EP1140983A1 - Bis-amino acid sulfonamides containing n-terminally a fluoro-substituted benzyl group as hiv protease inhibitors - Google Patents

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

Info

Publication number
EP1140983A1
EP1140983A1 EP00905604A EP00905604A EP1140983A1 EP 1140983 A1 EP1140983 A1 EP 1140983A1 EP 00905604 A EP00905604 A EP 00905604A EP 00905604 A EP00905604 A EP 00905604A EP 1140983 A1 EP1140983 A1 EP 1140983A1
Authority
EP
European Patent Office
Prior art keywords
compound
compound according
mmol
formula
hiv
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP00905604A
Other languages
German (de)
English (en)
French (fr)
Inventor
Robert F. Kaltenbach
George L. Trainor
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Bristol Myers Squibb Pharma Co
Original Assignee
Bristol Myers Squibb Pharma Co
DuPont Merck Pharmaceutical Co
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Bristol Myers Squibb Pharma Co, DuPont Merck Pharmaceutical Co filed Critical Bristol Myers Squibb Pharma Co
Publication of EP1140983A1 publication Critical patent/EP1140983A1/en
Withdrawn legal-status Critical Current

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.
EP00905604A 1999-01-13 2000-01-13 Bis-amino acid sulfonamides containing n-terminally a fluoro-substituted benzyl group as hiv protease inhibitors Withdrawn EP1140983A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US11574699P 1999-01-13 1999-01-13
US115746P 1999-01-13
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

Publications (1)

Publication Number Publication Date
EP1140983A1 true EP1140983A1 (en) 2001-10-10

Family

ID=22363180

Family Applications (1)

Application Number Title Priority Date Filing Date
EP00905604A Withdrawn EP1140983A1 (en) 1999-01-13 2000-01-13 Bis-amino acid sulfonamides containing n-terminally a fluoro-substituted benzyl group as hiv protease inhibitors

Country Status (15)

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

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
EP1188766A1 (en) 1995-03-10 2002-03-20 G.D. Searle & Co. Bis-amino acid hydroxyethylamino sulfonamide retroviral protease inhibitors
US6617310B2 (en) * 2000-07-19 2003-09-09 Bristol-Myers Squibb Pharma Company Phosphate esters of bis-amino acid sulfonamides containing substituted benzyl amines
US20020022659A1 (en) * 2000-07-19 2002-02-21 Harris Gregory D. Crystalline and salt forms of an HIV protease inhibitor
US20020022742A1 (en) * 2000-07-19 2002-02-21 Harris Gregory D. Salt forms of an HIV protease inhibitor

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5843946A (en) * 1992-08-25 1998-12-01 G.D. Searle & Co. α-and β-amino acid hydroxyethylamino sulfonamides useful as retroviral protease inhibitors
ES2127938T3 (es) * 1993-08-24 1999-05-01 Searle & Co Hidroxietilamino sulfonamidas utiles como inhibidores de proteasas retroviricas.
US6150556A (en) * 1995-03-10 2000-11-21 G. D. Dearle & Co. Bis-amino acid hydroxyethylamino sulfonamide retroviral protease inhibitors

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO0042060A1 *

Also Published As

Publication number Publication date
TR200102033T2 (tr) 2001-12-21
NO20013338L (no) 2001-09-13
CN1336934A (zh) 2002-02-20
WO2000042060A1 (en) 2000-07-20
JP2002536298A (ja) 2002-10-29
HUP0105235A2 (hu) 2002-04-29
PL349774A1 (en) 2002-09-09
KR20010101478A (ko) 2001-11-14
SK9522001A3 (en) 2002-03-05
NO20013338D0 (no) 2001-07-05
BR0007854A (pt) 2002-01-15
IL143763A0 (en) 2002-04-21
AU2725700A (en) 2000-08-01
HUP0105235A3 (en) 2002-08-28
CA2353088A1 (en) 2000-07-20
CZ20012435A3 (cs) 2002-02-13

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
EP1140983A1 (en) Bis-amino acid sulfonamides containing n-terminally a fluoro-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
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
US6313110B1 (en) Substituted 2H-1,3-diazapin-2-one useful as an 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
US7015214B2 (en) Cyanamide, alkoxyamino, and urea derivatives of 1,3-benzodiazepine as HIV reverse transcriptase inhibitors
MXPA01007047A (en) Bis-amino acid sulfonamides containing n-terminally a substituted benzyl group as hiv protease 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

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20010622

AK Designated contracting states

Kind code of ref document: A1

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

AX Request for extension of the european patent

Free format text: AL;LT PAYMENT 20010622;LV PAYMENT 20010622;MK;RO PAYMENT 20010622;SI PAYMENT 20010622

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: BRISTOL-MYERS SQUIBB PHARMA COMPANY

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: BRISTOL-MYERS SQUIBB PHARMA COMPANY

17Q First examination report despatched

Effective date: 20030929

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

RTI1 Title (correction)

Free format text: BIS-AMINO ACID SULFONAMIDES CONTAINING N-TERMINALLY A FLUORO-SUBSTITUTED BENZYL GROUP AS HIV PROTEASE INHIBITORS

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20040930