WO2013043778A1 - Novel betulinic acid derivatives with antiviral activity - Google Patents

Novel betulinic acid derivatives with antiviral activity Download PDF

Info

Publication number
WO2013043778A1
WO2013043778A1 PCT/US2012/056186 US2012056186W WO2013043778A1 WO 2013043778 A1 WO2013043778 A1 WO 2013043778A1 US 2012056186 W US2012056186 W US 2012056186W WO 2013043778 A1 WO2013043778 A1 WO 2013043778A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
aids
hiv
alkyl
compound
Prior art date
Application number
PCT/US2012/056186
Other languages
French (fr)
Inventor
Zheng Liu
Nicholas A. Meanwell
Alicia Regueiro-Ren
Original Assignee
Bristol-Myers Squibb 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 MX2014002936A priority Critical patent/MX2014002936A/en
Application filed by Bristol-Myers Squibb Company filed Critical Bristol-Myers Squibb Company
Priority to AU2012312485A priority patent/AU2012312485B2/en
Priority to JP2014531936A priority patent/JP6100786B2/en
Priority to EA201490643A priority patent/EA023463B1/en
Priority to SG11201400331WA priority patent/SG11201400331WA/en
Priority to EP12766825.9A priority patent/EP2758419B8/en
Priority to BR112014006559A priority patent/BR112014006559A2/en
Priority to KR1020147010087A priority patent/KR20140069167A/en
Priority to CN201280046037.7A priority patent/CN103814042B/en
Priority to ES12766825.9T priority patent/ES2611727T3/en
Priority to CA2849475A priority patent/CA2849475A1/en
Publication of WO2013043778A1 publication Critical patent/WO2013043778A1/en
Priority to IL231421A priority patent/IL231421A/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J63/00Steroids in which the cyclopenta(a)hydrophenanthrene skeleton has been modified by expansion of only one ring by one or two atoms
    • C07J63/008Expansion of ring D by one atom, e.g. D homo steroids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates to novel compounds useful against HIV and, more particularly, to compounds derived from betulinic acid and other structurally-related compounds which are useful as HIV maturation inhibitors, and to pharmaceutical compositions containing same, as well as to methods for their preparation.
  • HrV-1 human immunodeficiency virus -1 infection
  • HIV and AIDS immunodeficiency syndrome
  • RT nucleoside reverse transcriptase
  • AZT or Retrovir ® didanosine
  • Videx ® stavudine
  • Zerit ® lamivudine
  • 3TC or Epivir ® zalcitabine
  • DDC or Hivid ® abacavir succinate (or Ziagen ® ), Tenofovir disoproxil fumarate salt (or Viread ® ), emtricitabine (or FTC - Emtriva®), Combivir ® (contains -3TC plus AZT), Trizivir ® (contains abacavir, lamivudine, and zidovudine), Epzicom ® (contains abacavir and lamivudine), Truvada ® (contains Viread ® and Emtriva®); non-nucleo
  • rilpivirine or Edurant®
  • nevirapine or Viramune ®
  • delavirdine or Rescriptor ®
  • efavirenz or Sustiva ®
  • Atripla ® Truvada ® + Sustiva ®
  • Complera® Truvada ® + Edurant ®
  • etravirine and peptidomimetic protease inhibitors or approved formulations: saquinavir, indinavir, ritonavir, nelfinavir, amprenavir, lopinavir, Kaletra (lopinavir and Ritonavir), darunavir, atazanavir (Reyataz ) and tipranavir
  • novel anti-HIV agents exhibiting distinct resistance patterns, and favorable pharmacokinetic as well as safety profiles are needed to provide more treatment options.
  • Improved HIV fusion inhibitors and HIV entry coreceptor antagonists are two examples of new classes of anti-HIV agents further being studied by a number of investigators.
  • HIV attachment inhibitors are a further subclass of antiviral compounds that bind to the HIV surface glycoprotein gpl20, and interfere with the interaction between the surface protein gpl20 and the host cell receptor CD4. Thus, they prevent HIV from attaching to the human CD4 T-cell, and block HIV replication in the first stage of the HIV life cycle.
  • the properties of HIV attachment inhibitors have been improved in an effort to obtain compounds with maximized utility and efficacy as antiviral agents.
  • US 7,354, 924 and US 2005/0209246 are illustrative of HIV attachment inhibitors.
  • Another emerging class of compounds for the treatment of HIV are called HIV maturation inhibitors.
  • Maturation is the last of as many as 10 or more steps in HIV replication or the HIV life cycle, in which HIV becomes infectious as a consequence of several HIV protease-mediated cleavage events in the gag protein that ultimately results in release of the capsid (CA) protein. Maturation inhibitors prevent the HIV capsid from properly assembling and maturing, from forming a protective outer coat, or from emerging from human cells. Instead, non-infectious viruses are produced, preventing subsequent cycles of HIV infection.
  • Bevirimat or PA-457 One HIV maturation compound that has been in development has been identified as Bevirimat or PA-457, with the chemical formula of C 36 H5 6 O 6 and the IUPAC name of 3 -(3-carboxy-3-methyl-butanoyloxy) lup-20(29)-en-28-oic acid.
  • the present invention provides compounds of Formula I below, including pharmaceutically acceptable salts thereof, their pharmaceutical formulations, and their use in patients suffering from or susceptible to a virus such as HIV.
  • the compounds of Formula I are effective antiviral agents, particularly as inhibitors of HIV. They are useful for the treatment of HIV and AIDS.
  • One embodiment of the present invention is directed to a compound of Formula I, including pharmaceutically acceptable salts thereof:
  • X is -O
  • Ri is -C(0)CH 2 C(CH 3 ) 2 COOH, -C(0)C(CH 3 ) 2 CH 2 COOH, or - C(0)CH 2 C(CH 3 ) 2 CH 2 COOH;
  • R 2 is selected from the group of -H, methyl, isopropenyl and isopropyl;
  • R 3 and R4 are independently selected from the group of -H, -C 1-6 alkyl, -Ci_6
  • R3 and R4 are taken together with the adjacent N to form a cycle selected from the group of:
  • R5 is independently -H or -Ci-6 alkyl
  • R 7 is selected from the group of -C e alkyl, -Ci_6 alkylsubstituted alkyl, -C3-6 cycloalkyl and aryl;
  • Rs and R 9 are independently selected from the group of -H, -C 1-6 alkyl, and -C e alkylsubstituted alkyl.
  • a method for treating mammals infected with a virus, especially wherein said virus is HIV comprising administering to said mammal an antiviral effective amount of a compound of Formula I above, and one or more pharmaceutically acceptable carriers, excipients or diluents.
  • the compound of Formula I can be administered in combination with an antiviral effective amount of another AIDS treatment agent selected from the group consisting of: (a) an AIDS antiviral agent; (b) an anti-infective agent; (c) an immunomodulator; and (d) other HIV entry inhibitors.
  • Another embodiment of the present invention is a pharmaceutical composition
  • a pharmaceutical composition comprising an antiviral effective amount of a compound of Formula I, and one or more pharmaceutically acceptable carriers, excipients, and diluents; and optionally in combination with an antiviral effective amount of another AIDS treatment agent selected from the group consisting of: (a) an AIDS antiviral agent; (b) an anti-infective agent; (c) an immunomodulator; and (d) other HIV entry inhibitors.
  • another AIDS treatment agent selected from the group consisting of: (a) an AIDS antiviral agent; (b) an anti-infective agent; (c) an immunomodulator; and (d) other HIV entry inhibitors.
  • the present disclosure includes the individual diastereoisomeric and enantiomeric forms of the compounds of Formula I, in addition to the mixtures thereof.
  • H refers to hydrogen, including its isotopes, such as deuterium.
  • C l _ 6 alkyl as used herein and in the claims (unless specified otherwise) mean straight or branched chain alkyl groups such as methyl, ethyl, propyl, isopropyl, butyl, isobutyl, t-butyl, amyl, hexyl and the like.
  • C j -C 4 fluoroalkyl refers to F-substituted C x -C 4 alkyl wherein at least one H atom is substituted with F atom, and each H atom can be independently substituted by F atom;
  • Halogen refers to chlorine, bromine, iodine or fluorine.
  • aryl refers to an all carbon monocyclic or fused-ring polycyclic (i.e., rings which share adjacent pairs of carbon atoms) groups having a completely conjugated pi-electron system. Examples, without limitation, of aryl groups are phenyl, napthalenyl and anthracenyl. The aryl group may be substituted or unsubstituted.
  • the substituted group(s) is preferably one or more selected from alkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclic, hydroxy, alkoxy, aryloxy, heteroaryloxy, heteroalicycloxy, thiohydroxy, thioaryloxy, thioheteroaryloxy, thioheteroalicycloxy, cyano, halogen, nitro, carbonyl, O-carbamyl, N-carbamyl, C-amido, N-amido, C-carboxy, O-carboxy, sulfinyl, sulfonyl, sulfonamido, trihalomethyl, ureido, amino and -NR x R y , wherein R x and R y are independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, carbonyl, C-carboxy, sulfonyl, trihalomethyl,
  • heteroaryl refers to a monocyclic or fused ring (i.e., rings which share an adjacent pair of atoms) group having in the ring(s) one or more atoms selected from the group consisting of nitrogen, oxygen and sulfur and, in addition, having a completely conjugated pi-electron system. Unless otherwise indicated, the heteroaryl group may be attached at either a carbon or nitrogen atom within the heteroaryl group. It should be noted that the term heteroaryl is intended to encompass an N-oxide of the parent heteroaryl if such an N-oxide is chemically feasible as is known in the art.
  • heteroaryl groups are furyl, thienyl, benzothienyl, thiazolyl, imidazolyl, oxazolyl, oxadiazolyl, thiadiazolyl, benzothiazolyl, triazolyl, tetrazolyl, isoxazolyl, isothiazolyl, pyrrolyl, pyranyl, tetrahydropyranyl, pyrazolyl, pyridyl, pyrimidinyl, quinolinyl, isoquinolinyl, purinyl, carbazolyl, benzoxazolyl, benzimidazolyl, indolyl, isoindolyl, pyrazinyl.
  • the substituted group(s) is preferably one or more selected from alkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclic, hydroxy, alkoxy, aryloxy, heteroaryloxy, heteroalicycloxy, thioalkoxy, thiohydroxy, thioaryloxy, thioheteroaryloxy, thioheteroalicycloxy, cyano, halogen, nitro, carbonyl, O-carbamyl, N-carbamyl, C-amido, N-amido, C-carboxy, O-carboxy, sulfinyl, sulfonyl, sulfonamido, trihalomethyl, ureido, amino, and -NR x R y , wherein R x and R y are as defined above.
  • a heteroalicyclic group refers to a monocyclic or fused ring group having in the ring(s) one or more atoms selected from the group consisting of nitrogen, oxygen and sulfur. Rings are selected from those which provide stable arrangements of bonds and are not intended to encompass systems which would not exist. The rings may also have one or more double bonds. However, the rings do not have a completely conjugated pi-electron system.
  • heteroalicyclic groups examples, without limitation, of heteroalicyclic groups are azetidinyl, piperidyl, piperazinyl, imidazolinyl, thiazolidinyl, 3- pyrrolidin-l-yl, morpholinyl, thiomorpholinyl and tetrahydropyranyl.
  • the substituted group(s) is preferably one or more selected from alkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclic, hydroxy, alkoxy, aryloxy, heteroaryloxy, heteroalicycloxy, thiohydroxy, thioalkoxy, thioaryloxy, thioheteroaryloxy, thioheteroalicycloxy, cyano, halogen, nitro, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N- thiocarbamyl, C-amido, C-thioamido, N-amido, C-carboxy, O-carboxy, sulfinyl, sulfonyl, sulfonamido, trihalomethanesulfonamido, trihalomethanesulfonyl, silyl, guanyl, guanidino,
  • alkyl group refers to a saturated aliphatic hydrocarbon including straight chain and branched chain groups.
  • the alkyl group has 1 to 20 carbon atoms (whenever a numerical range; e.g., "1-20", is stated herein, it means that the group, in this case the alkyl group may contain 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc. up to and including 20 carbon atoms). More preferably, it is a medium size alkyl having 1 to 10 carbon atoms. Most preferably, it is a lower alkyl having 1 to 4 carbon atoms.
  • the alkyl group may be substituted or unsubstituted.
  • the substituent group(s) is preferably one or more individually selected from trihaloalkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclic, hydroxy, alkoxy, aryloxy, heteroaryloxy,
  • thioheteroalicycloxy cyano, halo, nitro, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, C-thioamido, N-amido, C-carboxy, O- carboxy, sulfinyl, sulfonyl, sulfonamido, trihalomethanesulfonamido,
  • a "cycloalkyl” group refers to an all-carbon monocyclic or fused ring (i.e., rings which share and adjacent pair of carbon atoms) group wherein one or more rings does not have a completely conjugated pi-electron system.
  • examples, without limitation, of cycloalkyl groups are cyclopropane, cyclobutane, cyclopentane, cyclopentene, cyclohexane, cyclohexene, cycloheptane, cycloheptene and adamantane.
  • a cycloalkyl group may be substituted or unsubstituted.
  • the substituent group(s) is preferably one or more individually selected from alkyl, aryl, heteroaryl, heteroalicyclic, hydroxy, alkoxy, aryloxy, heteroaryloxy, heteroalicycloxy, thiohydroxy, thioalkoxy, thioaryloxy, thioheteroaryloxy, thioheteroalicycloxy, cyano, halo, nitro, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, C- thioamido, N-amido, C-carboxy, O-carboxy, sulfinyl, sulfonyl, sulfonamido, trihalo- methanesulfonamido, trihalomethanesulfonyl, silyl, amidino, guanidino, ureid
  • alkenyl refers to an alkyl group, as defined herein, having at least two carbon atoms and at least one carbon-carbon double bond.
  • alkynyl group refers to an alkyl group, as defined herein, having at least two carbon atoms and at least one carbon-carbon triple bond.
  • a "hydroxy” group refers to an -OH group.
  • alkoxy refers to both an -O-alkyl and an -O-cycloalkyl group as defined herein.
  • aryloxy refers to both an -O-aryl and an -O-heteroaryl group, as defined herein.
  • heteroaryloxy refers to a heteroaryl-O- group with heteroaryl as defined herein.
  • heteroalicycloxy refers to a heteroalicyclic-O- group with
  • a "thiohydroxy” group refers to an -SH group.
  • a “thioalkoxy” group refers to both an S-alkyl and an -S-cycloalkyl group, defined herein.
  • a “thioaryloxy” group refers to both an -S-aryl and an -S-heteroaryl group, as defined herein.
