins.
  • HIV protease is responsible for regulating a cascade of cleavage events that lead to the virus particle's maturing into a virus that is capable of full infectivity.
  • the typical human immune system response killing the invading virion, is taxed because the virus infects and kills the immune system's T cells.
  • viral reverse transcriptase the enzyme used in making a new virion particle, is not very specific, and causes transcription mistakes that result in continually changed glycoproteins on the surface of the viral protective coat. This lack of specificity decreases the immune system's effectiveness because antibodies specifically produced against one glycoprotein may be useless against another, hence reducing the number of antibodies available to fight the virus.
  • the virus continues to reproduce while the immune response system continues to weaken. Eventually, the HIV largely holds free reign over the body's immune system, allowing opportunistic infections to set in and without the administration of antiviral agents, immunomodulators, or both, death may result.
  • Retroviral proteases most commonly process the gag precursor into the core proteins, and also process thepol precursor into reverse transcriptase and retroviral protease.
  • one object of the present invention is to provide novel protease inhibitors.
  • It is another object of the present invention to provide pharmaceutical compositions with protease inhibiting activity comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of at least one of the compounds of the present invention or a pharmaceutically acceptable salt form thereof.
  • R 1 is F
  • R 2 is F or H
  • R 3 is selected from the group: 4-aminophenyl, 3-aminophenyl, 2,3-dihydrobenzofuran-5- yl, and 1 ,3-benzodioxol-5-yl.
  • the present invention provides a novel compound of Formula II:
  • the present invention provides a novel compound of Formula Ila:
  • the present invention provides a novel compound of Formula Ila, wherein:
  • R 3 is 3-aminophenyl.
  • the present invention provides a novel compound of Formula Ila, wherein:
  • R 3 is 4-aminophenyl.
  • the present invention provides a novel compound of Formula Ila, wherein:
  • R 3 is 2,3-dihydrobenzofuran-5-yl or l,3-benzodioxol-5-yl.
  • the present invention provides a novel compound of Formula lib:
  • the present invention provides a novel compound of Formula lib, wherein:
  • R 3 is 3-aminophenyl.
  • R 3 is 4-aminophenyl.
  • the present invention provides a novel compound of Formula lib, wherein:
  • R 3 is 2,3-dihydrobenzofuran-5-yl or l,3-benzodioxol-5-yl.
  • the present invention provides a novel compound of Formula lie, wherein:
  • R 3 is 3-aminophenyl.
  • the present invention provides a novel compound of Formula lie, wherein:
  • R 3 is 4-aminophenyl.
  • the present invention provides a novel compound of Formula lie, wherein: R 3 is 2,3-dihydrobenzofuran-5-yl or 1 ,3-benzodioxol-5-yl.
  • the present invention provides a novel compound of Formula III:
  • the present invention provides a novel compound of Formula Ilia:
  • the present invention provides a novel compound of Formula IV:
  • the present invention provides a novel compound of Formula IVa:
  • the present invention provides a novel pharmaceutical composition
  • a novel pharmaceutical composition comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of a compound of formula I or pharmaceutically acceptable salt form thereof.
  • the present invention provides a novel method for treating HIV infection which comprises administering to a host in need of such treatment a therapeutically effective amount of a compound of formula I or pharmaceutically acceptable salt form thereof.
  • the present invention provides a novel method of treating HIV infection which comprises administering, in combination, to a host in need thereof a therapeutically effective amount of: (a) a compound of formula I; and,
  • the reverse transcriptase inhibitor is selected from the group AZT, ddC, ddl, d4T, 3TC, delavirdine, efavirenz, nevirapine, Ro 18,893, trovirdine, MKC-442, HBY 097, ACT, UC-781, UC-782, RD4-2025, and MEN 10979, and the protease inhibitor is selected from the group saquinavir, ritonavir, indinavir, amprenavir, nelfinavir, palinavir, BMS-232623, GS3333, KNI-413, KNI-272, LG-71350, CGP-61755, PD 173606, PD 177298, PD 178390, PD 178392, U- 140690, and ABT-378.
  • the reverse transcriptase inhibitor is selected from the group AZT, efavirenz, and 3TC and the protease inhibitor is selected from the group saquinavir, ritonavir, nelfinavir, and indinavir.
  • the reverse transcriptase inhibitor is AZT.
  • the protease inhibitor is ritonavir.
  • component (b) is a HIV reverse transcriptase inhibitor and a HIV protease inhibitor.
  • component (b) is two different HIV reverse transcriptase inhibitors.
  • the present invention provides a pharmaceutical composition useful for the treatment of HIV infection, which comprises a therapeutically effective amount of:
  • the present invention provides a novel method of inhibiting HIV present in a body fluid sample which comprises treating the body fluid sample with an effective amount of a compound of formula I.
  • the present invention to provides a novel a kit or container comprising a compound of formula I in an amount effective for use as a standard or reagent in a test or assay for determining the ability of a potential pharmaceutical to inhibit HIV protease, HIV growth, or both.
  • DEFINITIONS As used herein, the following terms and expressions have the indicated meanings. It will be appreciated that the compounds of the present invention contain asymmetrically substituted carbon atoms, and may be isolated in optically active or racemic forms. It is well known in the art how to prepare optically active forms, such as by resolution of racemic forms or by synthesis, from optically active starting materials. All chiral, diastereomeric, racemic forms and all geometric isomeric forms of a structure are intended, unless the specific stereochemistry or isomer form is specifically indicated.
  • Multigram scale is preferably the scale wherein at least one starting material is present in 10 grams or more, more preferably at least 50 grams or more, even more preferably at least 100 grams or more.
  • Multikilogram scale is intended to mean the scale wherein more than one kilogram of at least one starting material is used.
  • Industrial scale as used herein is intended to mean a scale which is other than a laboratory scale and which is sufficient to supply product sufficient for either clinical tests or distribution to consumers.
  • the present invention is intended to include all isotopes of atoms occurring on the present compounds.
  • Isotopes include those atoms having the same atomic number but different mass numbers.
  • isotopes of hydrogen include tritium and deuterium.
  • isotopes of carbon include C-13 and C-14.
  • HIV reverse transcriptase inhibitor is intended to refer to both nucleoside and non-nucleoside inhibitors of HIV reverse transcriptase (RT).
  • nucleoside RT inhibitors include, but are not limited to, AZT, ddC, ddl, d4T, and 3TC.
  • non-nucleoside RT inhibitors include, but are not limited to, delavirdine (Pharmacia and Upjohn, U90152S), efavirenz (DuPont), nevirapine (Boehringer Ingelheim), Ro 18,893 (Roche), trovirdine (Lilly), MKC-442 (Triangle), HBY 097 (Hoechst), HBY 1293 (Hoechst), ACT (Korean Research Institute), UC-781 (Rega Institute), UC-782 (Rega Institute), RD4-2025 (Tosoh Co. Ltd.), and MEN 10979 (Menarini Farmaceutici).
  • HIV protease inhibitor is intended to refer to compounds which inhibit HIV protease. Examples include, but are not limited, saquinavir (Roche, Ro31-8959), ritonavir (Abbott, ABT-538), indinavir (Merck, MK-639), amprenavir (Vertex/Glaxo Wellcome), nelfinavir (Agouron, AG-1343), palinavir (Boehringer Ingelheim), BMS-232623 (Bristol-Myers Squibb), GS3333 (Gilead Sciences), KNI-413 (Japan Energy), KNI-272 (Japan Energy), LG-71350 (LG Chemical), CGP-61755 (Ciba-Geigy), PD 173606 (Parke Davis), PD 177298 (Parke Davis), PD 178390 (Parke Davis), PD 178392 (Parke Davis), tipranavir (Pharmacia and Upjohn, U-
  • pharmaceutically acceptable salts refer to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof.
  • pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like.
  • the pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, and the like.
  • inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like
  • organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic,
  • the pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods.
  • such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred.
  • Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, PA, 1985, p. 1418, the disclosure of which is hereby incorporated by reference.
  • phrases "pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication commensurate with a reasonable benefit/risk ratio.
  • “Prodrugs” are intended to include any covalently bonded carriers which release the active parent drug according to formula (I) or other formulas or compounds of the present invention in vivo when such prodrug is administered to a mammalian subject.
  • Prodrugs of a compound of the present invention are prepared by modifying functional groups present in the compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound.
  • Prodrugs include compounds of the present invention wherein the hydroxy or amino group is bonded to any group that, when the prodrug is administered to a mammalian subject, cleaves to form a free hydroxyl or free amino, respectively.
  • Examples of prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of alcohol and amine functional groups in the compounds of the present invention, and the like.
  • Stable compound and “stable structure” are meant to indicate a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent. Only stable compounds are contempleted by the present invention.
  • Substituted is intended to indicate that one or more hydrogens on the atom indicated in the expression using “substituted” is replaced with a selection from the indicated group(s), provided that the indicated atom's normal valency is not exceeded, and that the substitution results in a stable compound.
  • “Therapeutically effective amount” is intended to include an amount of a compound of the present invention or an amount of the combination of compounds claimed effective to inhibit HIV infection or treat the symptoms of HIV infection in a host.
  • the combination of compounds is preferably a synergistic combination. Synergy, as described for example by Chou and Talalay, Adv. Enzyme Regul. 22:27-55 (1984), occurs when the effect (in this case, inhibition of HIV replication) of the compounds when administered in combination is greater than the additive effect of the compounds when administered alone as a single agent. In general, a synergistic effect is most clearly demonstrated at suboptimal concentrations of the compounds. Synergy can be in terms of lower cytotoxicity, increased antiviral effect, or some other beneficial effect of the combination compared with the individual components.
  • One diastereomer of a compound of Formula I may display superior activity compared with the other.
  • separation of the racemic material can be achieved by HPLC using a chiral column or by a resolution using a resolving agent such as camphonic chloride as in Thomas J. Tucker, et al, J. Med. Chem. 1994, 37, 2437-2444.
  • a chiral compound of Formula I may also be directly synthesized using a chiral catalyst or a chiral ligand, e.g. Mark A. Huffman, et al, J. Org. Chem. 1995, 60, 1590-1594.
  • the compounds of formula I possess HIV protease inhibitory activity and are therefore useful as antiviral agents for the treatment of HIV infection and associated diseases.
  • the compounds of formula I possess HIV protease inhibitory activity and are effective as inhibitors of HIV growth.
  • the ability of the compounds of the present invention to inhibit viral growth or infectivity is demonstrated in standard assay of viral growth or infectivity, for example, using the assay described below.
  • the compounds of formula I of the present invention are also useful for the inhibition of HIV in an ex vivo sample containing HIV or expected to be exposed to HIV.
  • the compounds of the present invention may be used to inhibit HIV present in a body fluid sample (for example, a serum or semen sample) which contains or is suspected to contain or be exposed to HIV.
  • the compounds provided by this invention are also useful as standard or reference compounds for use in tests or assays for determining the ability of an agent to inhibit viral clone replication and/or HIV protease, for example in a pharmaceutical research program.
  • the compounds of the present invention may be used as a control or reference compound in such assays and as a quality control standard.
  • the compounds of the present invention may be provided in a commercial kit or container for use as such standard or reference compound. Since the compounds of the present invention exhibit specificity for HIV protease, the compounds of the present invention may also be useful as diagnostic reagents in diagnostic assays for the detection of HIV protease.
  • ⁇ g denotes microgram
  • mg denotes milligram
  • g denotes gram
  • ⁇ L denotes microliter
  • mL denotes milliliter
  • L denotes liter
  • nM denotes nanomolar
  • ⁇ M denotes micromolar
  • mM denotes millimolar
  • M denotes molar
  • nm denotes nanometer.
  • Sigma stands for the Sigma- Aldrich Corp. of St. Louis, MO.
  • Plasmid pDAB 72 containing both gag and pol sequences of BH10 (bp 1 13-1816) cloned into PTZ 19R was prepared according to Erickson-Viitanen et al. AIDS Research and Human Retroviruses 1989, 5, 577.
  • the plasmid was linearized with Bam HI prior to the generation of in vitro RNA transcripts using the Riboprobe Gemini system II kit (Promega) with T7 RNA polymerase. Synthesized RNA was purified by treatment with RNase free DNAse (Promega), phenol-chloroform extraction, and ethanol precipitation. RNA transcripts were dissolved in water, and stored at -70°C. The concentration of RNA was determined from the A260-
  • Biotinylated capture probes were purified by HPLC after synthesis on an Applied source
  • Biosystems (Foster City, CA) DNA synthesizer by addition of biotin to the 5' teiminal end of the oligonucleotide, using the biotin-phosphoramidite reagent of Cocuzza, Tet. Lett. 1989, 30, 6287.
  • the gag biotinylated capture probe (5-biotin- CTAGCTCCCTGCTTGCCCATACTA 3*) was complementary to nucleotides 889-912 of HXB2 and the pol biotinylated capture probe (5'-biotin -
  • CCCTATCATTTTTGGTTTCCAT 3' was complementary to nucleotides 2374-2395 of HXB2.
  • Alkaline phosphatase conjugated oligonucleotides used as reporter probes were prepared by Syngene (San Diego, CA.).
  • the pol reporter probe (5' CTGTCTTACTTTGATAAAACCTC 3') was complementary to nucleotides 2403-2425 of HXB2.
  • the gag reporter probe (5' CCCAGTATTTGTCTACAGCCTTCT 3') was complementary to nucleotides 950-973 of HXB2.
  • the reporter probes were prepared as 0.5 ⁇ M stocks in 2 x SSC (0.3 M NaCl, 0.03 M sodium citrate), 0.05 M Tris pH 8.8, 1 mg/mL BSA.
  • the biotinylated capture probes were prepared as 100 ⁇ M stocks in water.
  • Streptavidin coated plates were obtained from Du Pont Biotechnology Systems (Boston, MA).
  • MT-2 and MT-4 cells were maintained in RPMI 1640 supplemented with 5% fetal calf serum (FCS) for MT-2 cells or 10% FCS for MT-4 cells, 2 mM L-glutamine and 50 ⁇ g/mL gentamycin, all from Gibco.
  • HIV-1 RF was propagated in MT-4 cells in the same medium. Virus stocks were prepared approximately 10 days after acute infection of MT-4 cells and stored as aliquots at -70°C. Infectious titers of HIV-1 (RF) stocks were 1-3 x 10 ⁇ PFU (plaque forming units)/mL as measured by plaque assay on MT-2 cells (see below). Each aliquot of virus stock used for infection was thawed only once.
