WO2009092293A1 - Composés à base de pyrollidine - Google Patents

Composés à base de pyrollidine Download PDF

Info

Publication number
WO2009092293A1
WO2009092293A1 PCT/CN2009/000067 CN2009000067W WO2009092293A1 WO 2009092293 A1 WO2009092293 A1 WO 2009092293A1 CN 2009000067 W CN2009000067 W CN 2009000067W WO 2009092293 A1 WO2009092293 A1 WO 2009092293A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
methyl
group
compound
allyl
Prior art date
Application number
PCT/CN2009/000067
Other languages
English (en)
Inventor
Li Chen
Eric Dale Jones
Dawei Ma
Dean Cameron Baylis
Ben Li
Jonathan Alan Victor Coates
Xin Xie
David Ian Rhodes
Renhai Chen
John Joseph Deadman
Original Assignee
Shanghai Targetdrug Co., Ltd.
Avexa Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from PCT/CN2008/000137 external-priority patent/WO2009089659A1/fr
Application filed by Shanghai Targetdrug Co., Ltd., Avexa Limited filed Critical Shanghai Targetdrug Co., Ltd.
Priority to CN200980110077.1A priority Critical patent/CN101990536B/zh
Publication of WO2009092293A1 publication Critical patent/WO2009092293A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D451/00Heterocyclic compounds containing 8-azabicyclo [3.2.1] octane, 9-azabicyclo [3.3.1] nonane, or 3-oxa-9-azatricyclo [3.3.1.0<2,4>] nonane ring systems, e.g. tropane or granatane alkaloids, scopolamine; Cyclic acetals thereof
    • C07D451/02Heterocyclic compounds containing 8-azabicyclo [3.2.1] octane, 9-azabicyclo [3.3.1] nonane, or 3-oxa-9-azatricyclo [3.3.1.0<2,4>] nonane ring systems, e.g. tropane or granatane alkaloids, scopolamine; Cyclic acetals thereof containing not further condensed 8-azabicyclo [3.2.1] octane or 3-oxa-9-azatricyclo [3.3.1.0<2,4>] nonane ring systems, e.g. tropane; Cyclic acetals thereof
    • C07D451/04Heterocyclic compounds containing 8-azabicyclo [3.2.1] octane, 9-azabicyclo [3.3.1] nonane, or 3-oxa-9-azatricyclo [3.3.1.0<2,4>] nonane ring systems, e.g. tropane or granatane alkaloids, scopolamine; Cyclic acetals thereof containing not further condensed 8-azabicyclo [3.2.1] octane or 3-oxa-9-azatricyclo [3.3.1.0<2,4>] nonane ring systems, e.g. tropane; Cyclic acetals thereof with hetero atoms directly attached in position 3 of the 8-azabicyclo [3.2.1] octane or in position 7 of the 3-oxa-9-azatricyclo [3.3.1.0<2,4>] nonane ring system