  • a “thioheteroaryloxy” group refers to a heteroaryl-S- group with heteroaryl as defined herein.
  • a “thioheteroalicycloxy” group refers to a heteroalicyclic-S- group with heteroalicyclic as defined herein.
  • An “aldehyde” group refers to a carbonyl group where R" is hydrogen.
  • O-carboxy refers to a R"C(-0)0-group, with R" as defined herein.
  • a “carboxylic acid” group refers to a C-carboxy group in which R" is hydrogen.
  • a “trihalomethyl” group refers to a -CZ 3 , group wherein Z is a halogen group as defined herein.
  • a “trihalomethanesulfonyl” group refers to an groups with Z as defined above.
  • a “trihalomethanesulfonamido” group refers to a group with Z as defined above and R x being H or (Ci-6)alkyl.
  • An “amino” group refers to an -NH 2 group.
  • a "cyano" group refers to a -CN group.
  • a “silyl” group refers to a -Si(R")3, with R" being (Ci_6)alkyl or phenyl.
  • a “hydrazino” group refers to a -NR x NR y R y2 group, with R x , R y , and R y2 independently being H or (Ci-6)alkyl.
  • a "4, 5, or 6 membered ring cyclic N-lactam" group refers to
  • Any two adjacent R groups may combine to form an additional aryl, cycli heteroaryl or heterocyclic ring fused to the ring initially bearing those R groups. It is known in the art that nitrogen atoms in heteroaryl systems can be
  • salts and prodrugs of compounds disclosed herein are within the scope of the invention.
  • pharmaceutically acceptable salt as used herein and in the claims is intended to include nontoxic base addition salts. Suitable salts include those derived from organic and inorganic acids such as, without limitation, hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid, methanesulfonic acid, acetic acid, tartaric acid, lactic acid, sulfinic acid, citric acid, maleic acid, fumaric acid, sorbic acid, aconitic acid, salicylic acid, phthalic acid, and the like.
  • organic and inorganic acids such as, without limitation, hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid, methanesulfonic acid, acetic acid, tartaric acid, lactic acid, sulfinic acid, citric acid, maleic acid, fumaric acid, sorbic acid, aconitic acid, salicylic acid, phthalic acid, and
  • pharmaceutically acceptable salt as used herein is also intended to include salts of acidic groups, such as a carboxylate, with such counterions as ammonium, alkali metal salts, particularly sodium or potassium, alkaline earth metal salts, particularly calcium or magnesium, and salts with suitable organic bases such as lower alkylamines
  • substituted lower alkylamines e.g. hydroxyl-substituted alkylamines such as diethanolamine
  • the compounds of the invention also include “prodrugs".
  • prodrug as used herein encompasses both the term “prodrug esters” and the term “prodrug ethers”.
  • prodrug esters as employed herein includes esters and carbonates formed by reacting one or more hydroxyls of compounds of Formula I with either alkyl, alkoxy, or aryl substituted acylating agents or phosphorylating agent employing procedures known to those skilled in the art to generate acetates, pivalates, methylcarbonates, benzoates, amino acid esters, phosphates, half acid esters such as malonates, succinates or glutarates, and the like. In certain embodiments, amino acid esters may be especially preferred.
  • X is -O
  • Ri is -C(0)CH 2 C(CH 3 ) 2 COOH, -C(0)C(CH 3 ) 2 CH 2 COOH, or - C(0)CH 2 C(CH 3 ) 2 CH 2 COOH;
  • R 2 is selected from the group of -H, methyl, isopropenyl and isopropyl;
  • R 3 and R4 are independently selected from the group of -H, -Ci_6 alkyl, -Ci_6
  • R5 is independently -H or -Ci_6 alkyl
  • R6 is selected from the group of -S0 2 R7, -S0 2 RsR9
  • R7 is selected from the group of -Ci-6 alkyl, -Ci-6 alkylsubstituted alkyl, -C3-6 cycloalkyl and aryl;
  • Rs and R 9 are independently selected from the group of -H, -Ci_6 alkyl, and -Ci_6 alkylsubstituted alkyl.
  • More preferred compounds of Formula I include those wherein a is 1. Also preferred are compounds wherein Ri is -C(0)CH 2 C(CH 3 ) 2 COOH.
  • a preferred compound of Formula I has the structural formula:
  • embodiments described above may be administered orally, parenterally (including subcutaneous injections, intravenous, intramuscular, intrasternal injection or infusion techniques), by inhalation spray, or rectally, and by other means, in dosage unit formulations containing non-toxic pharmaceutically acceptable carriers, excipients and diluents available to the skilled artisan.
  • One or more adjuvants may also be included.
  • a method of treatment for treating viral infections such as HIV infection and AIDS.
  • the treatment involves administering to a patient in need of such treatment a pharmaceutical composition which contains an antiviral effective amount of one or more of the compounds of Formula I, together with one or more pharmaceutically acceptable carriers, excipients or diluents.
  • antiviral effective amount means the total amount of each active component of the composition and method that is sufficient to show a meaningful patient benefit, i.e., inhibiting, ameliorating, or healing of acute conditions characterized by inhibition of the HIV infection.
  • the term refers to that ingredient alone.
  • the term refers to combined amounts of the active ingredients that result in the therapeutic effect, whether administered in combination, serially or simultaneously.
  • the terms "treat, treating, treatment” as used herein and in the claims means preventing, ameliorating or healing diseases associated with HIV infection.
  • compositions of the invention may be in the form of orally administrable suspensions or tablets; as well as nasal sprays, sterile injectable preparations, for example, as sterile injectable aqueous or oleaginous suspensions or suppositories.
  • Pharmaceutically acceptable carriers, excipients or diluents may be utilized in the pharmaceutical compositions, and are those utilized in the art of pharmaceutical preparations.
  • these compositions When administered orally as a suspension, these compositions are prepared according to techniques typically known in the art of pharmaceutical formulation and may contain microcrystalline cellulose for imparting bulk, alginic acid or sodium alginate as a suspending agent, methylcellulose as a viscosity enhancer, and sweeteners/flavoring agents known in the art.
  • these compositions may contain microcrystalline cellulose, dicalcium phosphate, starch, magnesium stearate and lactose and/or other excipients, binders, extenders, disintegrants, diluents, and lubricants known in the art.
  • the injectable solutions or suspensions may be formulated according to known art, using suitable non-toxic, parenterally acceptable diluents or solvents, such as mannitol, 1,3-butanediol, water, Ringer's solution or isotonic sodium chloride solution, or suitable dispersing or wetting and suspending agents, such as sterile, bland, fixed oils, including synthetic mono- or diglycerides, and fatty acids, including oleic acid.
  • suitable non-toxic, parenterally acceptable diluents or solvents such as mannitol, 1,3-butanediol, water, Ringer's solution or isotonic sodium chloride solution, or suitable dispersing or wetting and suspending agents, such as sterile, bland, fixed oils, including synthetic mono- or diglycerides, and fatty acids, including oleic acid.
  • the compounds herein set forth can be administered orally to humans in a dosage range of about 1 to 100 mg/kg body weight in divided doses, usually over an extended period, such as days, weeks, months, or even years.
  • One preferred dosage range is about 1 to 10 mg/kg body weight orally in divided doses.
  • Another preferred dosage range is about 1 to 20 mg/kg body weight in divided doses. It will be understood, however, that the specific dose level and frequency of dosage for any particular patient may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the host undergoing therapy.
  • the compounds of this disclosure may be effectively administered, whether at periods of pre-exposure and/or post-exposure, in combination with effective amounts of the AIDS antivirals, immunomodulators, antiinfectives, or vaccines, such as those in the following non-limiting table:
  • Famciclovir Smith Kline herpes zoster Famciclovir Smith Kline herpes zoster
  • ARC asymptomatic HIV positive, also in combination with AZT/ddl/ddC
  • Virazole Viratek/ICN asymptomatic HIV Ribavirin (Costa Mesa, CA) positive, LAS, ARC
  • VX-478 Vertex HIV infection, AIDS,
  • Emtriva R (Emtricitabine) Gilead HIV infection
  • Interleukin-2 CD4 cell counts (aldeslukin) Immune Globulin Cutter Biological Pediatric AIDS, in
  • Tumor Necrosis Genentech ARC in combination Factor; TNF w/gamma Interferon
  • HIV entry inhibitors examples include DRUGS OF THE FUTURE 1999, 24(12), pp. 1355-1362; CELL, Vol. 9, pp.
  • HIV attachment inhibitors are also set forth in US
  • Preferred combinations are simultaneous or alternating treatments with a compound of the present disclosure and an inhibitor of HIV protease and/or a non- nucleoside inhibitor of HIV reverse transcriptase.
  • An optional fourth component in the combination is a nucleoside inhibitor of HIV reverse transcriptase, such as AZT, 3TC, ddC or ddl.
  • a preferred inhibitor of HIV protease is Reyataz ® (active ingredient Atazanavir). Typically a dose of 300 to 600mg is administered once a day. This may be co-administered with a low dose of Ritonavir (50 to 500mgs).
  • Another preferred inhibitor of HIV protease is Kaletra ® .
  • indinavir is the sulfate salt of N-(2(R)-hydroxy-l-(S)-indanyl)-2(R)-phenylmethyl- 4-(S)-hydroxy-5-(l-(4-(3-pyridyl-methyl)-2(S)-N'-(t-butylcarboxamido)-piperazinyl))- pentaneamide ethanolate, and is synthesized according to U.S. 5,413,999.
  • Indinavir is generally administered at a dosage of 800 mg three times a day.
  • Other preferred protease inhibitors are nelfinavir and ritonavir.
  • HIV protease is saquinavir which is administered in a dosage of 600 or 1200 mg tid.
  • Preferred non- nucleoside inhibitors of HIV reverse transcriptase include efavirenz. These combinations may have unexpected effects on limiting the spread and degree of infection of HIV.
  • Preferred combinations include those with the following (1) indinavir with efavirenz, and, optionally, AZT and/or 3TC and/or ddl and/or ddC; (2) indinavir, and any of AZT and/or ddl and/or ddC and/or 3TC, in particular, indinavir and AZT and 3TC; (3) stavudine and 3TC and/or zidovudine; (4) tenofovir disoproxil fumarate salt and emtricitabine.
  • the compound of the present invention and other active agents may be administered separately or in conjunction.
  • the administration of one element may be prior to, concurrent to, or subsequent to the administration of other agent(s).
  • the present invention comprises compounds of Formula I, their pharmaceutical formulations, and their use in patients suffering from or susceptible to HIV infection.
  • the compounds of Formula I also include pharmaceutically acceptable salts thereof.
  • General procedures to construct compounds of Formula I and intermediates useful for their synthesis are described in the following Schemes (after the Abbreviations).
  • TBTU 0-(benzotriazol- 1 -yl)-N,N,N',N'-tetramethyluronium tetrafluoroborate
  • HATU 2-(lH-7-azabenzotriazol-l-yl)-l, l,3,3-tetramethyluronium hexafluorophosphate methanaminium
  • DIPEA diisopropylethylamine
  • DMAP 4-dimethylaminopyridine
  • DMSO dimethylsulfoxide
  • PCC pyridinium chlorochromate
  • ⁇ g microgram(s)
  • Betulinic acid can be converted into the corresponding C-17 amine analog via Curtius' rearrangement.
  • Selective alkylation of the amine can be achieved by treatment with an alkylating agent (R-LG), where LG is a leaving group such as, but not limited to Br, CI, I, mesylate, tosylate or triflate in the presence of a base. Heating maybe needed in some cases. In some cases, by prolonging the reaction times and heating the reaction mixture, the dialkylated product can also be formed. Acylation of the C-3 hydroxyl can be achieved by heating in the presence of the corresponding anhydride.
  • R-LG alkylating agent
  • acylation can also be accomplished by addition of the corresponding carboxylic acid and a couping agent such as, but not limited to HATU or TBTU. Saturation of the double bond can be performed under standard conditions in any stage of the synthesis.
  • CDCI 3 ( ⁇ ⁇ 7.26), CD 3 OD ( ⁇ ⁇ 3.30), Acetic-d4 (Acetic Acid d 4 ) ( ⁇ ⁇ 1 1.6, 2.07), DMSO mix or DMS0-D6_CDC1 3 (( H 2.50 and 8.25) (ratio 75%:25%), and DMSO-D6 ( ⁇ ⁇ 2.50).
  • Step 1 Preparation of (lR,3aS,5aR,5bR,7aR,9S,llaR,llbR,13aR,13bR)-3a-amino- 5a,5b,8,8,l la-pentamethyl-l-(prop-l-en-2-yl)icosahydro-lH-cyclopenta [a] chrysen-9- ol
  • Step 2 Preparation of (lR,3aS,5aR,5bR,7aR,9S,HaR,HbR,13aR,13bR)-3a-((2-(l,l- dioxido-4-thiomorpholinyl)ethyl)amino)-l-isopropenyl-5a,5b,8,8,lla- pentamethylicosahydro-lH-cyclopenta [a] chrysen-9-ol
  • Step 3 Preparation of 4-(((lR,3aS,5aR,5bR,7aR,9S,llaR,13aR,13bR)-3a-((2 dioxido-4-thiomorpholinyl)ethyl)amino)-l-isopropenyl-5a,5b,8,8,lla- pentamethylicosahydro-lH-cyclopenta[a]chrysen-9-yl)oxy)-3,3-dimethyl-4- oxobutanoic acid
  • ⁇ " ⁇ " means microliter
  • HIV cell culture assay - MT-2 cells and 293T cells were obtained from the NIH AIDS Research and Reference Reagent Program.
  • MT-2 cells were propagated in RPMI 1640 media supplemented with 10% heat inactivated fetal bovine serum, 100 ⁇ g/ml penicillin G and up to 100 units/ml streptomycin.
  • the 293T cells were propagated in DMEM media supplemented with 10% heat inactivated fetal bovine serum (FBS), 100 units/ml penicillin G and 100 ⁇ g/ml streptomycin.
  • FBS heat inactivated fetal bovine serum
  • the proviral DNA clone of L 4 _3 was obtained from the NIH AIDS Research and Reference Reagent Program.
  • a recombinant NL 4 _3 virus in which a section of the nef gene from NL4-3 was replaced with the Renilla luciferase gene, was used as a reference virus.
  • residue Gag P373 was converted to P373S.
  • the recombinant virus was prepared by transfection of the altered proviral clone of NL 4 _3. Transfections were performed in 293T cells using LipofectAMINE PLUS from Invitrogen (Carlsbad, CA), according to manufacturer's instruction. The virus was titered in MT-2 cells using luciferase enzyme activity as a marker. Luciferase was quantitated using the Dual Luciferase kit from Promega