  • cells to be infected were subcultured one day prior to infection. On the day of infection, cells were resuspended at 5 x 10 ⁇ cells/mL in RPMI 1640, 5% FCS for bulk infections or at 2 x 10 6 /mL in Dulbecco's modified Eagles medium with 5% FCS for infection in microtiter plates. Virus was added and culture continued for 3 days at 37°C.
  • HIV RNA assav Cell lysates or purified RNA in 3 M or 5 M GED were mixed with 5 M GED and capture probe to a final guanidinium isothiocyanate concentration of 3 M and a final biotin oligonucleotide concentration of 30 nM. Hybridization was carried out in sealed U bottom 96 well tissue culture plates (Nunc or Costar) for 16-20 hours at 37°C. RNA hybridization reactions were diluted three-fold with deionized water to a final guanidinium isothiocyanate concentration of 1 M and aliquots (150 ⁇ L) were transferred to streptavidin coated microtiter plates wells.
  • Binding of capture probe and capture probe-RNA hybrid to the immobilized streptavidin was allowed to proceed for 2 hours at room temperature, after which the plates were washed 6 times with DuPont ELISA plate wash buffer (phosphate buffered saline(PBS), 0.05% Tween 20.)
  • DuPont ELISA plate wash buffer phosphate buffered saline(PBS), 0.05% Tween 20.
  • a second hybridization of reporter probe to the immobilized complex of capture probe and hybridized target RNA was carried out in the washed streptavidin coated well by addition of 120 ⁇ l of a hybridization cocktail containing 4 X SSC, 0.66% Triton X 100, 6.66% deionized formamide, 1 mg/mL BSA and 5 nM reporter probe. After hybridization for one hour at 37 C, the plate was again washed 6 times.
  • Immobilized alkaline phosphatase activity was detected by addition of 100 ⁇ L of 0.2 mM 4-methylumbelliferyl phosphate (MUBP, JBL Scientific) in buffer ⁇ (2.5 M diethanolamine pH 8.9 (JBL Scientific), 10 mM MgCk>, 5 mM zinc acetate dihydrate and 5 mM N-hydroxyethyl-ethylene-diamine-triacetic acid).
  • MUBP 4-methylumbelliferyl phosphate
  • the final volume in each well was 200 ⁇ L. Eight wells per plate were left uninfected with 50 ⁇ L of medium added in place of virus, while eight wells were infected in the absence of any antiviral compound.
  • parallel plates were cultured without virus infection. After 3 days of culture at 37°C in a humidified chamber inside a CO2 incubator, all but 25 ⁇ L of medium/well was removed from the HIV infected plates. Thirty seven ⁇ L of 5 M GED containing biotinylated capture probe was added to the settled cells and remaining medium in each well to a final concentration of 3 M GED and 30 nM capture probe.
  • Hybridization of the capture probe to HIV RNA in the cell lysate was carried out in the same microplate well used for virus culture by sealing the plate with a plate sealer (Costar), and incubating for 16-20 hrs in a 37°C incubator. Distilled water was then added to each well to dilute the hybridization reaction three-fold and 150 ⁇ L of this diluted mixture was transferred to a streptavidin coated microtiter plate. HIV RNA was quantitated as described above. A standard curve, prepared by adding known amounts of pDAB 72 in vitro RNA transcript to wells containing lysed uninfected cells, was run on each microtiter plate in order to determine the amount of viral RNA made during the infection.
  • ddC dideoxycytidine
  • This concentration of virus corresponded to ⁇ 3 x 10 ⁇ PFU (measured by plaque assay on MT-2 cells) per assay well and typically produced approximately 75% of the maximum viral RNA level achievable at any virus inoculum.
  • IC90 values were determined from the percent reduction of net signal (signal from infected cell samples minus signal from uninfected cell samples) in the RNA assay relative to the net signal from infected, untreated cells on the same culture plate (average of eight wells). Valid performance of individual infection and RNA assay tests was judged according to three criteria. It was required that the virus infection should result in an RNA assay signal equal to or greater than the signal generated from 2 ng of pDAB 72 in vitro RNA transcript. The IC90 for ddC, determined in each assay run, should be between 0.1 and 0.3 ⁇ g/mL. Finally, the plateau level of viral RNA produced by an effective protease inhibitor should be less than 10% of the level achieved in an uninhibited infection. A compound was considered active if its IC90 was found to be less than l ⁇ M.
  • the antiviral compounds of this invention can be administered as treatment for viral infections by any means that produces contact of the active agent with the agent's site of action, i.e., the viral protease, in the body of a mammal. They can be administered by any conventional means available for use in conjunction with pharmaceuticals, either as individual therapeutic agents or in a combination of therapeutic agents. They can be administered alone, but preferably are administered with a pharmaceutical carrier selected on the basis of the chosen route of administration and standard pharmaceutical practice.
  • the dosage administered will, of course, vary depending upon known factors, such as the pharmacodynamic characteristics of the particular agent and its mode and route of administration; the age, health and weight of the recipient; the nature and extent of the symptoms; the kind of concurrent treatment; the frequency of treatment; and the effect desired.
  • a daily dosage of active ingredient can be expected to be about 0.001 to about 1000 milligrams per kilogram of body weight, with the preferred dose being about 0.1 to about 30 mg/kg.
  • compositions suitable for administration contain from about 1 mg to about 100 mg of active ingredient per unit.
  • the active ingredient will ordinarily be present in an amount of about 0.5-95% by weight based on the total weight of the composition.
  • the active ingredient can be administered orally in solid dosage forms, such as capsules, tablets and powders, or in liquid dosage forms, such as elixirs, syrups and suspensions. It can also be administered parenterally, in sterile liquid dosage forms.
  • Gelatin capsules contain the active ingredient and powdered carriers, such as lactose, starch, cellulose derivatives, magnesium stearate, stearic acid, and the like. Similar diluents can be used to make compressed tablets. Both tablets and capsules can be manufactured as sustained release products to provide for continuous release of medication over a period of hours. Compressed tablets can be sugar coated or film coated to mask any unpleasant taste and protect the tablet from the atmosphere, or enteric coated for selective disintegration in the gastrointestinal tract. Liquid dosage forms for oral administration can contain coloring and flavoring to increase patient acceptance.
  • water, a suitable oil, saline, aqueous dextrose (glucose), and related sugar solutions and glycols such as propylene glycol or polyethylene glycols are suitable carriers for parenteral solutions.
  • Solutions for parenteral administration preferably contain a water soluble salt of the active ingredient, suitable stabilizing agents, and if necessary, buffer substances.
  • Antioxidizing agents such as sodium bisulfite, sodium sulfite, or ascorbic acid, either alone or combined, are suitable stabilizing agents.
  • citric acid and its salts, and sodium EDTA are also used.
  • parenteral solutions can contain preservatives, such as benzalkonium chloride, methyl- or propyl-paraben and chlorobutanol.
  • Suitable pharmaceutical carriers are described in Remington's Pharmaceutical Sciences, supra, a standard reference text in this field.
  • Useful pharmaceutical dosage-forms for administration of the compounds of this invention can be illustrated as follows:
  • a large number of unit capsules can be prepared by filling standard two-piece hard gelatin capsules each with 100 mg of powdered active ingredient, 150 mg of lactose, 50 mg of cellulose, and 6 mg magnesium stearic.
  • a mixture of active ingredient in a digestible oil such as soybean oil, cottonseed oil or olive oil can be prepared and injected by means of a positive displacement pump into gelatin to form soft gelatin capsules containing 100 mg of the active ingredient. The capsules should then be washed and dried.
  • a digestible oil such as soybean oil, cottonseed oil or olive oil
  • a large number of tablets can be prepared by conventional procedures so that the dosage unit is 100 mg of active ingredient, 0.2 mg of colloidal silicon dioxide, 5 milligrams of magnesium stearate, 275 mg of microcrystalline cellulose, 11 mg of starch and 98.8 mg of lactose.
  • Appropriate coatings may be applied to increase palatability or delay absorption.
  • aqueous suspension can be prepared for oral administration so that each 5 mL contain 25 mg of finely divided active ingredient, 200 mg of sodium carboxymethyl cellulose, 5 mg of sodium benzoate, 1.0 g of sorbitol solution, U.S. P., and 0.025 mg of vanillin.
  • a parenteral composition suitable for administration by injection can be prepared by stirring 1.5% by weight of active ingredient in 10% by volume propylene glycol and water. The solution is sterilized by commonly used techniques.
  • Each therapeutic agent component of this invention can independently be in any dosage fo ⁇ n, such as those described above, and can also be administered in various ways, as described above.
  • component (b) is to be understood to represent one or more agents as described previously. Thus, if components (a) and (b) are to be treated the same or independently, each agent of component (b) may also be treated the same or independently.
  • Components (a) and (b) of the present invention may be formulated together, in a single dosage unit (that is, combined together in one capsule, tablet, powder, or liquid, etc.) as a combination product.
  • the component (a) may be administered at the same time as component (b) or in any order; for example component (a) of this invention may be administered first, followed by administration of component (b), or they may be administered in the revserse order.
  • component (b) contains more that one agent, e.g., one RT inhibitor and one protease inhibitor, these agents may be administered together or in any order.
  • component (a) and (b) When not administered at the same time, preferably the administration of component (a) and (b) occurs less than about one hour apart.
  • the route of administration of component (a) and (b) is oral.
  • component (a) and component (b) both be administered by the same route (that is, for example, both orally) or dosage form, if desired, they may each be administered by different routes (that is, for example, one component of the combination product may be administered orally, and another component may be administered intravenously) or dosage forms.
  • the dosage of the combination therapy of the invention may vary depending upon various factors such as the pharmacodynamic characteristics of the particular agent and its mode and route of administration, the age, health and weight of the recipient, the nature and extent of the symptoms, the kind of concurrent treatment, the frequency of treatment, and the effect desired, as described above.
  • a daily dosage may be about 100 milligrams to about 1.5 grams of each component. If component (b) represents more than one compound, then typically a daily dosage may be about 100 milligrams to about 1.5 grams of each agent of component (b).
  • the dosage amount of each component may be reduced by about 70-80% relative to the usual dosage of the component when it is administered alone as a single agent for the treatment of HIV infection, in view of the synergistic effect of the combination.
  • the combination products of this invention may be formulated such that, although the active ingredients are combined in a single dosage unit, the physical contact between the active ingredients is minimized.
  • one active ingredient may be enteric coated.
  • enteric coating one of the active ingredients it is possible not only to minimize the contact between the combined active ingredients, but also, it is possible to control the release of one of these components in the gastrointestinal tract such that one of these components is not released in the stomach but rather is released in the intestines.
  • Another embodiment of this invention where oral administration is desired provides for a combination product wherein one of the active ingredients is coated with a sustained-release material which effects a sustained-release throughout the gastrointestinal tract and also serves to minimize physical contact between the combined active ingredients.
  • the sustained-released component can be additionally enteric coated such that the release of this component occurs only in the intestine.
  • Still another approach would involve the formulation of a combination product in which the one component is coated with a sustained and/or enteric release polymer, and the other component is also coated with a polymer such as a lowviscosity grade of hydroxypropyl methylcellulose or other appropriate materials as known in the art, in order to further separate the active components.
  • the polymer coating serves to form an additional barrier to interaction with the other component.
  • contact may also be prevented between the individual agents of component (b).
  • Dosage forms of the combination products of the present invention wherein one active ingredient is enteric coated can be in the form of tablets such that the enteric coated component and the other active ingredient are blended together and then compressed into a tablet or such that the enteric coated component is compressed into one tablet layer and the other active ingredient is compressed into an additional layer.
  • one or more placebo layers may be present such that the placebo layer is between the layers of active ingredients.
  • dosage forms of the present invention can be in the form of capsules wherein one active ingredient is compressed into a tablet or in the form of a plurality of microtablets, particles, granules or non-perils, which are then enteric coated. These enteric coated microtablets, particles, granules or non-perils are then placed into a capsule or compressed into a capsule along with a granulation of the other active ingredient.
  • kits useful for the treatment of HIV infection which comprise a therapeutically effective amount of a pharmaceutical composition comprising a compound of component (a) and one or more compounds of component (b), in one or more sterile containers, are also within the ambit of the present invention. Sterilization of the container may be carried out using conventional sterilization methodology well known to those skilled in the art.
  • Component (a) and component (b) may be in the same sterile container or in separate sterile containers.
  • the sterile containers of materials may comprise separate containers, or one or more multi-part containers, as desired.
  • Component (a) and component (b) may be separate, or physically combined into a single dosage form or unit as described above.
  • kits may further include, if desired, one or more of various conventional pharmaceutical kit components, such as for example, one or more pharmaceutically acceptable carriers, additional vials for mixing the components, etc., as will be readily apparent to those skilled in the art.
  • kit components such as for example, one or more pharmaceutically acceptable carriers, additional vials for mixing the components, etc.
  • Instructions, either as inserts or as labels, indicating quantities of the components to be administered, guidelines for administration, and/or guidelines for mixing the components, may also be included in the kit.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Animal Behavior & Ethology (AREA)
  • Virology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • AIDS & HIV (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)