Definitions

  • the present invention provides compounds of Formula (I) and their use in the treatment or prophylaxis of HIV.
  • HIV human immunodeficiency virus
  • HIV infection follows a path of the virus particle interacting non-specifically with adhesion proteins on the host immune cell surface and then binding to the host cell surface receptor CD4.
  • Binding of the HIV-I gpl20 envelope glycoprotein to CD4 induces conformational changes in gpl20 that create or expose a binding site for a co-receptor that can be the CCR5 receptor (a G-protein coupled receptor also known as C-C chemokine receptor type 5; C-C CKR-5; CC-CKR-5; CCR-5; CCR5; CCR5 chemokine receptor; CD195 antigen; CHEMRl 3; HIV-I fusion coreceptor; MIP-I alpha receptor) resulting in fusion of the virus particle with the cell.
  • Strains of HIV that utilise the CCR5 receptor are called CCR5-tropic strains or R5 isolates.
  • CXCR4 residues dispersed throughout the extracellular domain of CXCR4 are involved in gpl20 docking, viral fusion and entry; each HIV-I isolate uses a slightly different subset of CXCR4 residues in order to gain entry into the target cell.
  • the gpl20 binding sites on CCR5 and CXCR4 comprise negatively charged and tyrosine residues. Certain mutations in CXCR4 even enable it to mediate the entry of R5 isolates. Similarities between CCR5 and CXCR4 gpl20-binding sites are further underscored by the ability of R5X4 isolates to interact with both co-receptors.
  • the contents of the virus are released into the cytoplasm where reverse transcription of the HIV genome occurs.
  • a double stranded proviral DNA copy is produced.
  • the proviral DNA is transported to the nucleus in a complex known as the pre integration complex (PIC) which contains integrase and other viral and possibly cellular proteins.
  • PIC pre integration complex
  • the proviral DNA is integrated into the host cell genome via the action of integrase.
  • transcription and translation of the viral genome can occur resulting in the production of viral proteins and a new viral RNA genome.
  • These proteins and genome assemble at the cell surface and, depending on cell type, possibly other intracellular membranous compartments. Assembled particles then bud out from the cell and during, or soon after, this process mature into infectious HIV particles through the action of the viral protease.
  • the integration of the proviral genome into the host cell genome requires the action of an integrase which carries out this process in at least three steps, possibly four.
  • the first step involves the assembly of the viral genome into a stable nucleoprotein complex, secondly, processing of two nucleotides from the 3' termini of the genome to give staggered ends with free 3' OH residues and thirdly the transfer of these ends into the host cell genome.
  • the final step involves the gap filling and repair of the insertion site in the host genome. There is still some conjecture over whether the integrase performs this final step or whether it is carried out by cellular repair enzymes.
  • HIV infection can be treated with a number of inhibitors on the market which target reverse transcriptase, protease, integrase or entry into the cell. Treatment of HIV infection with these, or a combination of these, drugs is known to be an effective treatment for AIDS and similar diseases. Shortcomings with the current inhibitors include the rapid emergence and increased incidence of resistance and numerous side effects and hence there is a need for new classes of inhibitors.
  • the present invention provides a compound of formula ( I )
  • Z is selected from carbonyl and -CH(CO 2 H)-;
  • R 1 is selected from the group consisting of alkyl, cycloalkyl, aryl, heteroaryl, heterocyclyl, which may be substituted with one or more suitable substituent(s);
  • R 2 is selected from the group consisting of -OH, -O-Ci -6 alkyl, -OC(O)-Ci -6 alkyl, and NR 8 Rg; each of R 8 and Rg are independently selected from the group consisting of H, C 1-6 alkyl, -O(CO)-C 1-6 alkyl, and -S(O) 2 -R 10 ; R 10 is selected from the group consisting of -C 1-6 alkyl and aryl;
  • R 3 is selected from the group consisting of H, C 1-6 alkyl, aryl, and heteroaryl;