Abstract

Compounds of Formula I: that possess unique antiviral activity are provided as HIV maturation inhibitors. These compounds are useful for the treatment of HIV and AIDS.

Description

NOVEL BETULINIC ACID DERIVATIVES WITH ANTIVIRAL ACTIVITY
CROSS REFERENCE TO RELATED APPLICATION This application claims the benefit of priority from U.S. Provisional Application
Serial Number 61/537,099 filed September 21, 2011.
FIELD OF THE INVENTION The present invention relates to novel compounds useful against HIV and, more particularly, to compounds derived from betulinic acid and other structurally-related compounds which are useful as HIV maturation inhibitors, and to pharmaceutical compositions containing same, as well as to methods for their preparation. BACKGROUND OF THE INVENTION
HrV-1 (human immunodeficiency virus -1) infection remains a major medical problem, with an estimated 45-50 million people infected worldwide at the end of 2010. The number of cases of HIV and AIDS (acquired immunodeficiency syndrome) has risen rapidly. In 2005, approximately 5.0 million new infections were reported, and 3.1 million people died from AIDS. Currently available drugs for the treatment of HIV include nucleoside reverse transcriptase (RT) inhibitors or approved single pill combinations: zidovudine (or AZT or Retrovir®), didanosine (or Videx®), stavudine (or Zerit®), lamivudine (or 3TC or Epivir®), zalcitabine (or DDC or Hivid®), abacavir succinate (or Ziagen®), Tenofovir disoproxil fumarate salt (or Viread®), emtricitabine (or FTC - Emtriva®), Combivir® (contains -3TC plus AZT), Trizivir® (contains abacavir, lamivudine, and zidovudine), Epzicom® (contains abacavir and lamivudine), Truvada® (contains Viread® and Emtriva®); non-nucleoside reverse transcriptase inhibitors:
rilpivirine (or Edurant®), nevirapine (or Viramune®), delavirdine (or Rescriptor®) and efavirenz (or Sustiva®), Atripla® (Truvada® + Sustiva®), Complera® (Truvada® + Edurant® ), and etravirine, and peptidomimetic protease inhibitors or approved formulations: saquinavir, indinavir, ritonavir, nelfinavir, amprenavir, lopinavir, Kaletra (lopinavir and Ritonavir), darunavir, atazanavir (Reyataz ) and tipranavir
® ®
(Aptivus ), and integrase inhibitors such as raltegravir (Isentress ), and entry inhibitors
® ®
such as enfuvirtide (T-20) (Fuzeon ) and maraviroc (Selzentry ).
Each of these drugs can only transiently restrain viral replication if used alone. However, when used in combination, these drugs have a profound effect on viremia and disease progression. In fact, significant reductions in death rates among AIDS patients have been recently documented as a consequence of the widespread application of combination therapy. However, despite these impressive results, 30 to 50% of patients may ultimately fail combination drug therapies. Insufficient drug potency, non- compliance, restricted tissue penetration and drug-specific limitations within certain cell types (e.g. most nucleoside analogs cannot be phosphorylated in resting cells) may account for the incomplete suppression of sensitive viruses. Furthermore, the high replication rate and rapid turnover of HIV- 1 combined with the frequent incorporation of mutations, leads to the appearance of drug-resistant variants and treatment failures when sub-optimal drug concentrations are present. Therefore, novel anti-HIV agents exhibiting distinct resistance patterns, and favorable pharmacokinetic as well as safety profiles are needed to provide more treatment options. Improved HIV fusion inhibitors and HIV entry coreceptor antagonists are two examples of new classes of anti-HIV agents further being studied by a number of investigators.
HIV attachment inhibitors are a further subclass of antiviral compounds that bind to the HIV surface glycoprotein gpl20, and interfere with the interaction between the surface protein gpl20 and the host cell receptor CD4. Thus, they prevent HIV from attaching to the human CD4 T-cell, and block HIV replication in the first stage of the HIV life cycle. The properties of HIV attachment inhibitors have been improved in an effort to obtain compounds with maximized utility and efficacy as antiviral agents. In particular, US 7,354, 924 and US 2005/0209246 are illustrative of HIV attachment inhibitors. Another emerging class of compounds for the treatment of HIV are called HIV maturation inhibitors. Maturation is the last of as many as 10 or more steps in HIV replication or the HIV life cycle, in which HIV becomes infectious as a consequence of several HIV protease-mediated cleavage events in the gag protein that ultimately results in release of the capsid (CA) protein. Maturation inhibitors prevent the HIV capsid from properly assembling and maturing, from forming a protective outer coat, or from emerging from human cells. Instead, non-infectious viruses are produced, preventing subsequent cycles of HIV infection.
Certain derivatives of betulinic acid have now been shown to exhibit potent anti- HIV activity as HIV maturation inhibitors. For example, US 7,365,221 discloses monoacylated betulin and dihydrobetuline derivatives, and their use as anti-HIV agents. As discussed in the '221 reference, esterification of betulinic acid (1) with certain substituted acyl groups, such as 3',3'-dimethylglutaryl and 3',3'-dimethylsuccinyl groups produced derivatives having enhanced activity (Kashiwada, Y., et al., J. Med. Chem. 39: 1016-1017 (1996)). Acylated betulinic acid and dihydrobetulinic acid derivatives that are potent anti-HIV agents are also described in U.S. Pat. No. 5,679,828. Esterification of the hydroxyl in the 3 carbon of betulin with succinic acid also produced a compound capable of inhibiting HIV- 1 activity.
Other references to the use of treating HIV infection with compounds derived from betulinic acid include US 2005/0239748 and US 2008/0207573, as well as
WO2006/053255, WO2009/100532 and WO2011/007230.
One HIV maturation compound that has been in development has been identified as Bevirimat or PA-457, with the chemical formula of C36H56O6 and the IUPAC name of 3 -(3-carboxy-3-methyl-butanoyloxy) lup-20(29)-en-28-oic acid.
Reference is also made herein to the applications by Bristol-Myers Squibb entitled "MODIFIED C-3 BETULINIC ACID DERIVATIVES AS HIV MATURATION
INHIBITORS" USSN 13/151,706 filed on June 2, 2011 (US 20120142707A1) and "C-28 AMIDES OF MODIFIED C-3 BETULINIC ACID DERIVATIVES AS HIV
MATURATION INHIBITORS" USSN 13/151,722, filed on June 2, 2011
(US20120142653A1). Reference is also made to the applications entitled "C-28
AMINES OF C-3 MODIFIED BETULINIC ACID DERIVATIVES AS HIV
MATURATION INHIBITORS" USSN 13/359,680, filed on January 27, 2012, and "C-17 AND C-3 MODIFIED TRITERPENOIDS WITH HIV MATURATION INHIBITORY ACTIVITY" USSN 13/359,727, filed on January 27, 2012.
What is now needed in the art are new compounds which are useful as HIV maturation inhibitors, as well as new pharmaceutical compositions containing these compounds.
SUMMARY OF THE INVENTION
The present invention provides compounds of Formula I below, including pharmaceutically acceptable salts thereof, their pharmaceutical formulations, and their use in patients suffering from or susceptible to a virus such as HIV. The compounds of Formula I are effective antiviral agents, particularly as inhibitors of HIV. They are useful for the treatment of HIV and AIDS.
One embodiment of the present invention is directed to a compound of Formula I, including pharmaceutically acceptable salts thereof:
Figure imgf000005_0001
Formula I
wherein a is 1 to 4;
X is -O;
Ri is -C(0)CH2C(CH3)2COOH, -C(0)C(CH3)2CH2COOH, or - C(0)CH2C(CH3)2CH2COOH;
R2 is selected from the group of -H, methyl, isopropenyl and isopropyl;
R3 and R4 are independently selected from the group of -H, -C1-6 alkyl, -Ci_6
alkylsubstituted alkyl and -C3-6 cycloalkyl;
or R3 and R4 are taken together with the adjacent N to form a cycle selected from the group of:
Figure imgf000006_0001
R5 is independently -H or -Ci-6 alkyl;
5 is selected from the group of -SO2R7, -SO2 R8 9
R7 is selected from the group of -C e alkyl, -Ci_6 alkylsubstituted alkyl, -C3-6 cycloalkyl and aryl; and
Rs and R9 are independently selected from the group of -H, -C1-6 alkyl, and -C e alkylsubstituted alkyl.
In a further embodiment, there is provided a method for treating mammals infected with a virus, especially wherein said virus is HIV, comprising administering to said mammal an antiviral effective amount of a compound of Formula I above, and one or more pharmaceutically acceptable carriers, excipients or diluents. Optionally, the compound of Formula I can be administered in combination with an antiviral effective amount of another AIDS treatment agent selected from the group consisting of: (a) an AIDS antiviral agent; (b) an anti-infective agent; (c) an immunomodulator; and (d) other HIV entry inhibitors.
Another embodiment of the present invention is a pharmaceutical composition comprising an antiviral effective amount of a compound of Formula I, and one or more pharmaceutically acceptable carriers, excipients, and diluents; and optionally in combination with an antiviral effective amount of another AIDS treatment agent selected from the group consisting of: (a) an AIDS antiviral agent; (b) an anti-infective agent; (c) an immunomodulator; and (d) other HIV entry inhibitors. In another embodiment of the invention there is provided one or more methods for making the compounds of Formula I.
Also provided herein are intermediate compounds useful in making the compounds of Formula I. The present invention is directed to these, as well as other important ends, hereinafter described.
DETAILED DESCRIPTION OF THE EMBODIMENTS
Since the compounds of the present invention may possess asymmetric centers and therefore occur as mixtures of diastereomers and enantiomers, the present disclosure includes the individual diastereoisomeric and enantiomeric forms of the compounds of Formula I, in addition to the mixtures thereof.
Definitions
Unless otherwise specifically set forth elsewhere in the application, one or more of the following terms may be used herein, and shall have the following meanings:
"H" refers to hydrogen, including its isotopes, such as deuterium.
The term "Cl _6 alkyl" as used herein and in the claims (unless specified otherwise) mean straight or branched chain alkyl groups such as methyl, ethyl, propyl, isopropyl, butyl, isobutyl, t-butyl, amyl, hexyl and the like.
"Cj -C4 fluoroalkyl" refers to F-substituted Cx -C4 alkyl wherein at least one H atom is substituted with F atom, and each H atom can be independently substituted by F atom;
"Halogen" refers to chlorine, bromine, iodine or fluorine.
An "aryl" or "Ar" group refers to an all carbon monocyclic or fused-ring polycyclic (i.e., rings which share adjacent pairs of carbon atoms) groups having a completely conjugated pi-electron system. Examples, without limitation, of aryl groups are phenyl, napthalenyl and anthracenyl. The aryl group may be substituted or unsubstituted. When substituted the substituted group(s) is preferably one or more selected from alkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclic, hydroxy, alkoxy, aryloxy, heteroaryloxy, heteroalicycloxy, thiohydroxy, thioaryloxy, thioheteroaryloxy, thioheteroalicycloxy, cyano, halogen, nitro, carbonyl, O-carbamyl, N-carbamyl, C-amido, N-amido, C-carboxy, O-carboxy, sulfinyl, sulfonyl, sulfonamido, trihalomethyl, ureido, amino and -NRxRy, wherein Rx and Ry are independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, carbonyl, C-carboxy, sulfonyl, trihalomethyl, and, combined, a five- or six-member heteroalicyclic ring.
As used herein, a "heteroaryl" group refers to a monocyclic or fused ring (i.e., rings which share an adjacent pair of atoms) group having in the ring(s) one or more atoms selected from the group consisting of nitrogen, oxygen and sulfur and, in addition, having a completely conjugated pi-electron system. Unless otherwise indicated, the heteroaryl group may be attached at either a carbon or nitrogen atom within the heteroaryl group. It should be noted that the term heteroaryl is intended to encompass an N-oxide of the parent heteroaryl if such an N-oxide is chemically feasible as is known in the art. Examples, without limitation, of heteroaryl groups are furyl, thienyl, benzothienyl, thiazolyl, imidazolyl, oxazolyl, oxadiazolyl, thiadiazolyl, benzothiazolyl, triazolyl, tetrazolyl, isoxazolyl, isothiazolyl, pyrrolyl, pyranyl, tetrahydropyranyl, pyrazolyl, pyridyl, pyrimidinyl, quinolinyl, isoquinolinyl, purinyl, carbazolyl, benzoxazolyl, benzimidazolyl, indolyl, isoindolyl, pyrazinyl. diazinyl, pyrazine, triazinyl, tetrazinyl, and tetrazolyl. When substituted the substituted group(s) is preferably one or more selected from alkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclic, hydroxy, alkoxy, aryloxy, heteroaryloxy, heteroalicycloxy, thioalkoxy, thiohydroxy, thioaryloxy, thioheteroaryloxy, thioheteroalicycloxy, cyano, halogen, nitro, carbonyl, O-carbamyl, N-carbamyl, C-amido, N-amido, C-carboxy, O-carboxy, sulfinyl, sulfonyl, sulfonamido, trihalomethyl, ureido, amino, and -NRxRy, wherein Rx and Ry are as defined above.
As used herein, a "heteroalicyclic" group refers to a monocyclic or fused ring group having in the ring(s) one or more atoms selected from the group consisting of nitrogen, oxygen and sulfur. Rings are selected from those which provide stable arrangements of bonds and are not intended to encompass systems which would not exist. The rings may also have one or more double bonds. However, the rings do not have a completely conjugated pi-electron system. Examples, without limitation, of heteroalicyclic groups are azetidinyl, piperidyl, piperazinyl, imidazolinyl, thiazolidinyl, 3- pyrrolidin-l-yl, morpholinyl, thiomorpholinyl and tetrahydropyranyl. When substituted the substituted group(s) is preferably one or more selected from alkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclic, hydroxy, alkoxy, aryloxy, heteroaryloxy, heteroalicycloxy, thiohydroxy, thioalkoxy, thioaryloxy, thioheteroaryloxy, thioheteroalicycloxy, cyano, halogen, nitro, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N- thiocarbamyl, C-amido, C-thioamido, N-amido, C-carboxy, O-carboxy, sulfinyl, sulfonyl, sulfonamido, trihalomethanesulfonamido, trihalomethanesulfonyl, silyl, guanyl, guanidino, ureido, phosphonyl, amino and -NRxRy, wherein Rx and Ry are as defined above.
An "alkyl" group refers to a saturated aliphatic hydrocarbon including straight chain and branched chain groups. Preferably, the alkyl group has 1 to 20 carbon atoms (whenever a numerical range; e.g., "1-20", is stated herein, it means that the group, in this case the alkyl group may contain 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc. up to and including 20 carbon atoms). More preferably, it is a medium size alkyl having 1 to 10 carbon atoms. Most preferably, it is a lower alkyl having 1 to 4 carbon atoms. The alkyl group may be substituted or unsubstituted. When substituted, the substituent group(s) is preferably one or more individually selected from trihaloalkyl, cycloalkyl, aryl, heteroaryl, heteroalicyclic, hydroxy, alkoxy, aryloxy, heteroaryloxy,
heteroalicycloxy, thiohydroxy, thioalkoxy, thioaryloxy, thioheteroaryloxy,
thioheteroalicycloxy, cyano, halo, nitro, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, C-thioamido, N-amido, C-carboxy, O- carboxy, sulfinyl, sulfonyl, sulfonamido, trihalomethanesulfonamido,
trihalomethanesulfonyl, and combined, a five- or six-member heteroalicyclic ring.