Abstract

This invention relates generally to bis-amino acid sulfonamides containing substituted benzyl amines of formula (I) or stereoisomeric forms, stereoisomeric mixtures, or pharmaceutically acceptable salt forms thereof, which are useful as HIV protease inhibitors, pharmaceutical compositions and diagnostic kits comprising the same, methods of using the same for treating viral infection or as assay standards or reagents, and intermediates and processes for making the same.

Description

TITLE
BIS-AMINO ACID SULFONAMIDES CONTAINING N-TERMINALLY A SUBSTITUTED BENZYL GROUP AS HIV PROTEASE INHIBITORS
FIELD OF THE INVENTION This invention relates generally to bis-amino acid sulfonamides containing substituted benzyl amines useful as HIN protease inhibitors, pharmaceutical compositions and diagnostic kits comprising the same, and methods of using the same for treating viral infection or as assay standards or reagents.
BACKGROUND OF THE INVENTION Two distinct retro viruses, human immunodeficiency virus (HIV) type-1 (HIV-1) or type-2 (HIV-2), have been etiologically linked to the immunosuppressive disease, acquired immunodeficiency syndrome (AIDS). HIV seropositive individuals are initially asymptomatic but typically develop AIDS related complex (ARC) followed by AIDS. Affected individuals exhibit severe immunosuppression which predisposes them to debilitating and ultimately fatal opportunistic infections.
The disease AIDS is the end result of an HIV-1 or HIV-2 virus following its own complex life cycle. The virion life cycle begins with the virion attaching itself to the host human T-4 lymphocyte immune cell through the bonding of a glycoprotein on the surface of the virion's protective coat with the CD4 glycoprotein on the lymphocyte cell. Once attached, the virion sheds its glycoprotein coat, penetrates into the membrane of the host cell, and uncoats its RNA. The virion enzyme, reverse transcriptase, directs the process of transcribing the RNA into single-stranded DNA. The viral RNA is degraded and a second DNA strand is created. The now double-stranded DNA is integrated into the human cell's genes and those genes are used for virus reproduction.
At this point, RNA polymerase transcribes the integrated DNA into viral RNA. The viral RNA is translated into the precursor gag-pol fusion polyprotein. The polyprotein is then cleaved by the HIV protease enzyme to yield the mature viral proteins. Thus, HIV protease is responsible for regulating a cascade of cleavage events that lead to the virus particle's maturing into a virus that is capable of full infectivity.
The typical human immune system response, killing the invading virion, is taxed because the virus infects and kills the immune system's T cells. In addition, viral reverse transcriptase, the enzyme used in making a new virion particle, is not very specific, and causes transcription mistakes that result in continually changed glycoproteins on the surface of the viral protective coat. This lack of specificity decreases the immune system's effectiveness because antibodies specifically produced against one glycoprotein may be useless against another, hence reducing the number of antibodies available to fight the virus. The virus continues to reproduce while the immune response system continues to weaken. Eventually, the HIV largely holds free reign over the body's immune system, allowing opportunistic infections to set in and without the administration of antiviral agents, immunomodulators, or both, death may result.
There are at least three critical points in the virus's life cycle which have been identified as possible targets for antiviral drugs: (1) the initial attachment of the virion to the T-4 lymphocyte or macrophage site, (2) the transcription of viral RNA to viral DNA (reverse transcriptase, RT), and (3) the processing of gag-pol protein by HIV protease. The genomes of retro viruses encode a protease that is responsible for the proteolytic processing of one or more polyprotein precursors such as thepol and gag gene products. See Wellink, Arch. Virol. 98 1 (1988). Retroviral proteases most commonly process the gag precursor into the core proteins, and also process thepol precursor into reverse transcriptase and retroviral protease. The correct processing of the precursor polyproteins by the retroviral protease is necessary for the assembly of the infectious virions. It has been shown that in vitro mutagenesis that produces protease-defective virus leads to the production of immature core forms which lack infectivity. See Crawford et al., J. Virol. 53 899 (1985); Katoh et al., Virology 145 280 (1985). Therefore, retroviral protease inhibition provides an attractive target for antiviral therapy. See Mitsuya, Nature 325 775 ( 1987).
As evidenced by the protease inhibitors presently marketed and in clinical trials, a wide variety of compounds have been studied as potential HIV protease inhibitors. One core, hydroxyethylamino-sulfonamides, has received significant attention. For example, PCT Applications WO94/05639, WO94/04492, WO95/06030, and WO96/28464 generically describe sulfonamides of the formula:
Figure imgf000004_0001
and methods of preparing them. Though some of the present compounds appear to fall within the generic descriptions of some of the above publications, they are not specifically disclosed, suggested, or claimed therein. Even with the current success of protease inhibitors, it has been found that HIV patients can become resistant to a single protease inhibitor. Thus, it is desirable to develop additional protease inhibitors to further combat HIV infection. SUMMARY OF THE INVENTION Accordingly, one object of the present invention is to provide novel protease inhibitors.
It is another object of the present invention to provide a novel method for treating HIV infection which comprises administering to a host in need of such treatment a therapeutically effective amount of at least one of the compounds of the present invention or a pharmaceutically acceptable salt form thereof.
It is another object of the present invention to provide a novel method for treating HIV infection which comprises administering to a host in need thereof a therapeutically effective combination of (a) one of the compounds of the present invention and (b) one or more compounds selected form the group consisting of HIV reverse transcriptase inhibitors and HIV protease inhibitors.
It is another object of the present invention to provide pharmaceutical compositions with protease inhibiting activity comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of at least one of the compounds of the present invention or a pharmaceutically acceptable salt form thereof.
It is another object of the present invention to provide a method of inhibiting HIV present in a body fluid sample which comprises treating the body fluid sample with an effective amount of a compound of the present invention. It is another object of the present invention to provide a kit or container containing at least one of the compounds of the present invention in an amount effective for use as a standard or reagent in a test or assay for determining the ability of a potential pharmaceutical to inhibit HIV protease, HIV growth, or both.
These and other objects, which will become apparent during the following detailed description, have been achieved by the inventors' discovery that compounds of formula (I):
Figure imgf000005_0001
I wherein R1, R2, and R3 are defined below, stereoisomeric forms, mixtures of stereoisomeric forms, or pharmaceutically acceptable salt forms thereof, are effective protease inhibitors. DETAILED DESCRIPTION OF PREFERRED EMBODIMENTS [1] Thus, in a first embodiment, the present invention provides a novel compound of Formula I:
Figure imgf000006_0001
I or a pharmaceutically acceptable salt form thereof, wherein:
R1 is F;
R2 is F or H; and,
R3 is selected from the group: 4-aminophenyl, 3-aminophenyl, 2,3-dihydrobenzofuran-5- yl, and 1 ,3-benzodioxol-5-yl.
[2] In a preferred embodiment, the present invention provides a novel compound of Formula II:
Figure imgf000006_0002
II.
[3] In a more preferred embodiment, the present invention provides a novel compound of Formula Ila:
Figure imgf000006_0003
Ila. [4] In an even more preferred embodiment, the present invention provides a novel compound of Formula Ila, wherein:
R3 is 3-aminophenyl.
[5] In another even more preferred embodiment, the present invention provides a novel compound of Formula Ila, wherein:
R3 is 4-aminophenyl.
[6] In another even more preferred embodiment, the present invention provides a novel compound of Formula Ila, wherein:
R3 is 2,3-dihydrobenzofuran-5-yl or l,3-benzodioxol-5-yl.
[7] In another more preferred embodiment, the present invention provides a novel compound of Formula lib:
Figure imgf000007_0001
lib.
[8] In another even more preferred embodiment, the present invention provides a novel compound of Formula lib, wherein:
R3 is 3-aminophenyl. [9] In another even more preferred embodiment, the present invention provides a novel compound of Formula lib, wherein:
R3 is 4-aminophenyl.
[10] In another even more preferred embodiment, the present invention provides a novel compound of Formula lib, wherein:
R3 is 2,3-dihydrobenzofuran-5-yl or l,3-benzodioxol-5-yl.
[1 1] In another more preferred embodiment, the present invention provides a novel compound of Formula lie:
He.
[ 12] In another even more preferred embodiment, the present invention provides a novel compound of Formula lie, wherein:
R3 is 3-aminophenyl.
[ 13] In another even more preferred embodiment, the present invention provides a novel compound of Formula lie, wherein:
R3 is 4-aminophenyl.
[14] In another even more preferred embodiment, the present invention provides a novel compound of Formula lie, wherein: R3 is 2,3-dihydrobenzofuran-5-yl or 1 ,3-benzodioxol-5-yl.
[15] In another preferred embodiment, the present invention provides a novel compound of Formula III:
Figure imgf000009_0001
III.
[16] In another more preferred embodiment, the present invention provides a novel compound of Formula Ilia:
Figure imgf000009_0002
Ilia.
[17] In another preferred embodiment, the present invention provides a novel compound of Formula IV:
Figure imgf000009_0003
IV.
[ 18] In another more preferred embodiment, the present invention provides a novel compound of Formula IVa:
Figure imgf000010_0001
IVa.
In another embodiment, the present invention provides a novel pharmaceutical composition comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of a compound of formula I or pharmaceutically acceptable salt form thereof.
In another embodiment, the present invention provides a novel method for treating HIV infection which comprises administering to a host in need of such treatment a therapeutically effective amount of a compound of formula I or pharmaceutically acceptable salt form thereof.
In another embodiment, the present invention provides a novel method of treating HIV infection which comprises administering, in combination, to a host in need thereof a therapeutically effective amount of: (a) a compound of formula I; and,
(b) at least one compound selected from the group consisting of HIV reverse transcriptase inhibitors and HIV protease inhibitors.
In another preferred embodiment, the reverse transcriptase inhibitor is selected from the group AZT, ddC, ddl, d4T, 3TC, delavirdine, efavirenz, nevirapine, Ro 18,893, trovirdine, MKC-442, HBY 097, ACT, UC-781, UC-782, RD4-2025, and MEN 10979, and the protease inhibitor is selected from the group saquinavir, ritonavir, indinavir, amprenavir, nelfinavir, palinavir, BMS-232623, GS3333, KNI-413, KNI-272, LG-71350, CGP-61755, PD 173606, PD 177298, PD 178390, PD 178392, U- 140690, and ABT-378. In an even more preferred embodiment, the reverse transcriptase inhibitor is selected from the group AZT, efavirenz, and 3TC and the protease inhibitor is selected from the group saquinavir, ritonavir, nelfinavir, and indinavir.
In a still further preferred ebodiment, the reverse transcriptase inhibitor is AZT.
In another still further preferred embodiment, the protease inhibitor is ritonavir.
In another preferred embodiment, component (b) is a HIV reverse transcriptase inhibitor and a HIV protease inhibitor.
In another preferred embodiment, component (b) is two different HIV reverse transcriptase inhibitors.
In another embodiment, the present invention provides a pharmaceutical composition useful for the treatment of HIV infection, which comprises a therapeutically effective amount of:
(a) a compound of formula I; and,
(b) at least one compound selected from the group consisting of HIV reverse transcriptase inhibitors and HIV protease inhibitors, in one or more sterile containers.
In another embodiment, the present invention provides a novel method of inhibiting HIV present in a body fluid sample which comprises treating the body fluid sample with an effective amount of a compound of formula I.
In a ninth embodiment, the present invention to provides a novel a kit or container comprising a compound of formula I in an amount effective for use as a standard or reagent in a test or assay for determining the ability of a potential pharmaceutical to inhibit HIV protease, HIV growth, or both. DEFINITIONS As used herein, the following terms and expressions have the indicated meanings. It will be appreciated that the compounds of the present invention contain asymmetrically substituted carbon atoms, and may be isolated in optically active or racemic forms. It is well known in the art how to prepare optically active forms, such as by resolution of racemic forms or by synthesis, from optically active starting materials. All chiral, diastereomeric, racemic forms and all geometric isomeric forms of a structure are intended, unless the specific stereochemistry or isomer form is specifically indicated.
The processes of the present invention are contemplated to be practiced on at least a multigram scale, kilogram scale, multikilogram scale, or industrial scale. Multigram scale, as used herein, is preferably the scale wherein at least one starting material is present in 10 grams or more, more preferably at least 50 grams or more, even more preferably at least 100 grams or more. Multikilogram scale, as used herein, is intended to mean the scale wherein more than one kilogram of at least one starting material is used. Industrial scale as used herein is intended to mean a scale which is other than a laboratory scale and which is sufficient to supply product sufficient for either clinical tests or distribution to consumers.
The present invention is intended to include all isotopes of atoms occurring on the present compounds. Isotopes include those atoms having the same atomic number but different mass numbers. By way of general example and without limitation, isotopes of hydrogen include tritium and deuterium. Isotopes of carbon include C-13 and C-14.
As used herein, "HIV reverse transcriptase inhibitor" is intended to refer to both nucleoside and non-nucleoside inhibitors of HIV reverse transcriptase (RT). Examples of nucleoside RT inhibitors include, but are not limited to, AZT, ddC, ddl, d4T, and 3TC. Examples of non-nucleoside RT inhibitors include, but are not limited to, delavirdine (Pharmacia and Upjohn, U90152S), efavirenz (DuPont), nevirapine (Boehringer Ingelheim), Ro 18,893 (Roche), trovirdine (Lilly), MKC-442 (Triangle), HBY 097 (Hoechst), HBY 1293 (Hoechst), ACT (Korean Research Institute), UC-781 (Rega Institute), UC-782 (Rega Institute), RD4-2025 (Tosoh Co. Ltd.), and MEN 10979 (Menarini Farmaceutici).
As used herein, "HIV protease inhibitor" is intended to refer to compounds which inhibit HIV protease. Examples include, but are not limited, saquinavir (Roche, Ro31-8959), ritonavir (Abbott, ABT-538), indinavir (Merck, MK-639), amprenavir (Vertex/Glaxo Wellcome), nelfinavir (Agouron, AG-1343), palinavir (Boehringer Ingelheim), BMS-232623 (Bristol-Myers Squibb), GS3333 (Gilead Sciences), KNI-413 (Japan Energy), KNI-272 (Japan Energy), LG-71350 (LG Chemical), CGP-61755 (Ciba-Geigy), PD 173606 (Parke Davis), PD 177298 (Parke Davis), PD 178390 (Parke Davis), PD 178392 (Parke Davis), tipranavir (Pharmacia and Upjohn, U- 140690), DMP- 450 (DuPont) and ABT-378.