  • R 4 is selected from the group consisting of H, alkylene, aryl; cycloalkyl and alkylcycloaklyl; or R 4 and Rn, together with the atoms to which they are linked, form an optionally substituted 5- or 7-membered heterocycle;
  • each of R 5 and R 5' is selected from the group consisting of H, CH 3 , and OH, or R 5 and R 5 • together with the carbon atom to which they are attached form a carbonyl group, provided that both R 5 and R 5' are not both OH;
  • R 6 is absent or is 1 to 5 substituents selected from the group consisting of CF 3 , OCF 3 , NO 2 , SO 2 R 12 , NC(O)OC 1-6 alkyl, CrCsalkyl, C 3 -C 6 cycloalkyl, Qaryl, heterocyclyl, heteroaryl, Q-CsalkylOH, alkylaryl, OH, OCrsalkyl, halo, CN 5 CO 2 H, CO 2 C 1-3 alkyl, CONH 2 , CONH(C 1-3 alkyl), CON(C 1-3 alkyl) 2 , NH 2 , NH(C 1-3 alkyl) and N(C 1-3 alkyl) 2 ;
  • R 12 is selected from the group consisting of Ci -6 alkenyl and NRi 3 R 14; each of R 13 and R 14 is independently selected from the group consisting of H, alkyl, and cycloalkyl, or R 13 and Ri 4 together with the nitrogen atom to which
  • each of R 7 and R 7' is selected from the group consisting of H, CH 3 , and OH, or R 7 and R 7' together with the carbon atom to which they are attached form a carbonyl group, provided that both R 7 and R 7 > are not both OH;
  • A is absent or is selected from the group consisting of -CH 2 CH 2 -, -CH 2 CH 2 CH 2 -, and -CH 2 OCH 2 -;
  • Y is absent or is carbonyl
  • L is absent or is selected from alkylene and alkenylene
  • C is selected from the group consisting of cycloalkyl, aryl, and heteroaryl;
  • X is absent or is selected from NR 11 and O;
  • R 11 is H or R 4 and R 11 , together with the atoms to which they are linked, form an optionally substituted 5- or 7-membered heterocycle;
  • R 6 is selected from the group consisting of SO 2 Ri 2 and OCF 3 ; or C and R 6 together with the atoms to which they are linked, form
  • a further object of present invention is to provide a method of treating or preventing HIV infection in a subject.
  • the method comprises administering to said subject an effective amount of a compound of Formula (I).
  • a yet further object of present invention is to provide a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula (I) and a pharmaceutically acceptable carrier, diluent or excipient.
  • the present invention provides a compound of formula (I)
  • Z is selected from carbonyl and -CH(CO 2 H)-;
  • R 1 is selected from the group consisting of alkyl, cycloalkyl, aryl, heteroaryl, heterocyclyl , which may be substituted with one or more suitable substituent(s);
  • R 2 is selected from the group consisting of -OH, -O-C 1-6 alkyl, -OC(O)-C 1-6 alkyl,and NR 8 R 9 ; each of R 8 and R 9 are independently selected from the group consisting of H, C 1-6 alkyl, -O(CO)-C 1-6 alkyl, and -S(O) 2 -R 10 ; R 10 is selected from the group consisting of -C 1-6 alkyl and aryl;
  • R 3 is selected from the group consisting of H, C 1-6 alkyl, aryl, and heteroaryl;
  • R 4 is selected from the group consisting of H, C 1-4 alkyl, alkylene, aryl, cycloalkyl and alkylcycloaklyl; or R 4 and R 11 , together with the atoms to which they are linked, form an optionally substituted 5- or 7-membered heterocycle;
  • each of R 5 and R 5 - is selected from the group consisting of H, CH 3 , and OH, or R 5 and R 5 - together with the carbon atom to which they are attached form a carbonyl group, provided that both R 5 and R 5 ' are not both OH;
  • R 6 is absent or is 1 to 5 substituents selected from the group consisting of CF 3 , OCF 3 , NO 2 , SO 2 Ri 2 , NC(O)OC 1-6 alkyl, Q-Csalkyl, C 3 -C 6 cycloalkyl, C 6 aryl, heterocyclyl, heteroaryl, C r C 3 alkylOH, alkylaryl, OH, OCr 3 alkyl, halo, CN, CO 2 H, CO 2 C 1-3 alkyl, CONH 2 , CONH(C 1-3 alkyl), CON(C 1- C 3 alkyl) 2 , NH 2 , NH(C 1-3 alkyl) and N(C 1-3 alkyl) 2 ; R 12 is selected from the group consisting of C 1-6 alkenyl and NR 13 R 14 ; each of R 13 and R 14 is independently selected from the group consisting of H, alkyl, and cycloalkyl, or R 13 and R 14 together with the nitrogen
  • each of R 7 and R 7 > is selected from the group consisting of H, CH 3 , and OH, or R 7 and R 7 ' together with the carbon atom to which they are attached form a carbonyl group, provided that both R 7 and R 7 ' are not both OH;