A "cycloalkyl" group refers to an all-carbon monocyclic or fused ring (i.e., rings which share and adjacent pair of carbon atoms) group wherein one or more rings does not have a completely conjugated pi-electron system. Examples, without limitation, of cycloalkyl groups are cyclopropane, cyclobutane, cyclopentane, cyclopentene, cyclohexane, cyclohexene, cycloheptane, cycloheptene and adamantane. A cycloalkyl group may be substituted or unsubstituted. When substituted, the substituent group(s) is preferably one or more individually selected from alkyl, aryl, heteroaryl, heteroalicyclic, hydroxy, alkoxy, aryloxy, heteroaryloxy, heteroalicycloxy, thiohydroxy, thioalkoxy, thioaryloxy, thioheteroaryloxy, thioheteroalicycloxy, cyano, halo, nitro, carbonyl, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, C- thioamido, N-amido, C-carboxy, O-carboxy, sulfinyl, sulfonyl, sulfonamido, trihalo- methanesulfonamido, trihalomethanesulfonyl, silyl, amidino, guanidino, ureido, phosphonyl, amino and -NRxRy with Rx and Ry as defined above.
An "alkenyl" group refers to an alkyl group, as defined herein, having at least two carbon atoms and at least one carbon-carbon double bond.
An "alkynyl" group refers to an alkyl group, as defined herein, having at least two carbon atoms and at least one carbon-carbon triple bond.
A "hydroxy" group refers to an -OH group.
An "alkoxy" group refers to both an -O-alkyl and an -O-cycloalkyl group as defined herein.
An "aryloxy" group refers to both an -O-aryl and an -O-heteroaryl group, as defined herein.
A "heteroaryloxy" group refers to a heteroaryl-O- group with heteroaryl as defined herein. A "heteroalicycloxy" group refers to a heteroalicyclic-O- group with
heteroalicyclic as defined herein.
A "thiohydroxy" group refers to an -SH group.
A "thioalkoxy" group refers to both an S-alkyl and an -S-cycloalkyl group, defined herein. A "thioaryloxy" group refers to both an -S-aryl and an -S-heteroaryl group, as defined herein.
A "thioheteroaryloxy" group refers to a heteroaryl-S- group with heteroaryl as defined herein.
A "thioheteroalicycloxy" group refers to a heteroalicyclic-S- group with heteroalicyclic as defined herein. A "carbonyl" group refers to a -C(=0)-R" group, where R" is selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heteroalicyclic (bonded through a ring carbon), as each is defined herein. An "aldehyde" group refers to a carbonyl group where R" is hydrogen.
A "thiocarbonyl" group refers to a -C(=S)-R" group, with R" as defined herein.
A "Keto" group refers to a -CC(=0)C- group wherein the carbon on either or both sides of the C=0 may be alkyl, cycloalkyl, aryl or a carbon of a heteroaryl or heteroalicyclic group.
A "trihalomethanecarbonyl" group refers to a Z3CC(=0)- group with said Z being a halogen.
A "C-carboxy" group refers to a -C(=0)0-R" groups, with R" as defined herein.
An "O-carboxy" group refers to a R"C(-0)0-group, with R" as defined herein. A "carboxylic acid" group refers to a C-carboxy group in which R" is hydrogen.
A "trihalomethyl" group refers to a -CZ3, group wherein Z is a halogen group as defined herein. A "trihalomethanesulfonyl" group refers to an
Figure imgf000012_0001
groups with Z as defined above. A "trihalomethanesulfonamido" group refers to a
Figure imgf000012_0002
group with Z as defined above and Rx being H or (Ci-6)alkyl.
A "sulfinyl" group refers to a -S(=0)-R" group, with R" being (Ci_6)alkyl. A "sulfonyl" group refers to a -S(=0)2R" group with R" being (C1-6)alky 1.
A "S-sulfonamido" group refers to a -S(=0)2NRxRY, with Rx and RY independently being H or (C^alkyl. A "N-Sulfonamido" group refers to a R"S(=0)2NRx- group, with Rx being H or
(Ci-6)alkyl.
A "O-carbamyl" group refers to a -OC(=0)NRxRy group, with Rx and RY independently being H or (C^alkyl.
A "N-carbamyl" group refers to a RxOC(=0)NRy group, with Rx and Ry independently being H or (C^alkyl.
A "O-thiocarbamyl" group refers to a -OC(=S)NRxRy group, with Rx and Ry independently being H or (C^alkyl.
A "N-thiocarbamyl" group refers to a RxOC(=S)NRy- group, with Rx and Ry independently being H or (C^alkyl. An "amino" group refers to an -NH2 group.
A "C-amido" group refers to a -C(=0)NRxRy group, with Rx and Ry
independently being H or (C^alkyl. A "C-thioamido" group refers to a -C(=S)NRxRy group, with Rx and Ry independently being H or (Ci_6)alkyl. A "N-amido" group refers to a RxC(=0)NRy- group, with Rx and Ry
independently being H or (Ci_6)alkyl.
An "ureido" group refers to a -NRxC(=0)NRyRy2 group, with Rx, Ry, and Ry2 independently being H or (Ci_6)alkyl.
A "guanidino" group refers to a -RxNC(=N)NRyRy2 group, with Rx, Ry, and Ry2 independently being H or (Ci_6)alkyl.
A "amidino" group refers to a RxRyNC(=N)- group, with Rx and Ry independently being H or (Ci-6)alkyl.
A "cyano" group refers to a -CN group.
A "silyl" group refers to a -Si(R")3, with R" being (Ci_6)alkyl or phenyl.
A "phosphonyl" group refers to a P(=0)(ORx)2 with Rx being (C1-6)alkyl.
A "hydrazino" group refers to a -NRxNRyRy2 group, with Rx, Ry, and Ry2 independently being H or (Ci-6)alkyl.
A "4, 5, or 6 membered ring cyclic N-lactam" group refers to
Figure imgf000013_0001
Any two adjacent R groups may combine to form an additional aryl, cycli heteroaryl or heterocyclic ring fused to the ring initially bearing those R groups. It is known in the art that nitrogen atoms in heteroaryl systems can be
"participating in a heteroaryl ring double bond", and this refers to the form of double bonds in the two tautomeric structures which comprise five-member ring heteroaryl groups. This dictates whether nitrogens can be substituted as well understood by chemists in the art. The disclosure and claims of the present disclosure are based on the known general principles of chemical bonding. It is understood that the claims do not encompass structures known to be unstable or not able to exist based on the literature.
Pharmaceutically acceptable salts and prodrugs of compounds disclosed herein are within the scope of the invention. The term "pharmaceutically acceptable salt" as used herein and in the claims is intended to include nontoxic base addition salts. Suitable salts include those derived from organic and inorganic acids such as, without limitation, hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid, methanesulfonic acid, acetic acid, tartaric acid, lactic acid, sulfinic acid, citric acid, maleic acid, fumaric acid, sorbic acid, aconitic acid, salicylic acid, phthalic acid, and the like. The term
"pharmaceutically acceptable salt" as used herein is also intended to include salts of acidic groups, such as a carboxylate, with such counterions as ammonium, alkali metal salts, particularly sodium or potassium, alkaline earth metal salts, particularly calcium or magnesium, and salts with suitable organic bases such as lower alkylamines
(methylamine, ethylamine, cyclohexylamine, and the like) or with substituted lower alkylamines (e.g. hydroxyl-substituted alkylamines such as diethanolamine,
triethanolamine or tris(hydroxymethyl)- aminomethane), or with bases such as piperidine or morpholine. As stated above, the compounds of the invention also include "prodrugs". The term "prodrug" as used herein encompasses both the term "prodrug esters" and the term "prodrug ethers". The term "prodrug esters" as employed herein includes esters and carbonates formed by reacting one or more hydroxyls of compounds of Formula I with either alkyl, alkoxy, or aryl substituted acylating agents or phosphorylating agent employing procedures known to those skilled in the art to generate acetates, pivalates, methylcarbonates, benzoates, amino acid esters, phosphates, half acid esters such as malonates, succinates or glutarates, and the like. In certain embodiments, amino acid esters may be especially preferred.
As set forth above, the invention is directed to compounds of Formula I, including pharmaceutically acceptable salts thereof:
Figure imgf000015_0001
Formula I
wherein a is 1 to 4;
X is -O;
Ri is -C(0)CH2C(CH3)2COOH, -C(0)C(CH3)2CH2COOH, or - C(0)CH2C(CH3)2CH2COOH;
R2 is selected from the group of -H, methyl, isopropenyl and isopropyl;
R3 and R4 are independently selected from the group of -H, -Ci_6 alkyl, -Ci_6
alkylsubstituted alkyl and -C3-6 cycloalkyl;
or R3 and R4 are taken together with the adjacent N to form a cycle selected from the group of:
Figure imgf000015_0002
R5 is independently -H or -Ci_6 alkyl;
R6 is selected from the group of -S02R7, -S02 RsR9
R7 is selected from the group of -Ci-6 alkyl, -Ci-6 alkylsubstituted alkyl, -C3-6 cycloalkyl and aryl; and
Rs and R9 are independently selected from the group of -H, -Ci_6 alkyl, and -Ci_6 alkylsubstituted alkyl.
More preferred compounds of Formula I include those wherein a is 1. Also preferred are compounds wherein Ri is -C(0)CH2C(CH3)2COOH. A preferred compound of Formula I has the structural formula:
Figure imgf000016_0001
Other compounds which are preferred as part of the invention include the following:
Figure imgf000016_0002
Figure imgf000017_0001
Figure imgf000018_0001
, including pharmaceutically acceptable salts thereof.
The compounds of the present invention, according to all the various
embodiments described above, may be administered orally, parenterally (including subcutaneous injections, intravenous, intramuscular, intrasternal injection or infusion techniques), by inhalation spray, or rectally, and by other means, in dosage unit formulations containing non-toxic pharmaceutically acceptable carriers, excipients and diluents available to the skilled artisan. One or more adjuvants may also be included.
Thus, in accordance with the present invention, there is further provided a method of treatment, and a pharmaceutical composition, for treating viral infections such as HIV infection and AIDS. The treatment involves administering to a patient in need of such treatment a pharmaceutical composition which contains an antiviral effective amount of one or more of the compounds of Formula I, together with one or more pharmaceutically acceptable carriers, excipients or diluents. As used herein, the term "antiviral effective amount" means the total amount of each active component of the composition and method that is sufficient to show a meaningful patient benefit, i.e., inhibiting, ameliorating, or healing of acute conditions characterized by inhibition of the HIV infection. When applied to an individual active ingredient, administered alone, the term refers to that ingredient alone. When applied to a combination, the term refers to combined amounts of the active ingredients that result in the therapeutic effect, whether administered in combination, serially or simultaneously. The terms "treat, treating, treatment" as used herein and in the claims means preventing, ameliorating or healing diseases associated with HIV infection.
The pharmaceutical compositions of the invention may be in the form of orally administrable suspensions or tablets; as well as nasal sprays, sterile injectable preparations, for example, as sterile injectable aqueous or oleaginous suspensions or suppositories. Pharmaceutically acceptable carriers, excipients or diluents may be utilized in the pharmaceutical compositions, and are those utilized in the art of pharmaceutical preparations.
When administered orally as a suspension, these compositions are prepared according to techniques typically known in the art of pharmaceutical formulation and may contain microcrystalline cellulose for imparting bulk, alginic acid or sodium alginate as a suspending agent, methylcellulose as a viscosity enhancer, and sweeteners/flavoring agents known in the art. As immediate release tablets, these compositions may contain microcrystalline cellulose, dicalcium phosphate, starch, magnesium stearate and lactose and/or other excipients, binders, extenders, disintegrants, diluents, and lubricants known in the art.
The injectable solutions or suspensions may be formulated according to known art, using suitable non-toxic, parenterally acceptable diluents or solvents, such as mannitol, 1,3-butanediol, water, Ringer's solution or isotonic sodium chloride solution, or suitable dispersing or wetting and suspending agents, such as sterile, bland, fixed oils, including synthetic mono- or diglycerides, and fatty acids, including oleic acid.
The compounds herein set forth can be administered orally to humans in a dosage range of about 1 to 100 mg/kg body weight in divided doses, usually over an extended period, such as days, weeks, months, or even years. One preferred dosage range is about 1 to 10 mg/kg body weight orally in divided doses. Another preferred dosage range is about 1 to 20 mg/kg body weight in divided doses. It will be understood, however, that the specific dose level and frequency of dosage for any particular patient may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the host undergoing therapy.
Also contemplated herein are combinations of the compounds of Formula I herein set forth, together with one or more other agents useful in the treatment of AIDS. For example, the compounds of this disclosure may be effectively administered, whether at periods of pre-exposure and/or post-exposure, in combination with effective amounts of the AIDS antivirals, immunomodulators, antiinfectives, or vaccines, such as those in the following non-limiting table:
ANTIVIRALS
Drug Name Manufacturer Indication 097 Hoechst/Bayer HIV infection,
AIDS, ARC
(non-nucleoside
reverse transcriptase (RT)
inhibitor)
Amprenavir Glaxo Wellcome HIV infection,
141 W94 AIDS, ARC
GW 141 (protease inhibitor)
Abacavir (1592U89) Glaxo Wellcome HIV infection,
GW 1592 AIDS, ARC
(RT inhibitor)
Acemannan Carrington Labs ARC
(Irving, TX)
Acyclovir Burroughs Wellcome HIV infection, AIDS,
ARC AD-439 Tanox Biosystems HIV infection, AIDS,
ARC
AD-519 Tanox Biosystems HIV infection, AIDS,
ARC
Adefovir dipivoxil Gilead Sciences HIV infection
AL-721 Ethigen ARC, PGL
(Los Angeles, CA) HIV positive, AIDS
Alpha Interferon Glaxo Wellcome Kaposi's sarcoma,
HIV in combination w/Retrovir Ansamycin Adria Laboratories ARC LM 427 (Dublin, OH)
Erbamont
(Stamford, CT)
Antibody which Advanced Biotherapy AIDS, ARC
Neutralizes pH Concepts
Labile alpha aberrant (Rockville, MD)
Interferon
AR177 Aronex Pharm HIV infection, AIDS,
ARC
Beta-fluoro-ddA Nat'l Cancer Institute AIDS-associated
diseases
BMS-234475 Bristol-Myers Squibb/ HIV infection,
(CGP-61755) Novartis AIDS, ARC
(protease inhibitor)
CI-1012 Warner-Lambert HIV-1 infection
Cidofovir Gilead Science CMV retinitis,
herpes, papillomavirus
Curdlan sulfate AJI Pharma USA HIV infection
Cytomegalovirus Medlmmune CMV retinitis
Immune globin
Cytovene Syntex Sight threatening
Ganciclovir CMV
peripheral CMV retinitis
Darunavir Tibotec- J & J HIV infection, AIDS, ARC
(protease inhibitor) Delaviridine Pharmacia-Upjohn HIV infection,
AIDS, ARC
(RT inhibitor) Dextran Sulfate Ueno Fine Chem. AIDS, ARC, HIV
Ind. Ltd. (Osaka, positive
Japan) asymptomatic
ddC Hoffman-La Roche HIV infection, AIDS,
Dideoxycytidine ARC ddl Bristol-Myers Squibb HIV infection, AIDS,
Dideoxyinosine ARC; combination
with AZT/d4T
DMP-450 AVID HIV infection,
(Camden, NJ) AIDS, ARC
(protease inhibitor)
Efavirenz Bristol Myers Squibb HIV infection,
(DMP 266, Sustiva®) AIDS, ARC
(-)6-Chloro-4-(S)- (non-nucleoside RT cyclopropylethynyl- inhibitor)
4(S)-trifluoro- methyl- 1 ,4-dihydro- 2H-3, 1 -benzoxazin- 2-one, STOCRI E
EL10 Elan Corp, PLC HIV infection
(Gainesville, GA)
Etravirine Tibotec/ J & J HIV infection, AIDS, ARC
(non-nucleoside reverse transcriptase inhibitor)