As used herein, "pharmaceutically acceptable salts" refer to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. The pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. For example, such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, and the like.
The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, PA, 1985, p. 1418, the disclosure of which is hereby incorporated by reference. The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication commensurate with a reasonable benefit/risk ratio. "Prodrugs" are intended to include any covalently bonded carriers which release the active parent drug according to formula (I) or other formulas or compounds of the present invention in vivo when such prodrug is administered to a mammalian subject. Prodrugs of a compound of the present invention, for example formula (I), are prepared by modifying functional groups present in the compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound. Prodrugs include compounds of the present invention wherein the hydroxy or amino group is bonded to any group that, when the prodrug is administered to a mammalian subject, cleaves to form a free hydroxyl or free amino, respectively. Examples of prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of alcohol and amine functional groups in the compounds of the present invention, and the like.
"Stable compound" and "stable structure" are meant to indicate a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent. Only stable compounds are contempleted by the present invention.
"Substituted" is intended to indicate that one or more hydrogens on the atom indicated in the expression using "substituted" is replaced with a selection from the indicated group(s), provided that the indicated atom's normal valency is not exceeded, and that the substitution results in a stable compound. When a substituent is keto (i.e., =O) group, then 2 hydrogens on the atom are replaced.
"Therapeutically effective amount" is intended to include an amount of a compound of the present invention or an amount of the combination of compounds claimed effective to inhibit HIV infection or treat the symptoms of HIV infection in a host. The combination of compounds is preferably a synergistic combination. Synergy, as described for example by Chou and Talalay, Adv. Enzyme Regul. 22:27-55 (1984), occurs when the effect (in this case, inhibition of HIV replication) of the compounds when administered in combination is greater than the additive effect of the compounds when administered alone as a single agent. In general, a synergistic effect is most clearly demonstrated at suboptimal concentrations of the compounds. Synergy can be in terms of lower cytotoxicity, increased antiviral effect, or some other beneficial effect of the combination compared with the individual components.
One diastereomer of a compound of Formula I may display superior activity compared with the other. When required, separation of the racemic material can be achieved by HPLC using a chiral column or by a resolution using a resolving agent such as camphonic chloride as in Thomas J. Tucker, et al, J. Med. Chem. 1994, 37, 2437-2444. A chiral compound of Formula I may also be directly synthesized using a chiral catalyst or a chiral ligand, e.g. Mark A. Huffman, et al, J. Org. Chem. 1995, 60, 1590-1594.
Other features of the invention will become apparent in the course of the following descriptions of exemplary embodiments which are given for illustration of the invention and are not intended to be limiting thereof.
Examples Abbreviations used in the Examples are defined as follows: "°C" for degrees Celsius, "d" for doublet, "dd" for doublet of doublets, "eq" for equivalent or equivalents, "g" for gram or grams, "mg" for milligram or milligrams, "mL" for milliliter or milliliters, "H" for hydrogen or hydrogens, "hr" for hour or hours, "m" for multiplet, "M" for molar, "min" for minute or minutes, "MHz" for megahertz, "MS" for mass spectroscopy, "nmr" or "NMR" for nuclear magnetic resonance spectroscopy, "t" for triplet, and "TLC" for thin layer chromatography.
Example 1
Figure imgf000015_0001
1B
1A
Figure imgf000015_0002
1G
Figure imgf000016_0001
IB To a solution of N-[3(S)-[N,N-bis(phenylmethyl)amino]-2(R)-hydroxy-4- phenylbutyl]-N-isobutylamine#oxalic acid salt 1A (127.6 g, 251 mmol) in toluene (1 L), water (500 mL) and CH2C12 (400 mL) was added NaOH (50% aqueous, 44.5 g). After stirring 10 min the reaction mixture was extracted with toluene. The combined organic layers were washed with brine, dried (MgSO4) and the solvent was removed under reduced pressure. The residue was taken up in THF (1 L), cooled to 0 °C, and was treated with triethylamine (28.15 g, 278 mmol) and di-tert-butyl dicarbonate (55.23 g, 253 mmol). The solution was warmed to room temperature and was stirred overnight. The solvent was removed under reduced pressure and the residue was taken up in EtOAc (1 L), washed with water, 5% citric acid, water, saturated NaHCO3, brine, and was dried (MgSO4). The solvent was removed under reduced pressure to give the carbamate IB which was used directly without further purification. CIMS (NH3) m/z: 517 (M+H+, 100%)
IC To a solution of crude IB (251 mmol possible) in methanol (500 mL) was added palladium hydroxide on carbon (20%, 10 g). The suspension was placed in a parr bottle and was charged with hydrogen (55 psi). After shaking overnight the reaction mixture was filtered through Celite and the solvent was removed under reduced pressure. The resulting solid was recrystallized (EtOAc/hexane) to give the amine IC as a white solid (56.6 g, 67% (2 steps)): CIMS (NH3) m/z: 337 (M+H+, 100%) ID To a solution of N-carbobenzyloxy-L-tert-leucine (47.5 g, 179 mmol) in DMF (250 mL) at 0 °C was added N-hydroxybenzotriazole (38.6 g, 285 mmol) and EDC (35.7 g, 186 mmol). After stirring 1.5 hours the solution was added to a suspension of IC (56.6 g, 167 mmol) and 4-methylmorpholine (52.9 g, 521 mmol) in DMF (200 mL). The reaction mixture was allowed to warm to room temperature. After stirring overnight N,N- dimethylethylenediamine (4 mL) was added, the solution was stirred 1.5 hours and the solvent was removed under reduced pressure. The residue was taken up in EtOAc (1 L), washed with water, 5% citric acid, water, saturated NaHCO3, brine, and was dried (MgSO ). The solvent was removed under reduced pressure to give ID (97.5 g, 100%) which was used without further purification. CIMS (NH3) m/z: 584 (M+H+, 100%)
IE To a solution of ID (97.5 g, 167 mmol) in methanol (300 mL) was added palladium hydroxide on carbon (20%, 10 g). The suspension was placed in a parr bottle and was charged with hydrogen (55psi). After shaking overnight the reaction mixture was filtered through Celite and the solvent was removed under reduced pressure. The resulting solid was recrystallized (EtOAc/hexane) to give the amine IE as a white solid (72.8 g, 97%): CIMS (NH3) m/z: 450 (M+H+ 100%)
IF To a solution of amine IE (43.8 g, 97.6 mmol) in EtOAc (400 mL) and water (270 mL) was added KHCO3 (27.7 g, 276 mmol) and cloroacetyl chloride (12.4 g, 111 mmol). After stirring 3 hours, EtOAc (1 L) was added and the solution was washed with water, 5% citric acid, water, saturated NaHCO3, brine, and was dried (MgSO4). The solvent was removed under reduced pressure to give IF as a white solid (51.0 g, 99%): CIMS (NH3) m/z: 526 (M+H+ 100%)
1G To a solution of IF (33.8 g, 64.2 mmol) in EtOAc (600 mL) was added 4N HC1 in dioxane (80 mL, 320 mmol) and the reaction mixture was stirred 6 hours. The solvent was removed under reduced pressure and the resulting solid was triturated with cold ether to give the hydrochloride salt 1G (28.75 g, 97%): CIMS (NH3) m/z: 426 (M+H+, 100%)
1H To a solution of the salt 1G (32.0 g, 69.2 mmol) in THF (350 mL) and water (450 mL) was added K2CO3 (56.7 g, 411 mmol) and 4-nitrobenzenesulfonyl chloride (16.9 g, 76.0 mmol). After stirring 4 hours, water was added and the suspension was extracted with EtOAc. The combined organic layers were washed with brine, 5% citric acid, water, saturated NaHCO3, brine, and was dried (MgSO4). The solvent was removed under reduced pressure and the resulting solid was recrystallized (EtOAc/hexane) to give the sulfonamide 1H as a white solid (35.8 g, 85%). CIMS (NH3) m/z: 61 1 (M+H+, 100%). II To a solution of the chloride 1H (16.0 g, 26.1 mmol) in THF (200 mL) was added 3- fluorobenzylamine (20.0 g, 160 mmol) and the reaction mixture was refluxed overnight. The solvent was removed under reduced pressure and the residue was taken up in EtOAc and was washed with water, brine, and dried (MgSO4). The solvent was removed under reduced pressure and the residue was chromatographed (silica gel, 4% methanol/CH2Cl2) to give the amine II as a white solid (16.3 g, 89%). CIMS (NH3) m/z: 700 (M+H+, 100%).
1 To a solution of II (14.6 g, 20.8 mmol) in methanol (500 mL) was added palladium hydroxide on carbon (20%, 1.5 g) and the reaction mixture was charged with hydrogen. After stirring 3 hours, the mixture was filtered through Celite and the solvent was removed under reduced pressure. The residue was chromatographed (silica gel, 5% methanol/CH2Cl ) to give the amine as a white solid (13.2 g, 95%). To a solution of the freebase (1 1.68 g, 17.4 mmol) in ether (300 mL) and EtOAc (100 mL) was added IN HC1 in ether (37 mL, 37 mmol). The resulting suspension was stirred 15 min and was filtered to give the bis-hydrochloride salt 1 as a white solid (12.5 g, 96%): CIMS (NH3) m/z: 670
(M+H+, 100%).
Example 2
Figure imgf000018_0001
2A
Figure imgf000018_0002
2A To a solution of the chloride 1H (16.0 g, 26.1 mmol) in THF (200 mL) was added 3,5-difluorobenzylamine (25.0 g, 174 mmol) and the reaction mixture was refluxed overnight. The solvent was removed under reduced pressure and the residue was taken up in EtOAc and was washed with water, brine, and dried (MgSO4). The solvent was removed under reduced pressure and the residue was chromatographed (silica gel, 4% methanol/CH2Cl2) to give the amine 2A as a white solid (15.2 g, 81%). CIMS (NH3) m/z: 718 (M+H+, 100%).
2 To a solution of 2A (15.2 g, 21.2 mmol) in methanol (500 mL) was added palladium hydroxide on carbon (20%, 1.5 g) and the reaction mixture was charged with hydrogen. After stirring 4 hours, the mixture was filtered through Celite and the solvent was removed under reduced pressure. The residue was chromatographed (silica gel, 5% methanol/CH2Cl2) to give the amine as a white solid (10.3 g, 71%). To a solution of the freebase in ether (300 mL) and EtOAc (100 mL) was added IN HC1 in ether (32 mL, 32 mmol). The resulting suspension was stirred 15 min and was filtered to give the bis- hydrochloride salt 2 as a white solid: CIMS (NH3) m z: 688 (M+H+ 100%).
Example 3
Figure imgf000019_0001
3A To a solution of the chloride 1H (300 mg, 0.49 mmol) in THF (4 mL) was added 2,5- difluorobenzylamine (1.2 g, 8.5 mmol) and the reaction mixture was refluxed 4 hours. The reaction mixture was diluted with EtOAc and was washed with water, brine, and dried (MgSO4). The solvent was removed under reduced pressure and the residue was chromatographed (silica gel, 5% methanol/CH2Cl2) to give the amine 3A as a white solid (270 mg, 77%). CIMS (NH3) m/z: 718 (M+H+, 100%)
3 To a solution of 3A (260 mg, 0.36 mmol) in methanol (25 mL) was added palladium hydroxide on carbon (20%, 50 mg) and the reaction mixture was charged with hydrogen. After stirring 1 hour, the mixture was filtered through Celite and the solvent was removed under reduced pressure. The residue was chromatographed (silica gel, 5% methanol/CH2Cl2) to give the amine as a white solid (226 mg, 91%). To a solution of the freebase in ether (30 mL) and EtOAc (10 mL) was added 4N HC1 in dioxane (0.2 mL, 0.8 mmol). The resulting suspension was stirred 15 min and was filtered to give the bis- hydrochloride salt 3 as a white solid: CIMS (NH3) m/z: 688 (M+H+ 100%).
Example 4
Figure imgf000020_0001
4A To a solution of the chloride IH (300 mg, 0.49 mmol) in THF (4 mL) was added 2,6- difluorobenzylamine (1.2 g, 8.5 mmol) and the reaction mixture was refluxed 4 hours. The reaction mixture was diluted with EtOAc and was washed with water, brine, and dried (MgSO4). The solvent was removed under reduced pressure and the residue was chromatographed (silica gel, 5% methanol/CH2Cl2) to give the amine 4A as a white solid (306 mg, 87%). CIMS (NH3) m/z: 718 (M+H+, 100%).
4 To a solution of 4A (295 mg, 0.41 mmol) in methanol (25 mL) was added palladium hydroxide on carbon (20%, 50 mg) and the reaction mixture was charged with hydrogen. After stirring 1 hour, the mixture was filtered through Celite and the solvent was removed under reduced pressure. The residue was chromatographed (silica gel, 5% methanol/CH2Cl2) to give the amine as a white solid (228 mg, 81%). To a solution of the freebase in ether (30 mL) and EtOAc (10 mL) was added 4N HC1 in dioxane (0.2 mL, 0.8 mmol). The resulting suspension was stirred 15 min and was filtered to give the bis- hydrochloride salt 4 as a white solid: CIMS (NH3) m/z: 688 (M+H+, 100%). Example 5
Figure imgf000021_0001
5B
Figure imgf000021_0002
5A To a solution of the salt 1G (28.8 g, 62.1 mmol) in THF (300 mL) and water (400 mL) was added K2CO3 (51.4 g, 370 mmol) and 3-nitrobenzenesulfonyl chloride (15.14 g, 68.3 mmol). After stirring 4 hours, water was added and the suspension was extracted with EtOAc. The combined organic layers were washed with brine, 5% citric acid, water, saturated NaHCO3, brine, and was dried (MgSO4). The solvent was removed under reduced pressure and the resulting solid was triturated with EtOAc and hexane to give the sulfonamide 5A as a white solid (32.1 g, 85%). CIMS (NH3) m/z: 611 (M+H+, 100%).
5B To a solution of the chloride 5A (16.0 g, 26.1 mmol) in THF (200 mL) was added 3- fluorobenzylamine (17.0 g, 135 mmol) and the reaction mixture was refluxed overnight. The solvent was removed under reduced pressure and the residue was taken up in EtOAc and was washed with water, brine, and dried (MgSO4). The solvent was removed under reduced pressure and the residue was chromatographed (silica gel, 4% methanol/ CH2C12) to give the amine 5B as a white solid (16.0 g, 87%). CIMS (NH3) m/z: 700 (M+H+, 100%). 5 To a solution of 5B (12.0 g, 17.22 mmol) in methanol (400 mL) was added palladium hydroxide on carbon (20%, 1.25 g) and the reaction mixture was charged with hydrogen. After stirring 3 hours, the mixture was filtered through Celite and the solvent was removed under reduced pressure. The residue was chromatographed (silica gel, 5% methanol/CH2Cl2) to give the amine as a white solid (11.2 g, 97%). To a solution of the freebase in ether (400 mL) and EtOAc (75 mL) was added IN HC1 in ether (36 mL, 36 mmol). The resulting suspension was stirred 15 min and was filtered to give the bis- hydrochloride salt 5 as a white solid. CIMS (NH3) m/z: 670 (M+H+, 100%).
Example 6
Figure imgf000022_0001
6A To a solution of the chloride 5 A (16.0 g, 26.1 mmol) in THF (200 mL) was added 3,5-difluorobenzylamine (25.0 g, 174 mmol) and the reaction mixture was stirred 2 hours and refluxed overnight. The solvent was removed under reduced pressure and the residue was taken up in EtOAc and was washed with water, brine, and dried (MgSO4). The solvent was removed under reduced pressure and the residue was chromatographed (silica gel, 3.5% methanol/CH2Cl2) to give the amine 6A as a white solid (15.6 g, 83%). CIMS (NH3) m/z: 718 (M+H+, 100%).
6 To a solution of 6A (14.4 g, 20.0 mmol) in methanol (500 mL) was added palladium hydroxide on carbon (20%, 1.5 g) and the reaction mixture was charged with hydrogen. After stirring 4 hours, the mixture was filtered through Celite and the solvent was removed under reduced pressure. The residue was chromatographed (silica gel, 5% methanol/CH2Cl2) to give the amine as a white solid (12.5 g, 91%). To a solution of the freebase (9.57 g, 13.9 mmol) in ether (300 mL) was added IN HC1 in ether (31 mL, 31 mmol). The resulting suspension was stirred 20 min and was filtered to give the bis- hydrochloride salt 6 as a white solid: (9.9 g, 94%). CIMS (NH3) m/z: 688 (M+H+, 100%).
Example 7
Figure imgf000023_0001
p96 7A
Figure imgf000023_0002