  • A is absent or is selected from the group consisting of -CH 2 CH 2 -, -CH 2 CH 2 CH 2 -, and -CH 2 OCH 2 -;
  • Y is absent or is carbonyl
  • L is absent or is selected from alkylene and alkenylene
  • C is selected from the group consisting of cycloalkyl, aryl, and heteroaryl;
  • X is absent or is selected from NR 11 and O;
  • R 11 is H or R 4 and R 11 , together with the atoms to which they are linked, form an optionally substituted 5- or 7-membered heterocycle;
  • R 6 is selected from the group consisting of SO 2 R 12 and OCF 3 ; or C and R 6 , together with the atoms to which they are linked, form
  • R 1 is selected from the group consisting of-OH, C 1-6 alkyl, C 3-8 cycloalkyl, aryl, heteroaryl, and heterocyclyl, preferably C 3-8 cycloalkyl, more preferably cyclopentyl.
  • R 2 is selected from the group consisting of -O-C 1-6 alkyl, -OC(O)-C 1-6 alkyl, and NR 8 R 9 .
  • R 4 is preferably selected from C 1-6 alkenyl and cycloalkyl, more preferably selected from allyl and -CH 2 cyclopropyl.
  • the aboved-mentioned compounds, wherein C is selected from the group consisting of benzodioxazole, m-methoxyphenyl, phenyl substituted with at least one substiruent selected from SO 2 C 1-6 alkyl and -OCF 3 .
  • the aboved-mentioned compounds, wherein Z is preferably carbonyl are preferably carbonyl.
  • R 6 is 1 to 5 substituents selected from the group consisting of CF 3 , OCF 3 , NO 2 , SO 2 R12 5 NC(O)OCi -6 alkyl, C ⁇ alkyl, C 3 - 6 cycloalkyl, C 6 aryl, heterocyclyl, heteroaryl, CrsalkylOH, alkylaryl, OH, OC r C 3 alkyl, halo, CN, CO 2 H, CO 2 C I-3 alkyl, CONH 2 , CONH(C 1-3 alkyl), CON(C I-3 alkyl) 2 , NH 2 , NH(C 1-3 alkyl) , and N(C 1-3 alkyl) 2 .
  • Z is carbonyl
  • R 1 is selected from the group consisting of cycloalkyl, aryl, heteroaryl, heterocyclyl;
  • each ofR 2 and R 3 is selected from the group consisting of H, C 1-6 alkyl, aryl, and heteroaryl;
  • R 4 is selected from the group consisting of H, Ci -4 alkyl, alkylene, aryl, cycloalkyl and alkylcycloaklyl; or R 4 and R 11 , together with the atoms to which they are linked, form an optionally substituted 5- or 7-membered heterocycle;
  • each ofR 5 and R 5 > is H
  • R 6 is absent or is 1 to 5 substituents selected from the group consisting of CF 3 , OCF 3 , NO 2 , SO 2 R 12 , NC(O)OC 1-6 alkyl, Ci-C 3 alkyl, C 3 -C 6 cycloalkyl, C 6 aryl, heterocyclyl, heteroaryl, Ci-QjalkylOH, alkylaryl, OH, OC ⁇ alkyl, halo, CN, CO 2 H, CO 2 C 1-3 alkyl, CONH 2 , CONH(Ci -3 alkyl), CON(d.C 3 alkyl)2, NH 2 , NH(C 1-3 alkyl) and N(C 1-3 alkyl) 2 ; R 12 is selected from the group consisting of C 1-6 alkenyl and NR 13 Rj 4; each OfRi 3 and R 14 is independently selected from the group consisting of H, alkyl, and cycloalkyl, or Ri 3 and R 14 together with the nitrogen atom to
  • each of R 7 and R 7 - is H; Y is carbonyl;
  • L is alkylene
  • C is aryl; the said groups which may be substituted with one or more suitable substituent(s).
  • the compound of Formula (I) is a compound of any one of the Examples, more preferably:
  • the invention provides an intermediate of formula II useful of formula I :
  • R 3 is selected from the group consisting of H, C 1-6 alkyl, aryl, and heteroary.
  • halo or halogen refers to fluorine (fluoro), chlorine (chloro), bromine (bromo) or iodine (iodo).
  • alkyl either used alone or in compound terms such as NH(alkyl) or N(alkyl) 2 , refers to monovalent straight chain or branched hydrocarbon groups, having 1 to 3, 1 to 6, 1 to 10 or 1 to 21 carbon atoms as appropriate.
  • suitable alkyl groups include, but are not limited to methyl, ethyl, propyl, isopropyl, n-butyl, sec-butyl, tert-butyl, pentyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 2-, 3- or 4-methylpentyl, 2-ethylbutyl, n-hexyl or 2-, 3-, 4- or 5-methylpentyl.
  • alkenyl refers to straight chain or branched hydrocarbon groups having one or more double bonds between carbon atoms. Suitable alkenyl groups include, but are not limited to ethenyl, allyl, propenyl, isopropenyl, butenyl, pentenyl and hexenyl.
  • alkynyl refers to straight chain or branched hydrocarbon groups containing one or more triple bonds. Suitable alkynyl groups include, but are not limited to ethynyl, propynyl, butynyl, pentynyl and hexenyl.