Famciclovir Smith Kline herpes zoster,
herpes simplex
GS 840 Gilead HIV infection,
AIDS, ARC
(reverse transcriptase inhibitor)
HBY097 Hoechst Marion HIV infection,
Roussel AIDS, ARC
(non-nucleoside reverse transcriptase inhibitor) Hypericin VIMRx Pharm. HIV infection, AIDS,
ARC Recombinant Human Triton Biosciences AIDS, Kaposi's Interferon Beta (Almeda, CA) sarcoma, ARC
Interferon alfa-n3 Interferon Sciences ARC, AIDS Indinavir Merck HIV infection, AIDS,
ARC, asymptomatic HIV positive, also in combination with AZT/ddl/ddC
ISIS 2922 ISIS Pharmaceuticals CMV retinitis
KNI-272 Nat'l Cancer Institute HrV-assoc. diseases Lamivudine, 3TC Glaxo Wellcome HIV infection,
AIDS, ARC
(reverse
transcriptase inhibitor); also with AZT
Lobucavir Bristol-Myers Squibb CMV infection Nelfinavir Agouron HIV infection,
Pharmaceuticals AIDS, ARC
(protease inhibitor)
Nevirapine Boeheringer HIV infection,
Ingleheim AIDS, ARC
(RT inhibitor)
Novapren Novaferon Labs, Inc. HIV inhibitor
(Akron, OH) Peptide T Peninsula Labs AIDS
Octapeptide (Belmont, CA)
Sequence Trisodium Astra Pharm. CMV retinitis, HIV Phosphonoformate Products, Inc. infection, other CMV infections
PNU- 140690 Pharmacia Upjohn HIV infection,
AIDS, ARC
(protease inhibitor) Probucol Vyrex HIV infection, AIDS
RBC-CD4 Sheffield Med. HIV infection,
Tech (Houston, TX) AIDS, ARC
Rilpivirine Tibotec HIV infection, AIDS, ARC
( RTI)
Ritonavir Abbott HIV infection,
AIDS, ARC
(protease inhibitor)
Saquinavir Hoffmann- HIV infection,
LaRoche AIDS, ARC
(protease inhibitor) Stavudine; d4T Bristol-Myers Squibb HIV infection, AIDS, Didehydrodeoxy- ARC
Thymidine
Tipranavir Boehringer Ingelheim HIV infection, AIDS, ARC
(protease inhibitor)
Valaciclovir Glaxo Wellcome Genital HSV & CMV infections
Virazole Viratek/ICN asymptomatic HIV Ribavirin (Costa Mesa, CA) positive, LAS, ARC
VX-478 Vertex HIV infection, AIDS,
ARC Zalcitabine Hoffmann-LaRoche HIV infection, AIDS,
ARC, with AZT
Zidovudine; AZT Glaxo Wellcome HIV infection, AIDS,
ARC, Kaposi's sarcoma, in combination with other therapies
Tenofovir disoproxil, HIV infection,
fumarate salt (Viread®) AIDS,
(reverse transcriptase inhibitor)
EmtrivaR (Emtricitabine) Gilead HIV infection,
(FTC) AIDS,
(reverse transcriptase inhibitor)
Combivir" GSK HIV infection,
AIDS,
(reverse transcriptase inhibitor)
Abacavir succinate GSK HIV infection,
(or Ziagen®) AIDS,
(reverse transcriptase inhibitor)
Reyataz Bristol-Myers Squibb HIV infection
(or atazanavir) AIDs, protease
inhibitor
Fuzeon Roche / Trimeris HIV infection
(Enfuvirtide or T-20) AIDs, viral Fusion
inhibitor
Lexiva® GSK/Vertex HIV infection
(or Fosamprenavir calcium) AIDs, viral protease
inhibitor
Selzentry
Maraviroc; (UK 427857) Pfizer HIV infection
AIDs, (CCR5 antagonist, in development)
Trizivir GSK HIV infection
AIDs, (three drug combination)
Sch-417690 (vicriviroc) Schering-Plough HrV infection
AIDs, (CCR5 antagonist, in development) TAK-652 Takeda HIV infection
AIDs, (CCR5 antagonist, in development)
GSK 873140 GSK/ONO HIV infection
(ONO-4128) AIDs, (CCR5 antagonist, in development)
Integrase Inhibitor Merck HIV infection
MK-0518 AIDs
Raltegravir
®
Truvada Gilead Combination of Tenofovir disoproxil fumarate salt
(Viread^) and Emtriva® (Emtricitabine)
Integrase Inhibitor Gilead/Japan Tobacco HIV Infection
GS917/JTK-303 AIDs
Elvitegravir in development
Triple drug combination Gilead/Bristol-Myers Squibb Combination of Tenofovir
Atripla disoproxil fumarate salt
(Viread®), Emtriva R (Emtricitabine), and Sustiva (Efavirenz)
®
Festinavir Oncolys BioPharma HIV infection
AIDs
in development
CMX-157 Chimerix HIV infection
Lipid conjugate of AIDs
nucleotide tenofovir
GSK1349572 GSK HIV infection
Integrase inhibitor AIDs
IMMUNOMODULATORS
Drug Name Manufacturer Indication AS-101 Wyeth-Ayerst AIDS Bropirimine Pharmacia Upjohn Advanced AIDS Acemannan Carrington Labs, Inc. AIDS, ARC
(Irving, TX)
CL246,738 Wyeth AIDS, Kaposi's
Lederle Labs sarcoma
FP-21399 Fuki ImmunoPharm Blocks HIV fusion with CD4+ cells
Gamma Interferon Genentech ARC, in combination w/TNF (tumor necrosis factor)
Granulocyte Genetics Institute AIDS
Macrophage Colony Sandoz
Stimulating Factor
Granulocyte Hoechst-Roussel AIDS
Macrophage Colony Immunex
Stimulating Factor Granulocyte Schering-Plough AIDS,
Macrophage Colony combination
Stimulating Factor w/AZT
HIV Core Particle Rorer Seropositive HIV Immunostimulant
IL-2 Cetus AIDS, in combination
Interleukin-2 w/AZT
IL-2 Hoffman-LaRoche AIDS, ARC, HIV, in Interleukin-2 Immunex combination w/AZT
IL-2 Chiron AIDS, increase in
Interleukin-2 CD4 cell counts (aldeslukin) Immune Globulin Cutter Biological Pediatric AIDS, in
Intravenous (Berkeley, CA) combination w/AZT
(human) IMREG-1 Imreg AIDS, Kaposi's
(New Orleans, LA) sarcoma, ARC, PGL
IMREG-2 Imreg AIDS, Kaposi's
(New Orleans, LA) sarcoma, ARC, PGL
Imuthiol Diethyl Merieux Institute AIDS, ARC
Dithio Carbamate
Alpha-2 Schering Plough Kaposi's sarcoma Interferon w/AZT, AIDS
Methionine- TNI Pharmaceutical AIDS, ARC
Enkephalin (Chicago, IL)
MTP-PE Ciba-Geigy Corp. Kaposi's sarcoma
Muramyl-Tripeptide
Granulocyte Amgen AIDS, in combination Colony Stimulating w/AZT
Factor
Remune Immune Response Immunotherapeutic
Corp. rCD4 Genentech AIDS, ARC
Recombinant
Soluble Human CD4 rCD4-IgG AIDS, ARC hybrids
Recombinant Biogen AIDS, ARC
Soluble Human CD4
Interferon Hoffman-La Roche Kaposi's sarcoma Alfa 2a AIDS, ARC,
in combination w/AZT SK&F 106528 Smith Kline HIV infection Soluble T4
Thymopentin Immunobiology HIV infection
Research Institute
(Annandale, NJ)
Tumor Necrosis Genentech ARC, in combination Factor; TNF w/gamma Interferon
ANTI-INFECTIVES
Drug Name Manufacturer Indication
Clindamycin with Pharmacia Upjohn PCP
Primaquine
Fluconazole Pfizer Cryptococcal
meningitis, candidiasis
Pastille Squibb Corp. Prevention of
Nystatin Pastille oral candidiasis
Ornidyl Merrell Dow PCP
Eflornithine
Pentamidine LyphoMed PCP treatment Isethionate (IM & IV) (Rosemont, IL) Trimethoprim Antibacterial
Trimethoprim/ sulfa Antibacterial
Piritrexim Burroughs Wellcome PCP treatment
Pentamidine Fisons Corporation PCP prophylaxis Isethionate for
Inhalation Spiramycin Rhone-Poulenc Cryptosporidia!
diarrhea
Intraconazole- Janssen-Pharm. Histoplasmosis;
R51211 cryptococcal
meningitis
Trimetrexate Warner-Lambert PCP Daunorubicin NeXstar, Sequus Kaposi's sarcoma
Recombinant Human Ortho Pharm. Corp. Severe anemia
Erythropoietin assoc. with AZT
therapy
Recombinant Human Serono AIDS-related
Growth Hormone wasting, cachexia
Megestrol Acetate Bristol-Myers Squibb Treatment of
anorexia assoc.
W/AIDS
Testosterone Alza, Smith Kline AIDS-related wasting
Total Enteral Norwich Eaton Diarrhea and
Nutrition Pharmaceuticals malabsorption
related to AIDS
Additionally, the compounds of the disclosure herein set forth may be used in combination with HIV entry inhibitors. Examples of such HIV entry inhibitors are discussed in DRUGS OF THE FUTURE 1999, 24(12), pp. 1355-1362; CELL, Vol. 9, pp.
243-246, Oct. 29, 1999; and DRUG DISCOVERY TODAY, Vol. 5, No. 5, May 2000, pp.
183-194 and Inhibitors of the entry of HIV into host cells. Meanwell, Nicholas A.;
Kadow, John F. Current Opinion in Drug Discovery & Development (2003), 6(4), 451- 461. Specifically the compounds can be utilized in combination with attachment inhibitors, fusion inhibitors, and chemokine receptor antagonists aimed at either the
CCR5 or CXCR4 coreceptor. HIV attachment inhibitors are also set forth in US
7,354,924 and US 2005/0209246. It will be understood that the scope of combinations of the compounds of this application with AIDS antivirals, immunomodulators, anti-infectives, HIV entry inhibitors or vaccines is not limited to the list in the above Table but includes, in principle, any combination with any pharmaceutical composition useful for the treatment of AIDS.
Preferred combinations are simultaneous or alternating treatments with a compound of the present disclosure and an inhibitor of HIV protease and/or a non- nucleoside inhibitor of HIV reverse transcriptase. An optional fourth component in the combination is a nucleoside inhibitor of HIV reverse transcriptase, such as AZT, 3TC, ddC or ddl. A preferred inhibitor of HIV protease is Reyataz® (active ingredient Atazanavir). Typically a dose of 300 to 600mg is administered once a day. This may be co-administered with a low dose of Ritonavir (50 to 500mgs). Another preferred inhibitor of HIV protease is Kaletra®. Another useful inhibitor of HIV protease is indinavir, which is the sulfate salt of N-(2(R)-hydroxy-l-(S)-indanyl)-2(R)-phenylmethyl- 4-(S)-hydroxy-5-(l-(4-(3-pyridyl-methyl)-2(S)-N'-(t-butylcarboxamido)-piperazinyl))- pentaneamide ethanolate, and is synthesized according to U.S. 5,413,999. Indinavir is generally administered at a dosage of 800 mg three times a day. Other preferred protease inhibitors are nelfinavir and ritonavir. Another preferred inhibitor of HIV protease is saquinavir which is administered in a dosage of 600 or 1200 mg tid. Preferred non- nucleoside inhibitors of HIV reverse transcriptase include efavirenz. These combinations may have unexpected effects on limiting the spread and degree of infection of HIV. Preferred combinations include those with the following (1) indinavir with efavirenz, and, optionally, AZT and/or 3TC and/or ddl and/or ddC; (2) indinavir, and any of AZT and/or ddl and/or ddC and/or 3TC, in particular, indinavir and AZT and 3TC; (3) stavudine and 3TC and/or zidovudine; (4) tenofovir disoproxil fumarate salt and emtricitabine.
In such combinations the compound of the present invention and other active agents may be administered separately or in conjunction. In addition, the administration of one element may be prior to, concurrent to, or subsequent to the administration of other agent(s). GENERAL CHEMISTRY (METHODS OF SYNTHESIS)
The present invention comprises compounds of Formula I, their pharmaceutical formulations, and their use in patients suffering from or susceptible to HIV infection. The compounds of Formula I also include pharmaceutically acceptable salts thereof. General procedures to construct compounds of Formula I and intermediates useful for their synthesis are described in the following Schemes (after the Abbreviations).
Abbreviations One or more of the following abbreviations, most of which are conventional abbreviations well known to those skilled in the art, may be used throughout the description of the disclosure and the examples:
BZ2O = benzoic anhydride
TBTU = 0-(benzotriazol- 1 -yl)-N,N,N',N'-tetramethyluronium tetrafluoroborate
HATU = 2-(lH-7-azabenzotriazol-l-yl)-l, l,3,3-tetramethyluronium hexafluorophosphate methanaminium
DCE = dichloroethane DCM = dichloromethane CDI = carbonyl diimidazole prep. HPLC = preparative high performance liquid chromatography rt = room temperature
DIPEA = diisopropylethylamine DMAP = 4-dimethylaminopyridine DMSO = dimethylsulfoxide
THF = tetrahydrofuran
KHMDS = potassium bis(trimethylsilyl)amide min = minute(s) h = hour(s)
sat. = saturated
TEA = triethylamine
EtOAc = ethyl acetate
TFA = trifluoroacetic acid
PCC = pyridinium chlorochromate
TLC = thin layer chromatography
Tf2NPh = (trifluoromethylsulfonyl)methanesulfonamide dioxane = 1,4-dioxane
PG = protective group
atm = atmosphere(s)
mol = mole(s)
mmol= milimole(s)
mg = milligram(s)
μg = microgram(s)
μΐ = microliter(s)
micrometer(s)
mm= millimeter(s)
HOAc= acetic acid
MeOH= methanol
DMF= N,N-dimethylformamide
TBAF= tetrabutylammonium fluoride Preparation of Compounds of Formula I General Chemistry Schemes:
Compounds of Formula I can be prepared from commercially available (Aldrich, others) betulinic acid by chemistry described in the following schemes.
General reaction schemes are set forth as follows:
Figure imgf000034_0001
Betulinic acid can be converted into the corresponding C-17 amine analog via Curtius' rearrangement. Selective alkylation of the amine can be achieved by treatment with an alkylating agent (R-LG), where LG is a leaving group such as, but not limited to Br, CI, I, mesylate, tosylate or triflate in the presence of a base. Heating maybe needed in some cases. In some cases, by prolonging the reaction times and heating the reaction mixture, the dialkylated product can also be formed. Acylation of the C-3 hydroxyl can be achieved by heating in the presence of the corresponding anhydride. Alternatively, acylation can also be accomplished by addition of the corresponding carboxylic acid and a couping agent such as, but not limited to HATU or TBTU. Saturation of the double bond can be performed under standard conditions in any stage of the synthesis. EXAMPLES
The following examples illustrate typical syntheses of the compounds of Formula I as described generally above. These examples are illustrative only and are not intended to limit the disclosure in any way. The reagents and starting materials are readily available to one of ordinary skill in the art.
Chemistry
Typical Procedures and Characterization of Selected Examples:
Unless otherwise stated, solvents and reagents were used directly as obtained from commercial sources, and reactions were performed under a nitrogen atmosphere. Flash chromatography was conducted on Silica gel 60 (0.040-0.063 particle size; EM Science supply). XH NMR spectra were recorded on Bruker DRX-500f at 500 MHz (or Bruker AV 400 MHz, Bruker DPX-300B or Varian Gemini 300 at 300 MHz as stated). The chemical shifts were reported in ppm on the δ scale relative to 6TMS = 0. The following internal references were used for the residual protons in the following solvents: CDCI3Η 7.26), CD3OD (δΗ 3.30), Acetic-d4 (Acetic Acid d4) (δΗ 1 1.6, 2.07), DMSO mix or DMS0-D6_CDC13 ((H 2.50 and 8.25) (ratio 75%:25%), and DMSO-D6 (δΗ 2.50).
Standard acronyms were employed to describe the multiplicity patterns: s (singlet), br. s (broad singlet), d (doublet), t (triplet), q (quartet), m (multiplet), b (broad), app (apparent). The coupling constant (J) is in Hertz. All Liquid Chromatography (LC) data were recorded on a Shimadzu LC-10AS liquid chromatograph using a SPD-10AV UV-Vis detector with Mass Spectrometry (MS) data determined using a Micromass Platform for LC in electrospray mode.
LC/MS methods: Method 1
Start %B = 0, final %B = 100 over 2 minute gradient
Flow rate = 1 mL / min
Solvent A = 95% H20/ 5% MeOH/ 10 mM NH4OAc
Solvent B = 5% H20 / 95% MeOH / 10 mM NH4OAc
Column = PHENOMENEX- LUNA 2.0 x 30 mm 3 μιη Preparation of compounds:
Example 1 and Example 2
Preparation of 4-(((lR,3aS,5aR,5bR,7aR,9S,llaR,13aR,13bR)-3a-((2-(l,l-dioxido-4- thiomorpholinyl)ethyl)amino)-l-isopropenyl-5a,5b,8,8,lla-pentamethylicosahydro- lH-cyclopenta[a]chrysen-9-yl)oxy)-3,3-dimethyl-4-oxobutanoic acid and 4- (((lR,3aS,5aR,5bR,7aR,9S,HaR,13aR,13bR)-3a-((2-(l,l-dioxido-4- thiomorpholinyl)ethyl)amino)-l-isopropenyl-5a,5b,8,8,lla-pentamethylicosahydro- lH-cyclopenta [a] chrysen-9-yl)oxy)-2,2-dimethyl-4-oxobutanoic acid
Figure imgf000036_0001
Scheme 1
Step 1: Preparation of (lR,3aS,5aR,5bR,7aR,9S,llaR,llbR,13aR,13bR)-3a-amino- 5a,5b,8,8,l la-pentamethyl-l-(prop-l-en-2-yl)icosahydro-lH-cyclopenta [a] chrysen-9- ol
Figure imgf000036_0002