7 To a solution of N-[2R-hydroxy-3-[[(2,3-dihydro2,3-dihydrobenzofuran-5- yl)sulfonyl](2-methylpropyl)amino]-lS-(phenylmethyl)propyl]-2S-[(chloroacetyl)amino]- 3,3-dimethylbutanamide 7A (100 mg, 0.16 mmol) in THF (2 mL) was added 3- fluorobenzylamine (550 mg, 4.4 mmol) and the reaction mixture was refluxed 6 hours. The reaction mixture was diluted with EtOAc and was washed with water (4x), brine, and dried (MgSO4). The solvent was removed under reduced pressure and the residue was chromatographed (silica gel, 5% methanol/CH2Cl2) to give the amine as a white solid (91 mg, 79%). To a solution of the freebase (91 mg, 0.13 mmol) in ether (25 mL) was added 4N HCI in dioxane (0.05 mL, 0.20 mmol). After stirring 10 min the solvent was removed under reduced pressure and the resulting solid was triturated with ether and filtered to give the hydrochloride salt 7 as a white solid (72 mg, 65%): CIMS (NH3) m/z: 697 (M+H+,
100%). Example 8
Figure imgf000024_0001
8 To a solution of 7A (100 mg, 0.16 mmol) in THF (2 mL) was added 3,5- difluorobenzylamine (605 mg, 4.2 mmol) and the reaction mixture was refluxed 6 hours. The reaction mixture was diluted with EtOAc and was washed with water (4x), brine, and dried (MgSO4). The solvent was removed under reduced pressure and the residue was chromatographed (silica gel, 5% methanol/CH2Cl2) to give the amine as a white solid (83 mg, 70%). To a solution of the freebase (83 mg, 0.1 1 mmol) in ether (25 mL) was added 4N HCI in dioxane (0.05 mL, 0.20 mmol). After stirring 10 min the solvent was removed under reduced pressure and the resulting solid was triturated with ether and filtered to give the hydrochloride salt 8 as a white solid (65 mg, 75%): Anal. (C37H49N4O6S,F2Cl1): Calc: C, 59.15; H, 6.45; N, 7.47. Found: C, 58.90; H, 6.51; N, 7.21.
Example 9
Figure imgf000024_0002
9 To a solution of 7A ( 100 mg, 0.16 mmol) in THF (2 mL) was added 2,5- difluorobenzylamine (610 mg, 4.3 mmol) and the reaction mixture was refluxed 6 hours. The reaction mixture was diluted with EtOAc and was washed with water (4x), brine, and dried (MgSO4). The solvent was removed under reduced pressure and the residue was chromatographed (silica gel, 5% methanol/CH2Cl2) to give the amine as a white solid (1 10 mg, 93%). To a solution of the freebase (1 10 mg, 0.15 mmol) in ether (25 mL) was added 4N HCI in dioxane (0.05 mL, 0.20 mmol). After stirring 10 min the solvent was removed under reduced pressure and the resulting solid was triturated with ether and filtered to give the hydrochloride salt 9 as a white solid (76 mg, 66%): CIMS (NH3) m/z: 715 (M+H+,
100%). Example 10
Figure imgf000025_0001
10
10 To a solution of 7A ( 100 mg, 0.16 mmol) in THF (2 mL) was added 2,6- difluorobenzylamine (600 mg, 4.2 mmol) and the reaction mixture was refluxed 6 hours. The reaction mixture was diluted with EtOAc and was washed with water (4x), brine, and dried (MgSO4). The solvent was removed under reduced pressure and the residue was chromatographed (silica gel, 5% methanol/CH2Cl2) to give the amine as a white solid (103 mg, 87%). To a solution of the freebase (103 mg, 0.14 mmol) in ether (25 mL) was added 4N HCI in dioxane (0.05 mL, 0.20 mmol). After stirring 10 min the solvent was removed under reduced pressure and the resulting solid was triturated with ether and filtered to give the hydrochloride salt 10 as a white solid (82 mg, 76%): CIMS (NH3) m/z: 715 (M+H+,
100%).
Example 11
Figure imgf000025_0002
11
11 To a solution of N-[2R-hydroxy-3-[[(l,3-benzodioxol-5-yl)sulfonyl](2- methylpropyl)amino]-lS-(phenylmethyl)propyl]-2S-[(chloroacetyl)amino]-3,3- dimethylbutanamide 11A (750 mg, 1.23 mmol) in THF (2 mL) was added 3- fluorobenzylamine (1.1 g, 8.8 mmol) and the reaction mixture was stirred overnight. The reaction mixture was diluted with EtOAc and was washed with water (4x), brine, and dried (MgSO4). The solvent was removed under reduced pressure and the residue was chromatographed (silica gel, 5% methanol/CH2Cl2) to give the amine as a white solid (532 mg, 62%). To a solution of the freebase (532 mg, 0.76 mmol) in ether (100 mL) was added 4N HCI in dioxane (0.22 mL, 0.88 mmol). After stirring 10 min the solvent was removed under reduced pressure and the resulting solid was triturated with ether and filtered to give the hydrochloride salt 11 as a white solid (417 mg, 75%): CIMS (NH3) m/z: 699 (M+H+, 100%).
Example 12
Figure imgf000026_0001
12 To a solution of 11A (2.0 g, 3.27 mmol) in THF (7 mL) was added 3,5- difluorobenzylamine (2.42 g, 16.9 mmol) and the reaction mixture was refluxed 5 hours. The reaction mixture was diluted with EtOAc and was washed with water (4x), brine, and dried (MgSO4). The solvent was removed under reduced pressure and the residue was chromatographed (silica gel, 5% methanol/CH2Cl2) to give the amine as a white solid (2.26g, 97%). To a solution of the freebase (1.8 g, 2.51 mmol) in ether (100 mL) was added 4N HCI in dioxane (0.66 mL, 2.67 mmol). After stirring 10 min the solvent was removed under reduced pressure and the resulting solid was triturated with ether and filtered to give the benzylamine salt 12 as a white solid (1.65 g, 87%): CIMS (NH3) m/z:
717 (M+H+, 100%). Anal. (C36H47N4O7S,F2Cl,): Calc: C, 57.40; H, 6.17; N, 7.45. Found: C, 57.25; H, 6.25; N, 7.24. Example 13
Figure imgf000027_0001
13
13 To a solution of 11 A (750 mg, 1.23 mmol) in THF (2 mL) was added 2,5- difluorobenzylamine (1.2 g, 8.5 mmol) and the reaction mixture was refluxed 6 hours. The reaction mixture was diluted with EtOAc and was washed with water (4x), brine, and dried (MgSO ). The solvent was removed under reduced pressure and the residue was chromatographed (silica gel, 5% methanol/CH2Cl2) to give the amine as a white solid (702 mg, 80%). To a solution of the freebase (702 mg, 0.98 mmol) in ether (100 mL) and EtOAc (25 mL) was added 4N HCI in dioxane (0.3 mL, 1.2 mmol). After stirring 10 min the solvent was removed under reduced pressure and the resulting solid was triturated with ether and filtered to give the hydrochloride salt 13 as a white solid (586 mg, 79%): CIMS (NH3) m/z: 717 (M+H+, 100%).
Example 14
Figure imgf000027_0002
14
14 To a solution of 11A (750 mg, 1.23 mmol) in THF (2 mL) was added 2,6- difluorobenzylamine (1.2 g, 8.3 mmol) and the reaction mixture was refluxed 6 hours. The reaction mixture was diluted with EtOAc and was washed with water (4x), brine, and dried (MgSO ). The solvent was removed under reduced pressure and the residue was chromatographed (silica gel, 5% methanol/CH2Cl2) to give the amine as a white solid (717 mg, 81%). To a solution of the freebase (717 mg, 1.00 mmol) in ether (100 mL) was added 4N HCI in dioxane (0.3 mL, 1.2 mmol). After stirring 10 min the solvent was removed under reduced pressure and the resulting solid was triturated with ether and filtered to give the hydrochloride salt 14 as a white solid (663 mg, 88%): CIMS (NH3) m/z: 717 (M+H+ 100%). Example 15
Figure imgf000028_0001
15
15 To a solution of 11 A (750 mg, 1.23 mmol) in THF (2 mL) was added 3,4- difluorobenzylamine (1.2 g, 8.3 mmol) and the reaction mixture was refluxed 3 hours. The reaction mixture was diluted with EtOAc and was washed with water (4x), brine, and dried (MgSO4). The solvent was removed under reduced pressure and the residue was chromatographed (silica gel, 5% methanol/CH2Cl2) to give the amine as a white solid (760 mg, 86%). To a solution of the freebase (760 mg, 1.06 mmol) in ether (100 mL) was added 4N HCI in dioxane (0.3 mL, 1.2 mmol). After stirring 10 min the resulting solid was filtered to give the hydrochloride salt 15 as a white solid (730 mg, 91%): CIMS (NH3) m/z: 717 (M+H+ 100%).
Example 16
Figure imgf000028_0002
16
16 To a solution of 11 A (750 mg, 1.23 mmol) in THF (2 mL) was added 2,4- difluorobenzylamine (1.2 g, 8.3 mmol) and the reaction mixture was refluxed 6 hours. The reaction mixture was diluted with EtOAc and was washed with water (4x), brine, and dried (MgSO4). The solvent was removed under reduced pressure and the residue was chromatographed (silica gel, 5% methanol/CH2Cl2) to give the amine as a white solid (693 mg, 79%). To a solution of the freebase (693 mg, 0.97 mmol) in ether (100 mL) was added 4N HCI in dioxane (0.3 mL, 1.2 mmol). After stirring 10 min the resulting solid was filtered to give the hydrochloride salt 16 as a white solid (638 mg, 88%): CIMS (NH3) m/z: 717 (M+H+, 100%). Example 17
Figure imgf000029_0001
17
17 To a solution of 11 A (500 mg, 0.82 mmol) in THF (2 mL) was added 4- fluorobenzylamine (1.0 g, 8.0 mmol) and the reaction mixture was stirred overnight. The reaction mixture was diluted with EtOAc and was washed with water (4x), brine, and dried (MgSO4). The solvent was removed under reduced pressure and the residue was chromatographed (silica gel, 5% methanol/CH2Cl2) to give the amine as a white solid (470 mg, 82%). To a solution of the freebase (400 mg, 0.57 mmol) in ether (30 mL) was added 4N HCI in dioxane (0.18 mL, 0.7 mmol). After stirring 15 min the resulting solid was filtered to give the hydrochloride salt 17 as a white solid (413 mg, 98%): CIMS (NH3) m/z: 699 (M+H+, 100%).
Utility The compounds of formula I possess HIV protease inhibitory activity and are therefore useful as antiviral agents for the treatment of HIV infection and associated diseases. The compounds of formula I possess HIV protease inhibitory activity and are effective as inhibitors of HIV growth. The ability of the compounds of the present invention to inhibit viral growth or infectivity is demonstrated in standard assay of viral growth or infectivity, for example, using the assay described below.
The compounds of formula I of the present invention are also useful for the inhibition of HIV in an ex vivo sample containing HIV or expected to be exposed to HIV. Thus, the compounds of the present invention may be used to inhibit HIV present in a body fluid sample (for example, a serum or semen sample) which contains or is suspected to contain or be exposed to HIV.
The compounds provided by this invention are also useful as standard or reference compounds for use in tests or assays for determining the ability of an agent to inhibit viral clone replication and/or HIV protease, for example in a pharmaceutical research program. Thus, the compounds of the present invention may be used as a control or reference compound in such assays and as a quality control standard. The compounds of the present invention may be provided in a commercial kit or container for use as such standard or reference compound. Since the compounds of the present invention exhibit specificity for HIV protease, the compounds of the present invention may also be useful as diagnostic reagents in diagnostic assays for the detection of HIV protease. Thus, inhibition of the protease activity in an assay (such as the assays described herein) by a compound of the present invention would be indicative of the presence of HIV protease and HIV virus. As used herein "μg" denotes microgram, "mg" denotes milligram, "g" denotes gram, "μL" denotes microliter, "mL" denotes milliliter, "L" denotes liter, "nM" denotes nanomolar, "μM" denotes micromolar, "mM" denotes millimolar, "M" denotes molar and "nm" denotes nanometer. "Sigma" stands for the Sigma- Aldrich Corp. of St. Louis, MO.
HIV RNA Assay
DNA Plasmids and in vitro RNA transcripts:
Plasmid pDAB 72 containing both gag and pol sequences of BH10 (bp 1 13-1816) cloned into PTZ 19R was prepared according to Erickson-Viitanen et al. AIDS Research and Human Retroviruses 1989, 5, 577. The plasmid was linearized with Bam HI prior to the generation of in vitro RNA transcripts using the Riboprobe Gemini system II kit (Promega) with T7 RNA polymerase. Synthesized RNA was purified by treatment with RNase free DNAse (Promega), phenol-chloroform extraction, and ethanol precipitation. RNA transcripts were dissolved in water, and stored at -70°C. The concentration of RNA was determined from the A260-
Probes: Biotinylated capture probes were purified by HPLC after synthesis on an Applied
Biosystems (Foster City, CA) DNA synthesizer by addition of biotin to the 5' teiminal end of the oligonucleotide, using the biotin-phosphoramidite reagent of Cocuzza, Tet. Lett. 1989, 30, 6287. The gag biotinylated capture probe (5-biotin- CTAGCTCCCTGCTTGCCCATACTA 3*) was complementary to nucleotides 889-912 of HXB2 and the pol biotinylated capture probe (5'-biotin -
CCCTATCATTTTTGGTTTCCAT 3' ) was complementary to nucleotides 2374-2395 of HXB2. Alkaline phosphatase conjugated oligonucleotides used as reporter probes were prepared by Syngene (San Diego, CA.). The pol reporter probe (5' CTGTCTTACTTTGATAAAACCTC 3') was complementary to nucleotides 2403-2425 of HXB2. The gag reporter probe (5' CCCAGTATTTGTCTACAGCCTTCT 3') was complementary to nucleotides 950-973 of HXB2. All nucleotide positions are those of the GenBank Genetic Sequence Data Bank as accessed through the Genetics Computer Group Sequence Analysis Software Package (Devereau Nucleic Acids Research 1984, 12, 387). The reporter probes were prepared as 0.5 μM stocks in 2 x SSC (0.3 M NaCl, 0.03 M sodium citrate), 0.05 M Tris pH 8.8, 1 mg/mL BSA. The biotinylated capture probes were prepared as 100 μM stocks in water.
Streptavidin coated plates:
Streptavidin coated plates were obtained from Du Pont Biotechnology Systems (Boston, MA).
Cells and virus stocks:
MT-2 and MT-4 cells were maintained in RPMI 1640 supplemented with 5% fetal calf serum (FCS) for MT-2 cells or 10% FCS for MT-4 cells, 2 mM L-glutamine and 50 μg/mL gentamycin, all from Gibco. HIV-1 RF was propagated in MT-4 cells in the same medium. Virus stocks were prepared approximately 10 days after acute infection of MT-4 cells and stored as aliquots at -70°C. Infectious titers of HIV-1 (RF) stocks were 1-3 x 10^ PFU (plaque forming units)/mL as measured by plaque assay on MT-2 cells (see below). Each aliquot of virus stock used for infection was thawed only once. For evaluation of antiviral efficacy, cells to be infected were subcultured one day prior to infection. On the day of infection, cells were resuspended at 5 x 10^ cells/mL in RPMI 1640, 5% FCS for bulk infections or at 2 x 106/mL in Dulbecco's modified Eagles medium with 5% FCS for infection in microtiter plates. Virus was added and culture continued for 3 days at 37°C.
HIV RNA assav: Cell lysates or purified RNA in 3 M or 5 M GED were mixed with 5 M GED and capture probe to a final guanidinium isothiocyanate concentration of 3 M and a final biotin oligonucleotide concentration of 30 nM. Hybridization was carried out in sealed U bottom 96 well tissue culture plates (Nunc or Costar) for 16-20 hours at 37°C. RNA hybridization reactions were diluted three-fold with deionized water to a final guanidinium isothiocyanate concentration of 1 M and aliquots (150 μL) were transferred to streptavidin coated microtiter plates wells. Binding of capture probe and capture probe-RNA hybrid to the immobilized streptavidin was allowed to proceed for 2 hours at room temperature, after which the plates were washed 6 times with DuPont ELISA plate wash buffer (phosphate buffered saline(PBS), 0.05% Tween 20.) A second hybridization of reporter probe to the immobilized complex of capture probe and hybridized target RNA was carried out in the washed streptavidin coated well by addition of 120 μl of a hybridization cocktail containing 4 X SSC, 0.66% Triton X 100, 6.66% deionized formamide, 1 mg/mL BSA and 5 nM reporter probe. After hybridization for one hour at 37 C, the plate was again washed 6 times. Immobilized alkaline phosphatase activity was detected by addition of 100 μL of 0.2 mM 4-methylumbelliferyl phosphate (MUBP, JBL Scientific) in buffer δ (2.5 M diethanolamine pH 8.9 (JBL Scientific), 10 mM MgCk>, 5 mM zinc acetate dihydrate and 5 mM N-hydroxyethyl-ethylene-diamine-triacetic acid). The plates were incubated at 37°C. Fluorescence at 450 nM was measured using a microplate fluorometer (Dynateck) exciting at 365 nM.
Microplate based compound evaluation in HIN-1 infected MT-2 cells:
Compounds to be evaluated were dissolved in DMSO and diluted in culture medium to twice the highest concentration to be tested and a maximum DMSO concentration of 2%. Further three-fold serial dilutions of the compound in culture medium were performed directly in U bottom microtiter plates (Νunc). After compound dilution, MT-2 cells (50 μL) were added to a final concentration of 5 x 10^ per mL (1 x 10^ per well). Cells were incubated with compounds for 30 minutes at 37°C in a CO2 incubator. For evaluation of antiviral potency, an appropriate dilution of HIV-1 (RF) virus stock (50 μL) was added to culture wells containing cells and dilutions of the test compounds. The final volume in each well was 200 μL. Eight wells per plate were left uninfected with 50 μL of medium added in place of virus, while eight wells were infected in the absence of any antiviral compound. For evaluation of compound toxicity, parallel plates were cultured without virus infection. After 3 days of culture at 37°C in a humidified chamber inside a CO2 incubator, all but 25 μL of medium/well was removed from the HIV infected plates. Thirty seven μL of 5 M GED containing biotinylated capture probe was added to the settled cells and remaining medium in each well to a final concentration of 3 M GED and 30 nM capture probe. Hybridization of the capture probe to HIV RNA in the cell lysate was carried out in the same microplate well used for virus culture by sealing the plate with a plate sealer (Costar), and incubating for 16-20 hrs in a 37°C incubator. Distilled water was then added to each well to dilute the hybridization reaction three-fold and 150 μL of this diluted mixture was transferred to a streptavidin coated microtiter plate. HIV RNA was quantitated as described above. A standard curve, prepared by adding known amounts of pDAB 72 in vitro RNA transcript to wells containing lysed uninfected cells, was run on each microtiter plate in order to determine the amount of viral RNA made during the infection.
In order to standardize the virus inoculum used in the evaluation of compounds for antiviral activity, dilutions of virus were selected which resulted in an IC90 value
(concentration of compound required to reduce the HIV RNA level by 90%) for dideoxycytidine (ddC) of 0.2 μg/mL. IC90 values of other antiviral compounds, both more and less potent than ddC, were reproducible using several stocks of HIV-1 (RF) when this procedure was followed. This concentration of virus corresponded to ~3 x 10^ PFU (measured by plaque assay on MT-2 cells) per assay well and typically produced approximately 75% of the maximum viral RNA level achievable at any virus inoculum. For the HIV RNA assay, IC90 values were determined from the percent reduction of net signal (signal from infected cell samples minus signal from uninfected cell samples) in the RNA assay relative to the net signal from infected, untreated cells on the same culture plate (average of eight wells). Valid performance of individual infection and RNA assay tests was judged according to three criteria. It was required that the virus infection should result in an RNA assay signal equal to or greater than the signal generated from 2 ng of pDAB 72 in vitro RNA transcript. The IC90 for ddC, determined in each assay run, should be between 0.1 and 0.3 μg/mL. Finally, the plateau level of viral RNA produced by an effective protease inhibitor should be less than 10% of the level achieved in an uninhibited infection. A compound was considered active if its IC90 was found to be less than lμM.
For antiviral potency tests, all manipulations in microtiter plates, following the initial addition of 2X concentrated compound solution to a single row of wells, were performed using a Perkin Elmer/Cetus ProPette. Dosage and Formulation
The antiviral compounds of this invention can be administered as treatment for viral infections by any means that produces contact of the active agent with the agent's site of action, i.e., the viral protease, in the body of a mammal. They can be administered by any conventional means available for use in conjunction with pharmaceuticals, either as individual therapeutic agents or in a combination of therapeutic agents. They can be administered alone, but preferably are administered with a pharmaceutical carrier selected on the basis of the chosen route of administration and standard pharmaceutical practice.
The dosage administered will, of course, vary depending upon known factors, such as the pharmacodynamic characteristics of the particular agent and its mode and route of administration; the age, health and weight of the recipient; the nature and extent of the symptoms; the kind of concurrent treatment; the frequency of treatment; and the effect desired. A daily dosage of active ingredient can be expected to be about 0.001 to about 1000 milligrams per kilogram of body weight, with the preferred dose being about 0.1 to about 30 mg/kg.
Dosage forms of compositions suitable for administration contain from about 1 mg to about 100 mg of active ingredient per unit. In these pharmaceutical compositions the active ingredient will ordinarily be present in an amount of about 0.5-95% by weight based on the total weight of the composition. The active ingredient can be administered orally in solid dosage forms, such as capsules, tablets and powders, or in liquid dosage forms, such as elixirs, syrups and suspensions. It can also be administered parenterally, in sterile liquid dosage forms.
Gelatin capsules contain the active ingredient and powdered carriers, such as lactose, starch, cellulose derivatives, magnesium stearate, stearic acid, and the like. Similar diluents can be used to make compressed tablets. Both tablets and capsules can be manufactured as sustained release products to provide for continuous release of medication over a period of hours. Compressed tablets can be sugar coated or film coated to mask any unpleasant taste and protect the tablet from the atmosphere, or enteric coated for selective disintegration in the gastrointestinal tract. Liquid dosage forms for oral administration can contain coloring and flavoring to increase patient acceptance.
In general, water, a suitable oil, saline, aqueous dextrose (glucose), and related sugar solutions and glycols such as propylene glycol or polyethylene glycols are suitable carriers for parenteral solutions. Solutions for parenteral administration preferably contain a water soluble salt of the active ingredient, suitable stabilizing agents, and if necessary, buffer substances. Antioxidizing agents such as sodium bisulfite, sodium sulfite, or ascorbic acid, either alone or combined, are suitable stabilizing agents. Also used are citric acid and its salts, and sodium EDTA. In addition, parenteral solutions can contain preservatives, such as benzalkonium chloride, methyl- or propyl-paraben and chlorobutanol. Suitable pharmaceutical carriers are described in Remington's Pharmaceutical Sciences, supra, a standard reference text in this field.
Useful pharmaceutical dosage-forms for administration of the compounds of this invention can be illustrated as follows:
Capsules
A large number of unit capsules can be prepared by filling standard two-piece hard gelatin capsules each with 100 mg of powdered active ingredient, 150 mg of lactose, 50 mg of cellulose, and 6 mg magnesium stearic.
Soft Gelatin Capsules
A mixture of active ingredient in a digestible oil such as soybean oil, cottonseed oil or olive oil can be prepared and injected by means of a positive displacement pump into gelatin to form soft gelatin capsules containing 100 mg of the active ingredient. The capsules should then be washed and dried.
Tablets
A large number of tablets can be prepared by conventional procedures so that the dosage unit is 100 mg of active ingredient, 0.2 mg of colloidal silicon dioxide, 5 milligrams of magnesium stearate, 275 mg of microcrystalline cellulose, 11 mg of starch and 98.8 mg of lactose. Appropriate coatings may be applied to increase palatability or delay absorption.
Suspension An aqueous suspension can be prepared for oral administration so that each 5 mL contain 25 mg of finely divided active ingredient, 200 mg of sodium carboxymethyl cellulose, 5 mg of sodium benzoate, 1.0 g of sorbitol solution, U.S. P., and 0.025 mg of vanillin.
Injectable
A parenteral composition suitable for administration by injection can be prepared by stirring 1.5% by weight of active ingredient in 10% by volume propylene glycol and water. The solution is sterilized by commonly used techniques.
Combination of components (a) and (b)
Each therapeutic agent component of this invention can independently be in any dosage foπn, such as those described above, and can also be administered in various ways, as described above. In the following description component (b) is to be understood to represent one or more agents as described previously. Thus, if components (a) and (b) are to be treated the same or independently, each agent of component (b) may also be treated the same or independently.
Components (a) and (b) of the present invention may be formulated together, in a single dosage unit (that is, combined together in one capsule, tablet, powder, or liquid, etc.) as a combination product. When component (a) and (b) are not formulated together in a single dosage unit, the component (a) may be administered at the same time as component (b) or in any order; for example component (a) of this invention may be administered first, followed by administration of component (b), or they may be administered in the revserse order. If component (b) contains more that one agent, e.g., one RT inhibitor and one protease inhibitor, these agents may be administered together or in any order. When not administered at the same time, preferably the administration of component (a) and (b) occurs less than about one hour apart. Preferably, the route of administration of component (a) and (b) is oral. The terms oral agent, oral inhibitor, oral compound, or the like, as used herein, denote compounds which may be orally administered. Although it is preferable that component (a) and component (b) both be administered by the same route (that is, for example, both orally) or dosage form, if desired, they may each be administered by different routes (that is, for example, one component of the combination product may be administered orally, and another component may be administered intravenously) or dosage forms.
As is appreciated by a medical practitioner skilled in the art, the dosage of the combination therapy of the invention may vary depending upon various factors such as the pharmacodynamic characteristics of the particular agent and its mode and route of administration, the age, health and weight of the recipient, the nature and extent of the symptoms, the kind of concurrent treatment, the frequency of treatment, and the effect desired, as described above.
The proper dosage of components (a) and (b) of the present invention will be readily ascertainable by a medical practitioner skilled in the art, based upon the present disclosure. By way of general guidance, typically a daily dosage may be about 100 milligrams to about 1.5 grams of each component. If component (b) represents more than one compound, then typically a daily dosage may be about 100 milligrams to about 1.5 grams of each agent of component (b). By way of general guidance, when the compounds of component (a) and component (b) are administered in combination, the dosage amount of each component may be reduced by about 70-80% relative to the usual dosage of the component when it is administered alone as a single agent for the treatment of HIV infection, in view of the synergistic effect of the combination.
The combination products of this invention may be formulated such that, although the active ingredients are combined in a single dosage unit, the physical contact between the active ingredients is minimized. In order to minimize contact, for example, where the product is orally administered, one active ingredient may be enteric coated. By enteric coating one of the active ingredients, it is possible not only to minimize the contact between the combined active ingredients, but also, it is possible to control the release of one of these components in the gastrointestinal tract such that one of these components is not released in the stomach but rather is released in the intestines. Another embodiment of this invention where oral administration is desired provides for a combination product wherein one of the active ingredients is coated with a sustained-release material which effects a sustained-release throughout the gastrointestinal tract and also serves to minimize physical contact between the combined active ingredients. Furthermore, the sustained-released component can be additionally enteric coated such that the release of this component occurs only in the intestine. Still another approach would involve the formulation of a combination product in which the one component is coated with a sustained and/or enteric release polymer, and the other component is also coated with a polymer such as a lowviscosity grade of hydroxypropyl methylcellulose or other appropriate materials as known in the art, in order to further separate the active components. The polymer coating serves to form an additional barrier to interaction with the other component. In each formulation wherein contact is prevented between components (a) and (b) via a coating or some other material, contact may also be prevented between the individual agents of component (b).
Dosage forms of the combination products of the present invention wherein one active ingredient is enteric coated can be in the form of tablets such that the enteric coated component and the other active ingredient are blended together and then compressed into a tablet or such that the enteric coated component is compressed into one tablet layer and the other active ingredient is compressed into an additional layer. Optionally, in order to further separate the two layers, one or more placebo layers may be present such that the placebo layer is between the layers of active ingredients. In addition, dosage forms of the present invention can be in the form of capsules wherein one active ingredient is compressed into a tablet or in the form of a plurality of microtablets, particles, granules or non-perils, which are then enteric coated. These enteric coated microtablets, particles, granules or non-perils are then placed into a capsule or compressed into a capsule along with a granulation of the other active ingredient.
These as well as other ways of minimizing contact between the components of combination products of the present invention, whether administered in a single dosage form or administered in separate forms but at the same time or concurrently by the same manner, will be readily apparent to those skilled in the art, based on the present disclosure.
Pharmaceutical kits useful for the treatment of HIV infection, which comprise a therapeutically effective amount of a pharmaceutical composition comprising a compound of component (a) and one or more compounds of component (b), in one or more sterile containers, are also within the ambit of the present invention. Sterilization of the container may be carried out using conventional sterilization methodology well known to those skilled in the art. Component (a) and component (b) may be in the same sterile container or in separate sterile containers. The sterile containers of materials may comprise separate containers, or one or more multi-part containers, as desired. Component (a) and component (b), may be separate, or physically combined into a single dosage form or unit as described above. Such kits may further include, if desired, one or more of various conventional pharmaceutical kit components, such as for example, one or more pharmaceutically acceptable carriers, additional vials for mixing the components, etc., as will be readily apparent to those skilled in the art. Instructions, either as inserts or as labels, indicating quantities of the components to be administered, guidelines for administration, and/or guidelines for mixing the components, may also be included in the kit. Obviously, numerous modifications and variations of the present invention are possible in light of the above teachings. It is therefore to be understood that within the scope of the appended claims, the invention may be practiced otherwise than as specifically described herein.