  • cycloalkyl refers to cyclic hydrocarbon groups. Suitable cycloalkyl groups include, but are not limited to cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.
  • aryl refers to a C 6 -C 10 aromatic hydrocarbon group, for example phenyl or naphthyl.
  • arylalkyl includes, for example, benzyl.
  • heterocycle when used alone or in compound words includes monocyclic, polycyclic, fused or conjugated hydrocarbon residues, preferably C 3-6 ,wherein one or more carbon atoms (and where appropriate, hydrogen atoms attached thereto) are replaced by a heteroatom so as to provide a non-aromatic residue.
  • the heterocycle may be saturated or unsaturated. Suitable heteroatoms include, O, N and S. Where two or more carbon atoms are replaced, this may be by two or more of the same heteroatom or by different heteroatoms.
  • heteroaryl includes a 5- or 6-membered heteroaromatic ring containing one or more heteroatoms selected from O, N and S.
  • Suitable examples of heteroaryl groups include tetrazolyl, 1,2,3-triazolyl, 1,2,4-triazolyl, imidazolyl, pyrazolyl, pyridinyl, pyrimidinyl, oxazolyl, oxadiazolyl etc.
  • the heteroaromatic ring may be fused to a 5- or 6- aromatic or heteroaromatic ring to form a bicyclic aromatic system eg benzofuran or benzooxadiazole.
  • Each alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heterocyclyl, or heteroaryl group may be optionally substituted with one or more of CrC 3 alkyl, C 3 -C 6 cycloalkyl, C 6 aryl, heterocyclyl, heteroaryl, Q-CsalkylOH, alkylaryl, OH, OCi-C 3 alkyl, halo, CN, NO 2 , CO 2 H, CO 2 Ci.C 3 alkyl, CONH 2 , CONH(Ci.C 3 alkyl), CON(C 1- C 3 alkyl) 2 , trifluoromethyl, NH 2 , NHCCj.Caalkyl), N(Ci-C 3 alkyl) 2 , S(O) 2 NH 2 , SO 2 NH(Ci -3 alkyl), SO 2 (C 1-3 alkyl) 2 , and S ⁇ 2 C 1-3 alkyl.
  • an optionally substituted aryl group may be 4-methylphenyl or 4-hydroxyphenyl group, and an optionally substituted alkyl group may be 2-hydroxyethyl, trifluoromethyl, or difluoromethyl.
  • Each optional substituent may also be optionally substituted.
  • optional substituents also include suitable nitrogen protecting groups (see “Protective Groups in Organic Synthesis” Theodora. Greene and Peter Wuts, third edition, Wiley Interscience, 1999).
  • salts of the compound of formula I are preferably pharmaceutically acceptable, but it will be appreciated that non-pharmaceutically acceptable salts also fall within the scope of the present invention, since these are useful as intermediates in the preparation of pharmaceutically acceptable salts.
  • Suitable pharmaceutically acceptable salts include, but are not limited to, salts of pharmaceutically acceptable inorganic acids such as hydrochloric, sulphuric, phosphoric, nitric, carbonic, boric, sulfamic, and hydrobromic acids, or salts of pharmaceutically acceptable organic acids such as acetic, propionic, butyric, tartaric, maleic, hydroxymaleic, fumaric, malic, citric, lactic, mucic, gluconic, benzoic, succinic, oxalic, phenylacetic, methanesulphonic, toluenesulphonic, benzenesulphonic, salicylic, sulphanilic, aspartic, glutamic, edetic, stearic, palmitic, oleic, lauric, pantothenic, tannic, ascorbic and valeric acids.
  • pharmaceutically acceptable inorganic acids such as hydrochloric, sulphuric, phosphoric, n
  • Base salts include, but are not limited to, those formed with pharmaceutically acceptable cations, such as sodium, potassium, lithium, calcium, magnesium, zinc, ammonium, alkylammonium such as salts formed from triethylamine, alkoxyammonium such as those formed with ethanolamine and salts formed from ethylenediamine, choline or amino acids such as arginine, lysine or histidine.
  • pharmaceutically acceptable cations such as sodium, potassium, lithium, calcium, magnesium, zinc, ammonium, alkylammonium such as salts formed from triethylamine, alkoxyammonium such as those formed with ethanolamine and salts formed from ethylenediamine, choline or amino acids such as arginine, lysine or histidine.
  • This invention also encompasses prodrugs of compounds of formula I.