a) A mixture of (lR,3aS,5aR,5bR,7aR,9S,l laR,l lbR,13aR,13bR)-9-hydroxy- 5a,5b,8,8,l la-pentamethyl-l-(prop-l-en-2-yl)icosahydro-lH-cyclopenta[a]chrysene-3a- carboxylic acid (5 g, 10.95 mmol), diphenyl phosphorazidate (3.54 mL, 16.42 mmol) and triethylamine (4.58 mL, 32.8 mmol) in toluene (50 mL) was refluxed for 16 h at 110 °C. The reaction mixture was concentrated under reduced pressure and the residue was purified in silica gel using 6-30% ethyl acetate/hexanes to provide the title compound as white solid (2.8 g). b) To this intermediate (2.8 g, 6.17 mmol) in THF (20 mL) was added HC1 ( 4N, 10.14 mL, 123 mmol) and the reaction mixture was heated up at 60 °C for 48 h. The reaction mixture was concentrated under reduced pressure and the residue was purified in silica gel using 0-10% methylene chloride/methanol/5% TEA. The resulting solid was recrystalized in methanol/dioxane/water to provide the title compound as a white solid (2.3 g, 50%). LCMS: m/e 428.23 (M+H)+, 2.26 min (method 1). 'H NMR (500MHZ, Acetic) δ 4.86 (s, 1H), 4.73 (s, 1H), 3.32 (dd, J=10.4, 5.5 Hz, 1H), 2.78 (d, J=6.1 Hz, 1H), 2.38 - 2.12 (m, 2H), 1.98 - 0.93 (m, 22H), 1.76 (s, 3H), 1.13 (s, 3H), 1.07 (s, 3H), 0.99 (s, 3H), 0.90 (s, 3H), 0.81 (s, 3H).
Step 2: Preparation of (lR,3aS,5aR,5bR,7aR,9S,HaR,HbR,13aR,13bR)-3a-((2-(l,l- dioxido-4-thiomorpholinyl)ethyl)amino)-l-isopropenyl-5a,5b,8,8,lla- pentamethylicosahydro-lH-cyclopenta [a] chrysen-9-ol
Figure imgf000037_0001
A mixture of (lR,3aS,5aR,5bR,7aR,9S,l laR,l lbR,13aR,13bR)-3a-amino- 5a,5b,8,8,l la-pentamethyl-l-(prop-l-en-2-yl)icosahydro-lH-cyclopenta[a]chrysen-9-ol (1.03 g, 2.408 mmol), 4-(2-chloroethyl)thiomorpholine (1.428 g, 7.22 mmol), potassium phosphate (2.045 g, 9.63 mmol) and potassium iodide (0.959 g, 5.78 mmol) in acetonitrile (5 mL) was heated up at 120 °C for 20 h. The reaction mixture was concentrated under reduced pressure and the crude was purified in silica gel using 10-100% ethyl acetate/hexanes to provide the title compound as a white solid (0.9 g, 64%). LCMS: m/e 589.38 (M+H)+, 2.46 min (method 1). Step 3: Preparation of 4-(((lR,3aS,5aR,5bR,7aR,9S,llaR,13aR,13bR)-3a-((2 dioxido-4-thiomorpholinyl)ethyl)amino)-l-isopropenyl-5a,5b,8,8,lla- pentamethylicosahydro-lH-cyclopenta[a]chrysen-9-yl)oxy)-3,3-dimethyl-4- oxobutanoic acid
Figure imgf000038_0001
Example 1
and 4-(((lR,3aS,5aR,5bR,7aR,9S,HaR,13aR,13bR)-3a-((2-(l,l-dioxido-4- thiomorpholinyl)ethyl)amino)-l-isopropenyl-5a,5b,8,8,lla-pentamethylicosahydro- lH-cyclopenta [a] chrysen-9-yl)oxy)-2,2-dimethyl-4-oxobutanoic acid
Figure imgf000038_0002
Example 2
A mixture of (lR,3aS,5aR,5bR,7aR,9S,l laR,l lbR,13aR,13bR)-3a-((2-(l,l- dioxido-4-thiomorpholinyl)ethyl)amino)-l-isopropenyl-5a,5b,8,8,l la- pentamethylicosahydro-lH-cyclopenta[a]chrysen-9-ol (100 mg, 0.170 mmol), 3,3- dimethyldihydrofuran-2,5-dione (436 mg, 3.4 mmol) and DMAP (208 mg, 1.7 mmol) in pyridine (1 mL) was heated up at 150 °C for 37 h. The reaction mixture was filtered and purified by prep. HPLC to provide the title compounds 4- (((lR,3aS,5aR,5bR,7aR,9S,l laR,13aR,13bR)-3a-((2-(l,l-dioxido-4- thiomorpholinyl)ethyl)amino)- l-isopropenyl-5a,5b,8,8, 11 a-pentamethylicosahydro- 1H- cyclopenta[a]chrysen-9-yl)oxy)-3,3-dimethyl-4-oxobutanoic acid, example 1, as a white solid (17 mg, 13%). LCMS: m/e 717.51 (M+H)+, 2.39 min (method 1). XH NMR
(500MHz, Acetic) δ 4.83 (s, 1H), 4.75 (s, 1H), 4.51 (dd, J=10.8, 5.6 Hz, 1H), 3.41 - 3.11 (m, 11H), 3.10 - 2.99 (m, 1H), 2.82 (td, J=10.8, 5.5 Hz, 1H), 2.75 - 2.63 (m, 2H), 2.27 - 1.04 (m, 24H), 1.76 (s, 3H), 1.31 (s, 6H), 1.19 (s, 3H), 1.13 (s, 3H), 0.95 (s, 3H), 0.92 (s, 3H), 0.92 (s, 3H) and 4-(((lR,3aS,5aR,5bR,7aR,9S,l laR,13aR,13bR)-3a-((2-(l,l- dioxido-4-thiomorpholinyl)ethyl)amino)-l-isopropenyl-5a,5b,8,8,l la- pentamethylicosahydro-lH-cyclopenta[a]chrysen-9-yl)oxy)-2,2-dimethyl-4-oxobutanoic acid, example 2, as a white solid (60 mg, 47%). LCMS: m/e 717.51 (M+H)+, 2.44 min (method 1). XH NMR (500MHz, Acetic) δ 4.82 (s, 1H), 4.72 (s, 1H), 4.57 (dd, J=10.2, 6.0 Hz, 1H), 3.44 (d, J=12.5 Hz, 1H), 3.35 - 3.20 (m, 6H), 3.19 - 3.09 (m, 3H), 3.05 (d, J=4.6 Hz, 2H), 2.97 - 2.85 (m, 1H), 2.81 - 2.58 (m, 2H), 2.32 - 2.13 (m, 2H), 1.79 - 1.05 (m, 22H), 1.75 (s, 3H), 1.33 (s, 3H), 1.31 (s, 3H), 1.23 (s, 3H), 1.11 (s, 3H), 0.96 (s, 3H), 0.90 (s, 3H), 0.89 (s, 3H).
Example 3
Preparation of 5-(((lR,3aS,5aR,5bR,7aR,9S,llaR,13aR,13bR)-3a-((2-(l,l-dioxido-4- thiomorpholinyl)ethyl)amino)-l-isopropenyl-5a,5b,8,8,lla-pentamethylicosahydro- lH-cyclopenta [a] chrysen-9-yl)oxy)-3,3-dimethyl-5-oxopentanoic acid
Figure imgf000039_0001
A mixture of (lR,3aS,5aR,5bR,7aR,9S,l laR,l lbR,13aR,13bR)-3a-((2-(l,l- dioxido-4-thiomorpholinyl)ethyl)amino)-l-isopropenyl-5a,5b,8,8,l la- pentamethylicosahydro-lH-cyclopenta[a]chrysen-9-ol (50 mg, 0.085 mmol), 4,4- dimethyldihydro-2H-pyran-2,6(3H)-dione (242mg, 1.7 mmol) and DMAP (104 mg, 0.85 mmol) in pyridine (1 mL) was heated up at 150 °C for 33 h. The reaction mixture was filtered to remove any solids and the clear solution was purified by prep. HPLC to provide the title compound as a white solid (48 mg, 74%). LCMS: m/e 731.48 (M+H)+, 2.44 min (method 1). ¾ NMR (500MHz, Acetic) δ 4.82 (s, 1H), 4.73 (s, 1H), 4.63 - 4.50 (m, 1H), 3.44 (d, J=12.5 Hz, 1H), 3.36 - 3.20 (m, 6H), 3.19 - 3.11 (m, 3H), 3.09 - 3.01 (m, 2H), 2.99 - 2.85 (m, 1H), 2.66 - 2.42 (m, 4H), 2.31 - 1.06 (m, 24H), 1.75 (s, 3H), 1.23 (s, 3H), 1.18 (s, 3H), 1.17 (s, 3H), 1.12 (s, 3H), 0.98 (s, 3H), 0.93 (s, 6H). Example 4 and Example 5
Preparation of 3,3-dimethyl-4-oxo-4-((lR,3aS,5aR,5bR,7aR,9S,llaR,13aR,13bR)- 5a,5b,8,8,lla-pentamethyl-3a-(2-morpholinoethylamino)-l-(prop-l-en-2- yl)icosahydro-lH-cyclopenta[a]chrysen-9-yloxy)butanoic acid and 2,2-dimethyl-4- 0X0-4-((lR,3aS,5aR,5bR,7aR,9S,llaR,13aR,13bR)-5a,5b,8,8,lla-pentamethyl-3a-(2- morpholinoethylamino)-l-(prop-l-en-2-yl)icosahydro-lH-cyclopenta[a]chrysen-9- yloxy)butanoic acid
Figure imgf000040_0001
example 4 example 5
The title compounds were prepared following the method described in scheme 1 using 4-(2-chloroethyl)morpholine as alkylating reagent instead of 4-(2- chloroethyl)thiomorpholine. The crude material was purified using reverse phase prep HPLC to afford 3,3-dimethyl-4-oxo-4-((lR,3aS,5aR,5bR,7aR,9S,l laR,13aR,13bR)- 5a,5b,8,8, 11 a-pentamethyl-3 a-(2-morpholinoethylamino)- 1 -(prop- 1 -en-2-yl)icosahydro- lH-cyclopenta[a]chrysen-9-yloxy)butanoic acid, example 4, as a white solid (4 mg, 8%). LCMS: m/e 669.36 (M+H)+, 2.51 min (method 1). 'H NMR (500MHZ, Acetic) δ 4.85 (s, 1H), 4.73 (s, 1H), 4.54 (dd, J=10.8, 5.6 Hz, 1H), 3.94 (br. s., 4H), 3.73 - 3.51 (m, 4H), 3.29 (br. s., 4H), 2.91 - 2.79 (m, 1H), 2.75 - 2.65 (m, 2H), 2.26 - 1.03 (m, 24H), 1.76 (s, 3H), 1.32 (s, 6H), 1.16 (s, 3H), 1.10 (s, 3H), 0.95 (s, 3H), 0.93 (s, 3H), 0.92 (s, 3H) and 2,2-dimethyl-4-oxo-4-((lR,3aS,5aR,5bR,7aR,9S,l laR,13aR,13bR)-5a,5b,8,8,l la- pentamethyl-3a-(2-morpholinoethylamino)- 1 -(prop- 1 -en-2-yl)icosahydro- 1 H- cyclopenta[a]chrysen-9-yloxy)butanoic acid, example 5, as a white solid (23 mg, 44%). LCMS: m/e 669.55 (M+H)+, 2.58 min (method 1). 'H NMR (500MHZ, Acetic) δ 4.84 (s, 1H), 4.73 (s, 1H), 4.61 - 4.52 (m, 1H), 3.94 (br. s., 4H), 3.72 - 3.52 (m, 4H), 3.28 (br. s., 4H), 2.89 - 2.79 (m, 1H), 2.78 - 2.62 (m, 2H), 2.28 - 1.19 (m, 24H), 1.75 (s, 3H), 1.33 (s, 3H), 1.31 (s, 3H), 1.15 (s, 3H), 1.10 (s, 3H), 0.94 (s, 3H), 0.90 (s, 3H), 0.88 (s, 3H). Example 6
Preparation of 4-(((lS,3aS,5aR,5bR,7aR,9S,HaR,13aR,13bR)-3a-((2-(l,l-dioxido-4- thiomorpholinyl)ethyl)amino)-l-isopropyl-5a,5b,8,8,lla-pentamethylicosahydro-lH- cyclopenta [a] chrysen-9-yl)oxy)-2,2-dimethyl-4-oxobutanoic acid
Figure imgf000041_0001
A mixture of 4-(((lR,3aS,5aR,5bR,7aR,9S,l laR,13aR,13bR)-3a-((2-(l,l-dioxido- 4-thiomorpholinyl)ethyl)amino)- 1 -isopropenyl-5a,5b,8,8, 11 a-pentamethylicosahydro- 1H- cyclopenta[a]chrysen-9-yl)oxy)-2,2-dimethyl-4-oxobutanoic acid (10 mg, 0.014 mmol) and 10% palladium on carbon (14.84 mg, 0.014 mmol) in methanol (5 mL) was stirred at 20 °C for 18 h under hydrogen at 40 psi. The reaction mixture was filtered to remove the catalyst and the solution was purified by reverse phase prep HPLC to provide the title compound as a white solid (4.1 mg, 39%). LCMS: m/e 719.53 (M+H)+, 2.41 min
(method 1). 'H NMR (500MHZ, Acetic) δ 4.58 (dd, J=10.5, 5.6 Hz, IH), 3.43 - 3.36 (m, IH), 3.34 - 3.19 (m, 6H), 3.18 - 3.01 (m, 5H), 2.80 - 2.61 (m, 2H), 2.27 - 0.89 (m, 26H), 1.33 (s, 3H),1.32 (s, 3H), 1.23 (s, 3H), 1.09 (s, 3H), 0.98 (s, 3H), 0.93 (d, J=6.7 Hz, 3H), 0.90 (s, 6H), 0.84 (d, J=6.7 Hz, 3H).
Example 7
Preparation of 5-(((lS,3aS,5aR,5bR,7aR,9S,HaR,13aR,13bR)-3a-((2-(l,l-dioxido-4- thiomorpholinyl)ethyl)amino)-l-isopropyl-5a,5b,8,8,lla-pentamethylicosahydro-lH- cyclopenta[a]chrysen-9-yl)oxy)-3,3-dimethyl-5-oxopentanoic acid
Figure imgf000041_0002
A mixture of 5-(((lR,3aS,5aR,5bR,7aR,9S,l laR,13aR,13bR)-3a-((2-(l,l-dioxido- 4-thiomorpholinyl)ethyl)amino)- 1 -isopropenyl-5a,5b,8,8, 11 a-pentamethylicosahydro- 1H- cyclopenta[a]chrysen-9-yl)oxy)-3,3-dimethyl-5-oxopentanoic acid (15 mg, 0.021 mmol) and 10% palladium on carbon (21.83 mg, 0.021 mmol) in methanol (5 mL) was stirred at 20 °C for 18 h under hydrogen at 35 psi. The reaction mixture was filtered to remove the catalyst and the solution was purified using reverse phase prep HPLC to provide the title compound as colorless oil (7 mg, 44%). LCMS: m/e 733.30 (M+H)+, 2.44 min (method 1). XH NMR (500MHz, Acetic) δ 4.66 - 4.46 (m, 1H), 3.38 (d, J=l 1.6 Hz, 1H), 3.34 - 2.99 (m, 11H), 2.61 - 2.44 (m, 4H), 2.26 - 1.03 (m, 26H), 1.24 (s, 3H), 1.19 (s, 3H), 1.18 (s, 3H), 1.10 (s, 3H), 0.99 (s, 3H), 0.93 (s, 6H), 0.92 (d, J=6.7 Hz, 3H), 0.85 (d, J=6.7 Hz, 3H).
Example 8 and Example 9
Preparation of 3,3-dimethyl-4-oxo-4-(((lR,3aS,5aR,5bR,7aR,9S,llaR,13aR,13bR)- 5a,5b,8,8,lla-pentamethyl-3a-((2-(4-(methylsulfonyl)piperidin-l-yl)ethyl)amino)-l- (prop-l-en-2-yl)icosahydro-lH-cyclopenta[a]chrysen-9-yl)oxy)butanoic acid and 2,2- dimethyl-4-oxo-4-(((lR,3aS,5aR,5bR,7aR,9S,llaR,13aR,13bR)-5a,5b,8,8,lla- pentamethyl-3a-((2-(4-(methylsulfonyl)piperidin-l-yl)ethyl)amino)-l-(prop-l-en-2- yl)icosahydro-lH-cyclopenta[a]chrysen-9-yl)oxy)butanoic acid
Figure imgf000042_0001
The title compounds were prepared following the method described in scheme 1 using l-(2-chloroethyl)-4-(methylsulfonyl)piperidine as alkylating reagent instead of 4- (2-chloroethyl)thiomorpholine. 3,3-dimethyl-4-oxo-4-
(((1 R,3 aS,5aR,5bR,7aR,9S, 11 aR, 13aR, 13bR)-5a,5b,8,8, 11 a-pentamethyl-3 a-((2-(4- (methylsulfonyl)piperidin- 1 -yl)ethyl)amino)- 1 -(prop- 1 -en-2-yl)icosahydro- 1 H- cyclopenta[a]chrysen-9-yl)oxy)butanoic acid, example 8, was isolated as a white solid (1.5 mg, 7%). LCMS: m/e 745.6 (M+H)+, 2.3 min (method 1). ¾ NMR (500MHz,
Acetic) δ 4.85 (s, 1H), 4.74 (s, 1H), 4.54 (dd, J=10.8, 5.0 Hz, 1H), 3.80 - 3.50 (m, 6H), 3.46 - 3.26 (m, 1H), 3.09 - 3.02 (m, 2H), 3.02 (s, 3H), 2.90 - 2.74(m, 1H), 2.76 - 2.61 (m, 2H), 2.47 - 2.32 (m, 2H), 2.26 - 1.05 (m, 26H), 1.76 (s, 3H), 1.32 (s, 6H), 1.16 (s, 3H), 1.10 (s, 3H), 0.95 (s, 3H), 0.93 (s, 3H), 0.92 (s, 3H). 2,2-dimethyl-4-oxo-4- (((1 R,3 aS,5aR,5bR,7aR,9S, 11 aR, 13aR, 13bR)-5a,5b,8,8, 11 a-pentamethyl-3 a-((2-(4- (methylsulfonyl)piperidin- 1 -yl)ethyl)amino)- 1 -(prop- 1 -en-2-yl)icosahydro- 1 H- cyclopenta[a]chrysen-9-yl)oxy)butanoic acid, example 9, was isolated as a white solid (8.2 mg, 36%). LCMS: m/e 743.55 (M-H)", 2.34 min (method 1). 'H NMR (400MHZ, Acetic) δ 4.81 (s, 1H), 4.69 (s, 1H), 4.60 - 4.45 (m, 1H), 3.77 - 3.43 (m, 6H), 3.38 - 3.22 (m, 1H), 3.06 - 2.92 (m, 2H), 2.97 (s, 3H), 2.82 - 2.73 (m, 1H), 2.73 - 2.59 (m, 2H), 2.44 - 2.27 (m, 2H), 2.19 - 1.01 (m, 26H), 1.72 (s, 3H), 1.29 (s, 3H), 1.27 (s, 3H), 1.12 (s, 3H), 1.06 (s, 3H), 0.90 (s, 3H), 0.86 (s, 3H), 0.85 (s, 3H).
Example 10
Preparation of 4-(((lR,3aS,5aR,5bR,7aR,9S,HaR,13aR,13bR)-3a-((2- (dimethylamino)ethyl)amino)-5a,5b,8,8,lla-pentamethyl-l-(prop-l-en-2- yl)icosahydro-lH-cyclopenta [a] chrysen-9-yl)oxy)-2,2-dimethyl-4-oxobutanoic acid
Figure imgf000043_0001
The title compound was prepared following the method described in scheme 1 using 2-chloro-N,N-dimethylethanamine hydrochloride as alkylating reagent instead of 4- (2-chloroethyl)thiomorpholine. The product was isolated as a white solid (0.6 mg, 23%). LCMS: m/e 627.6 (M+H)+, 2.39 min (method 1). ¾ NMR (500MHz, Acetic) δ 4.85 (s, 1H), 4.74 (s, 1H), 4.63 - 4.47 (m, 1H), 3.94 - 3.59 (m, 4H), 3.00 (s, 6H), 2.88 - 2.77 (m, 1H), 2.77 - 2.59 (m, 2H), 2.39 - 1.18 (m, 24H), 1.76 (s, 3H), 1.32 (s, 3H), 1.31 (s, 3H), 1.12 (s, 3H), 1.10 (s, 3H), 0.93 (s, 3H), 0.89 (s, 3H), 0.88 (s, 3H).
Preparation of 2,2-dimethyl-4-oxo-4-
(((lR,3aS,5aR,5bR,7aR,9S,llaR,HbR,13aR,13bR)-5a,5b,8,8,lla-pentamethyl-3a ((2-(4-(methylsulfonyl)piperazin-l-yl)ethyl)amino)-l-(prop-l-en-2-yl)icosahydi cyclopenta [a] chrysen-9-yl)oxy)butanoic acid
Figure imgf000044_0001
The title compound was prepared following the method described in scheme 1 using 1 -(2-chloroethyl)-4-(methylsulfonyl)piperazine as alkylating reagent instead of 4- (2-chloroethyl)thiomorpholine. The product was isolated as a white solid (5.1 mg, 40%). LCMS: m/e 746.6 (M+H)+, 2.15 min (method 1). ¾ NMR (500MHz, Acetic) δ 4.84 (s, 1H), 4.73 (s, 1H), 4.62 - 4.47 (m, 1H), 3.70 - 3.37 (m, 7H), 3.31 (dd, J=12.2, 6.1 Hz, 1H), 3.16 (br. s., 4H), 2.93 (s, 3H), 2.90 - 2.82 (m, 1H), 2.79 - 2.62 (m, 2H), 2.29 - 1.00 (m, 24H), 1.75 (s, 3H), 1.33 (s, 3H), 1.31 (s, 3H), 1.19 (s, 3H), 1.10 (s, 3H), 0.94 (s, 3H), 0.89 (s, 3H), 0.88 (s, 3H).
Biology Data for the Examples
• "μΜ" means micromolar;
• "mL" means milliliter;
· "μΐ" means microliter;
• "mg" means milligram;
• "| g" means microgram;
The materials and experimental procedures used to obtain the results reported in Tables 1-2 are described below.
HIV cell culture assay - MT-2 cells and 293T cells were obtained from the NIH AIDS Research and Reference Reagent Program. MT-2 cells were propagated in RPMI 1640 media supplemented with 10% heat inactivated fetal bovine serum, 100 μg/ml penicillin G and up to 100 units/ml streptomycin. The 293T cells were propagated in DMEM media supplemented with 10% heat inactivated fetal bovine serum (FBS), 100 units/ml penicillin G and 100 μg/ml streptomycin. The proviral DNA clone of L4_3 was obtained from the NIH AIDS Research and Reference Reagent Program. A recombinant NL4_3 virus, in which a section of the nef gene from NL4-3 was replaced with the Renilla luciferase gene, was used as a reference virus. In addition, residue Gag P373 was converted to P373S. Briefly, the recombinant virus was prepared by transfection of the altered proviral clone of NL4_3. Transfections were performed in 293T cells using LipofectAMINE PLUS from Invitrogen (Carlsbad, CA), according to manufacturer's instruction. The virus was titered in MT-2 cells using luciferase enzyme activity as a marker. Luciferase was quantitated using the Dual Luciferase kit from Promega
(Madison, WI), with modifications to the manufacturer's protocol. The diluted Passive Lysis solution was pre-mixed with the re-suspended Luciferase Assay Reagent and the re-suspended Stop & Glo Substrate (2: 1 : 1 ratio). Fifty (50) of the mixture was added to each aspirated well on assay plates and luciferase activity was measured immediately on a Wallac TriLux (Perkin-Elmer). Antiviral activities of inhibitors toward the recombinant virus were quantified by measuring luciferase activity in cells infected for 4- 5 days with NLRluc recombinants in the presence serial dilutions of the inhibitor. The EC50 data for the compounds is shown in Table 2. Table 1 is the key for the data in Table 2. Results
Table 1. Biological Data Key for EC50
Figure imgf000045_0001
Table 2
Figure imgf000046_0001
Figure imgf000047_0001
The foregoing description is merely illustrative and should not be understood to limit the scope or underlying principles of the invention in any way. Indeed, various modifications of the invention, in addition to those shown and described herein, will become apparent to those skilled in the art from the following examples and the foregoing description. Such modifications are also intended to fall within the scope of the appended claims.