Claims

WHAT IS CLAIMED:
1. A compound of formula I:
Figure imgf000039_0001
I or a pharmaceutically acceptable salt form thereof, wherein:
R s F;
R2 is F or H; and,
R3 is selected from the group: 4-aminophenyl, 3-aminophenyl, 2,3-dihydrobenzofuran-5- yl, and l,3-benzodioxol-5-yl.
2. A compound according to Claim 1, wherein the compound is of Formula II:
Figure imgf000039_0002
II.
3. A compound according to Claim 2, wherein the compound is of Formula Ila:
Figure imgf000039_0003
Ila.
4. A compound according to Claim 3, wherein: R3 is 3-aminophenyl.
5. A compound according to Claim 3, wherein:
R3 is 4-aminophenyl.
6. A compound according to Claim 3, wherein:
R3 is 2,3-dihydrobenzofuran-5-yl or l,3-benzodioxol-5-yl.
7. A compound according to Claim 2, wherein the compound is of Formula lib:
Figure imgf000040_0001
lib.
8. A compound according to Claim 7, wherein:
R3 is 3-aminophenyl.
9. A compound according to Claim 7, wherein:
R3 is 4-aminophenyl.
10. A compound according to Claim 7, wherein:
R3 is 2,3-dihydrobenzofuran-5-yl or 1 ,3-benzodioxol-5-yl.
1 1. A compound according to Claim 2, wherein the compound is of Formula He:
Figure imgf000041_0001
lie.
12. A compound according to Claim 1 1, wherein:
R3 is 3-aminophenyl.
13. A compound according to Claim 11, wherein:
R3 is 4-aminophenyl.
14. A compound according to Claim 11, wherein:
R3 is 2,3-dihydrobenzofuran-5-yl or l,3-benzodioxol-5-yl.
15. A compound according to Claim 1, wherein the compound is of Formula III:
Figure imgf000041_0002
III.
16. A compound according to Claim 15, wherein the compound is of Formula Ilia:
Figure imgf000042_0001
Ilia.
17. A compound according to Claim 1 , wherein the compound is of Formula IV:
Figure imgf000042_0002
IV.
18. A compound according to Claim 17, wherein the compound is of Formula IVa:
Figure imgf000042_0003
IVa.
19. A pharmaceutical composition, comprising: a pharmaceutically acceptable carrier and a therapeutically effective amount of a compound according to one of Claims 1-19.
20. A method for treating HIV infection, comprising: administering to a host in need of such treatment a therapeutically effective amount of a compound according to one of Claims 1-19, or a pharmaceutically acceptable salt form thereof.
21. A method of treating HIV infection which comprises administering, in combination, to a host in need thereof a therapeutically effective amount of:
(a) a compound according to one of Claims 1-19 or stereoisomeric forms, mixtures of stereoisomeric forms, or pharmaceutically acceptable salts thereof; and, (b) at least one compound selected from the group consisting of HIV reverse transcriptase inhibitors and HIV protease inhibitors.
22. A method according to Claim 21, wherein the reverse transcriptase inhibitor is selected from the group AZT, ddC, ddl, d4T, 3TC, delavirdine, efavirenz, nevirapine, Ro 18,893, trovirdine, MKC-442, HBY 097, ACT, UC-781, UC-782, RD4-2025, and MEN 10979 and the protease inhibitor is selected from the group saquinavir, ritonavir, indinavir, amprenavir, nelfinavir, palinavir, BMS-232623, GS3333, KNI-413, KNI-272, LG-71350, CGP-61755, PD 173606, PD 177298, PD 178390, PD 178392, U- 140690, and ABT-378.
23. A method according to Claim 22, wherein the reverse transcriptase inhibitor is selected from the group AZT, efavirenz, and 3TC and the protease inhibitor is selected from the group saquinavir, ritonavir, nelfinavir, and indinavir.
24. A method according to Claim 21, wherein compound (b) is ritonavir.
PCT/US2000/000820 1999-01-13 2000-01-13 Bis-amino acid sulfonamides containing n-terminally a substituted benzyl group as hiv protease inhibitors WO2000042060A1 (en)