  • This invention also encompasses methods of treating or preventing disorders in a subject that can be treated or prevented by the inhibition of AIDS and other disorders that can be treated by inhibition of the integrase enzyme by administering prodrugs of compounds of the formula I.
  • Compounds of formula I having free amino, amido, hydroxy or carboxylic groups can be converted into prodrugs.
  • Prodrugs include compounds wherein an amino acid residue, or a polypeptide chain of two or more (eg, two, three or four) amino acid residues which are covalently joined through peptide bonds to free amino, hydroxy and carboxylic acid groups of compounds of formula I.
  • the compounds of formula I may possess asymmetric centres and are therefore capable of existing in more than one stereoisomeric form.
  • the invention thus also relates to compounds in substantially pure isomeric form at one or more asymmetric centres eg., greater than about 90% ee, such as about 95% or 97% ee or greater than 99% ee, as well as mixtures, including racemic mixtures, thereof.
  • Such isomers may be prepared by asymmetric synthesis, for example using chiral intermediates, or by chiral resolution.
  • a further object of the present invention is to provide a method of treating of treatment or prophylaxis of a HIV infection in a subject, the method comprising administering to said subject an effective amunt of a compound of Formula (I) or a pharmaceutically acceptable derivative, salt or prodrug thereof.
  • effective amount means the amount of the subject composition that will elicit the reduction in viral load or inhibition of viral replication that is being sought by the researcher, veterinarian, medical doctor or other clinician.
  • treatment does not necessarily mean that the viral infection is completely cured.
  • treatment encompasses any level of reduction of the viral load and/or inhibition of replication in the subject being treated.
  • administering a should be understood to mean providing a compound of the invention to the individual in need of treatment.
  • a yet further object of the present invention is to provide a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable derivative, salt or prodrug thereof and a pharmaceutically acceptable carrier, diluent or excipient.
  • compositions of the present invention may contain other therapeutic agents and may be formulated, for example, by employing conventional solid or liquid vehicles or diluents, as well as pharmaceutical additives of a type appropriate to the mode of desired administration (for example, excipients, binders, preservatives, stabilizers, flavors, etc.) according to techniques such as those well known in the art of pharmaceutical formulation.
  • the compounds of the present invention may be administered by any suitable means, for example, parenterally, such as by subcutaneous, intravenous, intramuscular, or intracisternal injection or infusion techniques (e.g., as sterile injectable aqueous or non-aqueous solutions or suspensions).
  • parenterally such as by subcutaneous, intravenous, intramuscular, or intracisternal injection or infusion techniques (e.g., as sterile injectable aqueous or non-aqueous solutions or suspensions).
  • compositions include those for oral, rectal, nasal, topical (including buccal and sub-lingual), vaginal or parenteral (including intramuscular, sub-cutaneous and intravenous) administration or in a form suitable for administration by inhalation or insufflation.
  • the compounds of the invention may thus be placed into the form of pharmaceutical compositions and unit dosages thereof, and in such form may be employed as solids, such as tablets or filled capsules, or liquids as solutions, suspensions, emulsions, elixirs or capsules filled with the same, all for oral use, in the form of suppositories for rectal administration; or in the form of sterile injectable solutions for parenteral (including subcutaneous) use.
  • mammals including, but not limited to, cows, sheep, goats, horses, dogs, cats, guinea pigs, rats or other bovine, ovine, equine, canine, feline, rodent or murine species can be treated.
  • the method can also be practiced in other species, such as avian species (e.g., chickens).