Claims

What is claimed is:
1. A compound of Formula I, including pharmaceutically acceptable salts thereof:
Figure imgf000048_0001
Formula I
wherein a is 1 to 4;
X is -O;
Ri is -C(0)CH2C(CH3)2COOH, -C(0)C(CH3)2CH2COOH, or - C(0)CH2C(CH3)2CH2COOH;
R2 is selected from the group of -H, methyl, isopropenyl and isopropyl;
R3 and R4 are independently selected from the group of -H, -C1-6 alkyl, -Ci_6
alkylsubstituted alkyl and -C3-6 cycloalkyl;
or R3 and R4 are taken together with the adjacent N to form a cycle selected from the group of:
Figure imgf000048_0002
R5 is independently -H or -Ci_6 alkyl;
R6 is selected from the group of -S02R7, -S02 RsR9
R7 is selected from the group of -C e alkyl, -Ci_6 alkylsubstituted alkyl, -C3-6 cycloalkyl and aryl; and
Rs and R9 are independently selected from the group of -H, -C1-6 alkyl, and -C e alkylsubstituted alkyl.
2. The compound as claimed in claim 1, wherein a is 1. compound as claimed in claim 1, wherein Ri is -C(0)CH2C(CH3)2COOH.
4. The compound as claimed in claim 2, wherein Ri is -C(0)CH2C(CH3)2COOH.
5. The compound, including pharmaceutically acceptable salts thereof, which is:
Figure imgf000049_0001
6. A compound which is selected from:
Figure imgf000049_0002
Figure imgf000050_0001
Figure imgf000051_0001
including pharmaceutically
7. A pharmaceutical composition which comprises an antiviral effective amount of one or more of the compounds as claimed in claim 1, together with one or more pharmaceutically acceptable carriers, excipients or diluents.
8. A pharmaceutical composition which comprises an antiviral effective amount of one or more of the compounds as claimed in claim 5, together with one or more pharmaceutically acceptable carriers, excipients or diluents.
9. A pharmaceutical composition which comprises an antiviral effective amount of one or more of the compounds as claimed in claim 6, together with one or more pharmaceutically acceptable carriers, excipients or diluents.
10. The pharmaceutical composition of claim 1, useful for treating infection by HIV, which additionally comprises an antiviral effective amount of an AIDS treatment agent selected from the group consisting of: (a) an AIDS antiviral agent; (b) an anti-infective agent; (c) an immunomodulator; and (d) another HIV entry inhibitor.
11. A method for treating a mammal infected with the HIV virus comprising administering to said mammal an antiviral effective amount of a compound as claimed in claim 1, and one or more pharmaceutically acceptable carriers, excipients or diluents.
12. A method for treating a mammal infected with the HIV virus comprising administering to said mammal an antiviral effective amount of a compound as claimed in claim 5, and one or more pharmaceutically acceptable carriers, excipients or diluents.
13. A method for treating a mammal infected with the HIV virus comprising administering to said mammal an antiviral effective amount of a compound as claimed in claim 6, and one or more pharmaceutically acceptable carriers, excipients or diluents.
PCT/US2012/056186 2011-09-21 2012-09-20 Novel betulinic acid derivatives with antiviral activity WO2013043778A1 (en)