Priority Applications (11)

Application Number Priority Date Filing Date Title
IL14376300A IL143763A0 (en) 1999-01-13 2000-01-13 Bis-amino acid sulfonamides containing n-terminally a substituted benzyl group as hiv protease inhibitors
KR1020017008803A KR20010101478A (en) 1999-01-13 2000-01-13 Bis-Amino Acid Sulfonamides Containing N-Terminally a Substituted Benzyl Group as HIV Protease Inhibitors
CA002353088A CA2353088A1 (en) 1999-01-13 2000-01-13 Bis-amino acid sulfonamides containing n-terminally a substituted benzyl group as hiv protease inhibitors
SK952-2001A SK9522001A3 (en) 1999-01-13 2000-01-13 Bis-amino acid sulfonamides containing n-terminally a substituted benzyl group as hiv protease inhibitors
HU0105235A HUP0105235A3 (en) 1999-01-13 2000-01-13 Bis-amino acid sulfonamides containing n-terminally a substituted benzyl group as hiv protease inhibitors, pharmaceutical compositions comprising thereof and their use
BR0007854-9A BR0007854A (en) 1999-01-13 2000-01-13 Sulfonamide compounds bis-amino acids hiv protease inhibitors, pharmaceutical composition and method of treatment of hiv infections
JP2000593627A JP2002536298A (en) 1999-01-13 2000-01-13 Bis-amino acid sulfonamide containing N-terminally substituted benzyl group as HIV protease inhibitor
EP00905604A EP1140983A1 (en) 1999-01-13 2000-01-13 Bis-amino acid sulfonamides containing n-terminally a fluoro-substituted benzyl group as hiv protease inhibitors
PL00349774A PL349774A1 (en) 1999-01-13 2000-01-13 Bis-amino acid sulfonamides containing n-terminally a substituted benzyl group as hiv protease inhibitors
AU27257/00A AU2725700A (en) 1999-01-13 2000-01-13 Bis-amino acid sulfonamides containing n-terminally a substituted benzyl group as hiv protease inhibitors
NO20013338A NO20013338L (en) 1999-01-13 2001-07-05 Bis-amino acid sulfonamides containing N-terminally a substituted benzyl group as HIV protease inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US11574699P 1999-01-13 1999-01-13
US60/115,746 1999-01-13

Publications (1)

Publication Number Publication Date
WO2000042060A1 true WO2000042060A1 (en) 2000-07-20

Family

ID=22363180

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2000/000820 WO2000042060A1 (en) 1999-01-13 2000-01-13 Bis-amino acid sulfonamides containing n-terminally a substituted benzyl group as hiv protease inhibitors

Country Status (15)

Country Link
EP (1) EP1140983A1 (en)
JP (1) JP2002536298A (en)
KR (1) KR20010101478A (en)
CN (1) CN1336934A (en)
AU (1) AU2725700A (en)
BR (1) BR0007854A (en)
CA (1) CA2353088A1 (en)
CZ (1) CZ20012435A3 (en)
HU (1) HUP0105235A3 (en)
IL (1) IL143763A0 (en)
NO (1) NO20013338L (en)
PL (1) PL349774A1 (en)
SK (1) SK9522001A3 (en)
TR (1) TR200102033T2 (en)
WO (1) WO2000042060A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002006292A1 (en) * 2000-07-19 2002-01-24 Bristol-Myers Sqibb Company Phosphate esters of bis-amino acid sulfonamides containing substituted benzyl amines
WO2002006190A2 (en) * 2000-07-19 2002-01-24 Bristol-Myers Squibb Pharma Company Crystalline and salt forms of an hiv protease inhibitor
WO2002010124A2 (en) * 2000-07-19 2002-02-07 Dupont Pharmaceuticals Company Salt forms of an hiv protease inhibitor
EP1188766A1 (en) * 1995-03-10 2002-03-20 G.D. Searle & Co. Bis-amino acid hydroxyethylamino sulfonamide retroviral protease inhibitors
US6861539B1 (en) 1995-03-10 2005-03-01 G. D. Searle & Co. Bis-amino acid hydroxyethylamino sulfonamide retroviral protease inhibitors
US7339078B2 (en) 1995-03-10 2008-03-04 G.D. Searle Llc Bis-amino acid hydroxyethylamino sulfonamide retroviral protease inhibitors

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994004492A1 (en) * 1992-08-25 1994-03-03 G.D. Searle & Co. Hydroxyethylamino sulfonamides useful as retroviral protease inhibitors
WO1995006030A1 (en) * 1993-08-24 1995-03-02 G.D. Searle & Co. Hydroxyethylamino sulphonamides useful as retroviral protease inhibitors
WO1996028464A1 (en) * 1995-03-10 1996-09-19 G.D. Searle & Co. Bis-amino acid hydroxyethylamino sulfonamide retroviral protease inhibitors

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994004492A1 (en) * 1992-08-25 1994-03-03 G.D. Searle & Co. Hydroxyethylamino sulfonamides useful as retroviral protease inhibitors
WO1995006030A1 (en) * 1993-08-24 1995-03-02 G.D. Searle & Co. Hydroxyethylamino sulphonamides useful as retroviral protease inhibitors
WO1996028464A1 (en) * 1995-03-10 1996-09-19 G.D. Searle & Co. Bis-amino acid hydroxyethylamino sulfonamide retroviral protease inhibitors

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1188766A1 (en) * 1995-03-10 2002-03-20 G.D. Searle & Co. Bis-amino acid hydroxyethylamino sulfonamide retroviral protease inhibitors
US6683210B2 (en) 1995-03-10 2004-01-27 G. D. Searle & Co. Bis-amino acid hydroxyethylamino sulfonamide retroviral protease inhibitors
US6861539B1 (en) 1995-03-10 2005-03-01 G. D. Searle & Co. Bis-amino acid hydroxyethylamino sulfonamide retroviral protease inhibitors
US7161033B2 (en) 1995-03-10 2007-01-09 G. D. Searle & Co. Bis-amino acid hydroxyethylamino sulfonamide retroviral protease inhibitors
US7339078B2 (en) 1995-03-10 2008-03-04 G.D. Searle Llc Bis-amino acid hydroxyethylamino sulfonamide retroviral protease inhibitors
WO2002006292A1 (en) * 2000-07-19 2002-01-24 Bristol-Myers Sqibb Company Phosphate esters of bis-amino acid sulfonamides containing substituted benzyl amines
WO2002006190A2 (en) * 2000-07-19 2002-01-24 Bristol-Myers Squibb Pharma Company Crystalline and salt forms of an hiv protease inhibitor
WO2002010124A2 (en) * 2000-07-19 2002-02-07 Dupont Pharmaceuticals Company Salt forms of an hiv protease inhibitor
WO2002006190A3 (en) * 2000-07-19 2002-06-20 Bristol Myers Squibb Pharma Co Crystalline and salt forms of an hiv protease inhibitor
WO2002010124A3 (en) * 2000-07-19 2003-05-01 Du Pont Pharm Co Salt forms of an hiv protease inhibitor
US6617310B2 (en) 2000-07-19 2003-09-09 Bristol-Myers Squibb Pharma Company Phosphate esters of bis-amino acid sulfonamides containing substituted benzyl amines

Also Published As

Publication number Publication date
CN1336934A (en) 2002-02-20
NO20013338L (en) 2001-09-13
EP1140983A1 (en) 2001-10-10
CZ20012435A3 (en) 2002-02-13
NO20013338D0 (en) 2001-07-05
TR200102033T2 (en) 2001-12-21
SK9522001A3 (en) 2002-03-05
HUP0105235A3 (en) 2002-08-28
KR20010101478A (en) 2001-11-14
PL349774A1 (en) 2002-09-09
JP2002536298A (en) 2002-10-29
BR0007854A (en) 2002-01-15
HUP0105235A2 (en) 2002-04-29
IL143763A0 (en) 2002-04-21
CA2353088A1 (en) 2000-07-20
AU2725700A (en) 2000-08-01

Similar Documents

Publication Publication Date Title
US6825210B2 (en) Tricyclic compounds useful as HIV reverse transcriptase inhibitors
US6391919B1 (en) Bis-amino acid sulfonamides containing substituted benzyl amines HIV protease inhibitors
WO2000042060A1 (en) Bis-amino acid sulfonamides containing n-terminally a substituted benzyl group as hiv protease inhibitors
US5932570A (en) 1-(3-aminoindazol-5-yl)-3-phenylmethyl-cyclic ureas useful as HIV protease inhibitors
EP0937067B1 (en) 1-(3-aminoindazol-5-yl)-3-phenylmethyl-cyclic ureas useful as hiv protease inhibitors
US6218386B1 (en) A1-(3-aminoindazol-5-yl)-3 butyl-cyclic urea useful as a HIV protease inhibitor
US6562848B1 (en) Bis-amino acid sulfonamides as HIV protease inhibitors
US6313110B1 (en) Substituted 2H-1,3-diazapin-2-one useful as an HIV protease inhibitor
AU722489B2 (en) (4r,5s,6s,7r)-hexahydro-1- {5-(3-aminoinazole)methyl} -3-butyl-5,6-dihydr oxy-4,7-bis {phaenylmethyl} -2h-1,3-diazepin-2-one, its preparation and its use as HIV protease inhibitor
US6943170B2 (en) N-cycloalkylglycines as HIV protease inhibitors
US20020022742A1 (en) Salt forms of an HIV protease inhibitor
US20040063734A1 (en) 4,4-Disubstituted-3,4-dihydro-2 (1H)-quinazoliniones useful as HIV reverse transcriptase inhibitors
MXPA01007047A (en) Bis-amino acid sulfonamides containing n-terminally a substituted benzyl group as hiv protease inhibitors
US7015214B2 (en) Cyanamide, alkoxyamino, and urea derivatives of 1,3-benzodiazepine as HIV reverse transcriptase inhibitors
US20020022659A1 (en) Crystalline and salt forms of an HIV protease inhibitor
US6265406B1 (en) Substituted quinolin-2 (1H) -ones useful as HIV reverse transcriptase inhibitors
US20060128634A1 (en) Alpha, alpha-disubstituted benzylglycine derivatives as HIV protease inhibitors
HRP970595A2 (en) 1-(3-aminoindazol-5-yl)-3-butyl-cyclic urea useful as a hiv proteaze inhibitor
HRP970586A2 (en) 1-(3-aminoindazol-5-yl)-3-phenylmethyl-cyclic ureas useful as hiv protease inhibitors
MXPA99004294A (en) (4r,5s,6s,7r)-hexahydro-1- [5-(3-aminoinazole)methyl]-3-butyl-5,6-dihydroxy-4,7-bis [phaenylmethyl]-2h-1,3-diazepin-2-one, its preparation and its use as hiv protease inhibitor
MXPA99004286A (en) 1-(3-aminoindazol-5-yl)-3-phenylmethyl-cyclic ureas useful as hiv protease inhibitors
AU2002254652A1 (en) Tricyclic compounds useful as HIV reverse transcriptase inhibitors

Legal Events

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

Ref document number: 00802755.2

Country of ref document: CN

AK Designated states

Kind code of ref document: A1

Designated state(s): AU BR CA CN CZ EE HU IL IN JP KR LT LV MX NO NZ PL RO SG SI SK TR UA VN ZA

AL Designated countries for regional patents

Kind code of ref document: A1

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
ENP Entry into the national phase

Ref document number: 2353088

Country of ref document: CA

Ref document number: 2353088

Country of ref document: CA

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 200104499

Country of ref document: ZA

WWE Wipo information: entry into national phase

Ref document number: 512151

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 27257/00

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 143763

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: IN/PCT/2001/00723/MU

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2000905604

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: PV2001-2435

Country of ref document: CZ

WWE Wipo information: entry into national phase

Ref document number: 9522001

Country of ref document: SK

ENP Entry into the national phase

Ref document number: 2000 593627

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: PA/a/2001/007047

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 1020017008803

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 2001/02033

Country of ref document: TR

WWP Wipo information: published in national office

Ref document number: 2000905604

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 09980529

Country of ref document: US

WWP Wipo information: published in national office

Ref document number: 1020017008803

Country of ref document: KR

WWP Wipo information: published in national office

Ref document number: PV2001-2435

Country of ref document: CZ

WWR Wipo information: refused in national office

Ref document number: PV2001-2435

Country of ref document: CZ

WWW Wipo information: withdrawn in national office

Ref document number: 2000905604

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1020017008803

Country of ref document: KR