  • the subjects treated in the above method are mammals, including, but not limited to, cows, sheep, goats, horses, dogs, cats, guinea pigs, rats or other bovine, ovine, equine, canine, feline, rodent or murine species, and preferably a human being, male or female.
  • composition as used herein is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts.
  • pharmaceutically acceptable it is meant the carrier, diluent or excipient must be compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
  • compositions for the administration of the compounds of this invention may conveniently be presented in dosage unit form and may be prepared by any of the methods well known in the art of pharmacy. All methods include the step of bringing the active ingredient into association with the carrier which constitutes one or more accessory ingredients.
  • the pharmaceutical compositions are prepared by uniformly and intimately bringing the active ingredient into association with a liquid carrier or a finely divided solid carrier or both, and then, if necessary, shaping the product into the desired formulation.
  • the active object compound is included in an amount sufficient to produce the desired effect upon the process or condition of diseases.
  • composition is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts.
  • the pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleagenous suspension.
  • This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example as a solution in 1,3-butane diol.
  • the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • the pharmaceutical composition and method of the present invention may further comprise other therapeutically active compounds which are usually applied in the treatment of the above mentioned pathological conditions. Selection of the appropriate agents for use in combination therapy may be made by one of ordinary skill in the art, according to conventional pharmaceutical principles.
  • the combination of therapeutic agents may act synergistically to effect the treatment or prevention of the various disorders described above. Using this approach, one may be able to achieve therapeutic efficacy with lower dosages of each agent, thus reducing the potential for adverse side effects.
  • an appropriate dosage level will generally be about 0.01 to 500 mg per kg patient body weight per day which can be administered in single or multiple doses.
  • the dosage level will be about 0.1 to about 250 mg/kg per day; more preferably about 0.5 to about 100 mg/kg per day.
  • a suitable dosage level may be about 0.01 to 250 mg/kg per day, about 0.05 to 100 mg/kg per day, or about 0.1 to 50 mg/kg per day. Within this range the dosage may be 0.05 to 0.5, 0..5 to 5 or 5 to 50 mg/kg per day.
  • compositions are preferably provided in the form of tablets containing 1.0 to 1000 milligrams of the active ingredient, particularly 1.0, 5.0, 10.0, 15.0. 20.0, 25.0, 50.0, 75.0, 100.0, 150.0, 200.0, 250.0, 300.0, 400.0, 500.0, 600.0, 750.0, 800.0, 900.0, and 1000.0 milligrams of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated.
  • the compounds may be administered on a regimen of 1 to 4 times per day, preferably once or twice per day.
  • the compounds of the present invention can be prepared using the methods described in Schemes 1-10.
  • Compound 4 can be synthesized starting from benzyl alcohol 1. Reaction of benzyl alcohol 1 with triphosgene followed by treatment of the resulting chloroformate 2 with the amine 3 provides compound 4.
  • a variety of acid 5 can be coupled with the amine 3 using coupling agents such as EDCI and HOBt to give the amide 6.
  • the urea 8 can be obtained by reaction of a variety of isocyanates 7 with the amine 3 out lined in Scheme 3.
  • the urea 11 can be synthesized starting from benzyl amine 9. Treatment of compound 9 with triphosgene converts the amine to isocyanate 10 and the resulting compound is reacted with the amine 3 to give urea 11.
  • Scheme 5
  • a variety of final compounds 24 can be synthesized using the sequence shown in Scheme 7.
  • Boc protection of amine 18 followed by oxidation using an oxidant such as DMSO and (COCl) 2 affords ketone 20.
  • Reductive amination of an amine with ketone 20 using a reducing agent such as sodium triacetoxyborohydride provides intermediate 21.