Priority Applications (12)

Application Number Priority Date Filing Date Title
EP12766825.9A EP2758419B8 (en) 2011-09-21 2012-09-20 Novel betulinic acid derivatives with antiviral activity
AU2012312485A AU2012312485B2 (en) 2011-09-21 2012-09-20 Novel betulinic acid derivatives with antiviral activity
JP2014531936A JP6100786B2 (en) 2011-09-21 2012-09-20 Novel betulinic acid derivatives with antiviral activity
EA201490643A EA023463B1 (en) 2011-09-21 2012-09-20 Betulinic acid derivatives with antiviral activity
SG11201400331WA SG11201400331WA (en) 2011-09-21 2012-09-20 Novel betulinic acid derivatives with antiviral activity
MX2014002936A MX2014002936A (en) 2011-09-21 2012-09-20 Novel betulinic acid derivatives with antiviral activity.
BR112014006559A BR112014006559A2 (en) 2011-09-21 2012-09-20 betulinic acid derivatives with antiviral activity
ES12766825.9T ES2611727T3 (en) 2011-09-21 2012-09-20 Novel betulinic acid derivatives with antiviral activity
CN201280046037.7A CN103814042B (en) 2011-09-21 2012-09-20 There is the betulinic acid derivative of antiviral activity
KR1020147010087A KR20140069167A (en) 2011-09-21 2012-09-20 Novel betulinic acid derivatives with antiviral activity
CA2849475A CA2849475A1 (en) 2011-09-21 2012-09-20 Novel betulinic acid derivatives with antiviral activity
IL231421A IL231421A (en) 2011-09-21 2014-03-09 Lupane and its analogs substituted in position 3ß with carboxyalkylcarbonyloxy and in position 17 with (substituted alkyl)amino, as well as anti-hiv pharmaceutical compositions comprising them

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161537099P 2011-09-21 2011-09-21
US61/537,099 2011-09-21

Publications (1)

Publication Number Publication Date
WO2013043778A1 true WO2013043778A1 (en) 2013-03-28

Family

ID=46964090

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/056186 WO2013043778A1 (en) 2011-09-21 2012-09-20 Novel betulinic acid derivatives with antiviral activity

Country Status (15)

Country Link
US (1) US8754069B2 (en)
EP (1) EP2758419B8 (en)
JP (1) JP6100786B2 (en)
KR (1) KR20140069167A (en)
CN (1) CN103814042B (en)
AU (1) AU2012312485B2 (en)
BR (1) BR112014006559A2 (en)
CA (1) CA2849475A1 (en)
EA (1) EA023463B1 (en)
ES (1) ES2611727T3 (en)
IL (1) IL231421A (en)
MX (1) MX2014002936A (en)
SG (1) SG11201400331WA (en)
TW (1) TW201317255A (en)
WO (1) WO2013043778A1 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017017607A1 (en) 2015-07-28 2017-02-02 Glaxosmithkline Intellectual Property (No.2) Limited Betuin derivatives for preventing or treating hiv infections
WO2017017609A1 (en) 2015-07-28 2017-02-02 Glaxosmithkline Intellectual Property (No.2) Limited Betuin derivatives for preventing or treating hiv infections
WO2017051355A1 (en) 2015-09-24 2017-03-30 Glaxosmithkline Intellectual Property (No.2) Limited Compounds with hiv maturation inhibitory activity
WO2017125870A1 (en) 2016-01-20 2017-07-27 Glaxosmithkline Intellectual Property (No.2) Limited Amine derivatives of lupanes with hiv maturation inhibitory activity
CN107428798A (en) * 2015-04-14 2017-12-01 Viiv保健英国第四有限公司 The method for producing HIV maturation inhibitor
WO2019207460A1 (en) 2018-04-24 2019-10-31 VIIV Healthcare UK (No.5) Limited Compounds with hiv maturation inhibitory activity
WO2020165741A1 (en) 2019-02-11 2020-08-20 Hetero Labs Limited Novel triterpene derivatives as hiv inhibitors

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8906889B2 (en) * 2012-02-15 2014-12-09 Bristol-Myers Squibb Company C-3 cycloalkenyl triterpenoids with HIV maturation inhibitory activity
US8889854B2 (en) 2012-05-07 2014-11-18 Bristol-Myers Squibb Company C-17 bicyclic amines of triterpenoids with HIV maturation inhibitory activity
JP2019519571A (en) * 2016-06-30 2019-07-11 ヴィーブ ヘルスケア ユーケー(ナンバー5)リミテッド Aza substitution inhibitors of human immunodeficiency virus replication
US11236122B2 (en) 2018-06-29 2022-02-01 Dfh Therapeutics Triterpene amine derivatives
US20230174570A1 (en) * 2020-02-11 2023-06-08 Hetero Labs Limited Novel triterpene derivatives as hiv inhibitors

Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5413999A (en) 1991-11-08 1995-05-09 Merck & Co., Inc. HIV protease inhibitors useful for the treatment of AIDS
US5679828A (en) 1995-06-05 1997-10-21 Biotech Research Labs, Inc. Betulinic acid and dihydrobetulinic acid derivatives and uses therefor
US20050209246A1 (en) 2004-03-15 2005-09-22 Yasutsugu Ueda Prodrugs of piperazine and substituted piperidine antiviral agents
US20050239748A1 (en) 2004-03-17 2005-10-27 Panacos Pharmaceuticals, Inc. Pharmaceutical salts of 3-O-(3',3'-dimethylsuccinyl) betulinic acid
WO2006053255A2 (en) 2004-11-12 2006-05-18 Panacos Pharmaceuticals, Inc. Novel betulin derivatives, preparation thereof and use thereof
US7354924B2 (en) 2001-02-02 2008-04-08 Bristol-Myers Squibb Company Composition and antiviral activity of substituted azaindoleoxoacetic piperazine derivatives
US7365221B2 (en) 2002-09-26 2008-04-29 Panacos Pharmaceuticals, Inc. Monoacylated betulin and dihydrobetulin derivatives, preparation thereof and use thereof
US20080207573A1 (en) 2006-10-16 2008-08-28 Myriad Genetics, Incorporated Compounds for treating viral infections
WO2009100532A1 (en) 2008-02-14 2009-08-20 Virochem Pharma Inc. NOVEL 17ß LUPANE DERIVATIVES
WO2011007230A2 (en) 2009-07-14 2011-01-20 Hetero Research Foundation Lupeol-type triterpene derivatives as antivirals
US20120142653A1 (en) 2010-06-04 2012-06-07 Bristol-Myers Squibb Company C-28 amides of modified c-3 betulinic acid derivatives as hiv maturation inhibitors
US20120142707A1 (en) 2010-06-04 2012-06-07 Bristol-Myers Squibb Company Modified c-3 betulinic acid derivatives as hiv maturation inhibitors

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5869535A (en) 1995-03-21 1999-02-09 The Board Of Trustees Of The University Of Illinois Method and composition for selectively inhibiting melanoma
US5962527A (en) 1995-03-21 1999-10-05 The Board Of Trustees Of The University Of Illinois Method and composition for treating cancers
WO2004089357A2 (en) 2003-04-02 2004-10-21 Regents Of The University Of Minnesota Anti-fungal formulation of triterpene and essential oil
WO2008127364A2 (en) * 2006-10-13 2008-10-23 Myriad Genetics, Inc. Antiviral compounds and use thereof
KR101886467B1 (en) 2011-01-31 2018-08-07 비브 헬스케어 유케이 (넘버4) 리미티드 C-17 and c-3 modified triterpenoids with hiv maturation inhibitory activity
JP6000283B2 (en) 2011-01-31 2016-09-28 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company C-28 amines of C-3 modified betulinic acid derivatives as HIV maturation inhibitors

Patent Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5413999A (en) 1991-11-08 1995-05-09 Merck & Co., Inc. HIV protease inhibitors useful for the treatment of AIDS
US5679828A (en) 1995-06-05 1997-10-21 Biotech Research Labs, Inc. Betulinic acid and dihydrobetulinic acid derivatives and uses therefor
US7354924B2 (en) 2001-02-02 2008-04-08 Bristol-Myers Squibb Company Composition and antiviral activity of substituted azaindoleoxoacetic piperazine derivatives
US7365221B2 (en) 2002-09-26 2008-04-29 Panacos Pharmaceuticals, Inc. Monoacylated betulin and dihydrobetulin derivatives, preparation thereof and use thereof
US20050209246A1 (en) 2004-03-15 2005-09-22 Yasutsugu Ueda Prodrugs of piperazine and substituted piperidine antiviral agents
US20050239748A1 (en) 2004-03-17 2005-10-27 Panacos Pharmaceuticals, Inc. Pharmaceutical salts of 3-O-(3',3'-dimethylsuccinyl) betulinic acid
WO2006053255A2 (en) 2004-11-12 2006-05-18 Panacos Pharmaceuticals, Inc. Novel betulin derivatives, preparation thereof and use thereof
US20080207573A1 (en) 2006-10-16 2008-08-28 Myriad Genetics, Incorporated Compounds for treating viral infections
WO2009100532A1 (en) 2008-02-14 2009-08-20 Virochem Pharma Inc. NOVEL 17ß LUPANE DERIVATIVES
WO2011007230A2 (en) 2009-07-14 2011-01-20 Hetero Research Foundation Lupeol-type triterpene derivatives as antivirals
US20120142653A1 (en) 2010-06-04 2012-06-07 Bristol-Myers Squibb Company C-28 amides of modified c-3 betulinic acid derivatives as hiv maturation inhibitors
US20120142707A1 (en) 2010-06-04 2012-06-07 Bristol-Myers Squibb Company Modified c-3 betulinic acid derivatives as hiv maturation inhibitors

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
CELL, vol. 9, 29 October 1999 (1999-10-29), pages 243 - 246
DRUG DISCOVERY TODAY, vol. 5, no. 5, May 2000 (2000-05-01), pages 183 - 194
DRUGS OF THE FUTURE, vol. 24, no. 12, 1999, pages 1355 - 1362
KASHIWADA, Y. ET AL., J. MED. CHEM., vol. 39, 1996, pages 1016 - 1017
MEANWELL, NICHOLAS A.; KADOW, JOHN F.: "Inhibitors of the entry of HIV into host cells", CURRENT OPINION IN DRUG DISCOVERY & DEVELOPMENT, vol. 6, no. 4, 2003, pages 451 - 461

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107428798A (en) * 2015-04-14 2017-12-01 Viiv保健英国第四有限公司 The method for producing HIV maturation inhibitor
WO2017017607A1 (en) 2015-07-28 2017-02-02 Glaxosmithkline Intellectual Property (No.2) Limited Betuin derivatives for preventing or treating hiv infections
WO2017017609A1 (en) 2015-07-28 2017-02-02 Glaxosmithkline Intellectual Property (No.2) Limited Betuin derivatives for preventing or treating hiv infections
WO2017051355A1 (en) 2015-09-24 2017-03-30 Glaxosmithkline Intellectual Property (No.2) Limited Compounds with hiv maturation inhibitory activity
WO2017125870A1 (en) 2016-01-20 2017-07-27 Glaxosmithkline Intellectual Property (No.2) Limited Amine derivatives of lupanes with hiv maturation inhibitory activity
WO2019207460A1 (en) 2018-04-24 2019-10-31 VIIV Healthcare UK (No.5) Limited Compounds with hiv maturation inhibitory activity
EP4321164A2 (en) 2018-04-24 2024-02-14 ViiV Healthcare UK (No.5) Limited Compounds with hiv maturation inhibitory activity
WO2020165741A1 (en) 2019-02-11 2020-08-20 Hetero Labs Limited Novel triterpene derivatives as hiv inhibitors
EP4248960A2 (en) 2019-02-11 2023-09-27 Hetero Labs Limited Novel triterpene derivatives as hiv inhibitors

Also Published As

Publication number Publication date
AU2012312485A1 (en) 2014-05-08
CN103814042B (en) 2015-08-19
JP2014526554A (en) 2014-10-06
JP6100786B2 (en) 2017-03-22
US8754069B2 (en) 2014-06-17
KR20140069167A (en) 2014-06-09
EP2758419B8 (en) 2017-04-19
ES2611727T3 (en) 2017-05-10
BR112014006559A2 (en) 2017-03-28
CA2849475A1 (en) 2013-03-28
MX2014002936A (en) 2014-04-25
EA023463B1 (en) 2016-06-30
TW201317255A (en) 2013-05-01
CN103814042A (en) 2014-05-21
EA201490643A1 (en) 2014-06-30
US20130072465A1 (en) 2013-03-21
EP2758419B1 (en) 2016-10-19
SG11201400331WA (en) 2014-08-28
AU2012312485B2 (en) 2016-09-01
IL231421A0 (en) 2014-04-30
IL231421A (en) 2015-11-30
EP2758419A1 (en) 2014-07-30

Similar Documents

Publication Publication Date Title
AU2012312485B2 (en) Novel betulinic acid derivatives with antiviral activity
EP2670764B1 (en) C-28 amines of c-3 modified betulinic acid derivatives as hiv maturation inhibitors
EP2576585B1 (en) Modified c-3 betulinic acid derivatives as hiv maturation inhibitors
EP2576586B1 (en) C-28 amides of modified c-3 betulinic acid derivatives as hiv maturation inhibitors
AU2014218754B2 (en) C-3 alkyl and alkenyl modified betulinic acid derivatives useful in the treatment of HIV
EP2847208B1 (en) C-17 bicyclic amines of triterpenoids with hiv maturation inhibitory activity
WO2015195776A1 (en) Betulinic acid derivatives with hiv maturation inhibitory activity
AU2015346303B2 (en) Oxolupene derivatives
AU2015346314B2 (en) Extended betulinic acid analogs
US10047118B2 (en) C17-aryl substituted betulinic acid analogs
NZ614871A (en) C-17 and c-3 modified triterpenoids with hiv maturation inhibitory activity
NZ614871B2 (en) C-17 and c-3 modified triterpenoids with hiv maturation inhibitory activity

Legal Events

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

Ref document number: 12766825

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 231421

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: MX/A/2014/002936

Country of ref document: MX

REEP Request for entry into the european phase

Ref document number: 2012766825

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2012766825

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2849475

Country of ref document: CA

Ref document number: 2014531936

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 201490643

Country of ref document: EA

ENP Entry into the national phase

Ref document number: 20147010087

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2012312485

Country of ref document: AU

Date of ref document: 20120920

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112014006559

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112014006559

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20140319