  • Treatment of compound 21 with chloroformate 2 or isocyanate 10 provides compound 22. Removal of the Boc by reaction with trifluoroacetic acid followed by coupling with an acid affords the amide 24.
  • Step F and G Benzoic] [1, 2, 5]oxadiazol-5-ylmethyl allyl(l-(((3S, 4R)-l-(cyclopentanecarbonyl)-4-
  • This compound was prepared substantially as described in Example 24 using the starting material from step A, which was used without further purification in the next step.
  • step C To the alcohol from step C (0.5 g, 1.73 mmol) as a solution in DMF (6 mL) were added TBDMSCl (0.286 g, 1.9 mmol) and imidazole (0.294 g, 4.325 mmol) and the mixture was stirred for 18 h at room temperature. After dilution with water and extraction with EA, the organic phases were washed, dried, and evaporated, affording the title product (0.65 g, 92 %) as pale yellow foam, which was used without further purification in the next step.
  • Step E To the alcohol from step C (0.5 g, 1.73 mmol) as a solution in DMF (6 mL) were added TBDMSCl (0.286 g, 1.9 mmol) and imidazole (0.294 g, 4.325 mmol) and the mixture was stirred for 18 h at room temperature. After dilution with water and extraction with EA, the organic phases were washed, dried, and evaporated, affording
  • Tetrabutylammonium fluoride (0.5 mL, 0.5 mmol, 1 N in THF) was added to a solution of the silyl ether from step E (0.14 g, 0.335 mmol) in THF (2 mL) and stirred at room temperature overnight in the dark. The reaction mixture was filtered over silica gel and purified by chromatography (eluant: PE/EA, 1/1) to afford the product as white crystals (0.095 g, 98 %).
  • the reaction was allowed to warm to room temperature as the ice melted and stirred for 12 h at room temperature.
  • the reaction was diluted with EA, then washed with saturated bicarbonate and brine, dried over Na 2 SO 4 , and concentrated.
  • the residue was chromatographed on silica gel (50%- 100% EA in PE) to give the title compound as a white foam (0.12 g, 60%).
  • PBMC peripheral blood mononuclear cells
  • buffy coats obtained from the Australian Red Cross Blood Service, South Melbourne
  • HIV-I and hepatitis B virus-seronegative donors using a Ficoll gradient.
  • PBMC peripheral blood mononuclear cells
  • PHA PHA
  • Cells were seeded into 96 well microtitre plates at 200,000 cells per 50 ⁇ l per well in RF-10 containing 10U/mL IL-2 (RF-10/IL2). Compounds were prepared to 4 x final concentration in RF-10/IL2, and 30 ⁇ L added to cells.
  • Virus (40 ⁇ L in RF-10/IL2 containing 1400 pfu) was added to each well or 40 ⁇ L RF-10/IL2 for negative controls and for assaying compound cytotoxicity. After 24 hrs, an additional 90 ⁇ L of media or media containing 1 x compound was added to each well. At 4 days post infection, 100 ⁇ L of media was removed from each well and replaced with 100 ⁇ l of fresh media with or without compound. Forty eight hours later supernatants were harvested and levels of extracellular p24 determined. Supernatants were diluted 1 in 10,000 and p24 levels assayed using the Vironostika p24 assay kit. EC 50 was calculated as the concentration required to inhibit HIV p24 production to 50% that of no drug controls.

Abstract

L'invention concerne un composé de formule (I) ou un dérivé, sel ou promédicament de celui-ci acceptable sur le plan pharmaceutique, qui peut inhiber la réplication du VIH.
PCT/CN2009/000067 2008-01-18 2009-01-16 Composés à base de pyrollidine WO2009092293A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN200980110077.1A CN101990536B (zh) 2008-01-18 2009-01-16 基于吡咯烷的化合物

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
PCT/CN2008/000137 WO2009089659A1 (fr) 2008-01-18 2008-01-18 Composés à base de pyrollidine
CNPCT/CN2008/000137 2008-01-18
US2257808P 2008-01-21 2008-01-21
US61/022578 2008-01-21

Publications (1)

Publication Number Publication Date
WO2009092293A1 true WO2009092293A1 (fr) 2009-07-30

Family

ID=40900770

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2009/000067 WO2009092293A1 (fr) 2008-01-18 2009-01-16 Composés à base de pyrollidine

Country Status (1)

Country Link
WO (1) WO2009092293A1 (fr)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101885721A (zh) * 2010-06-11 2010-11-17 银杏树药业(苏州)有限公司 作为ccr5拮抗剂的吡咯衍生物、制备该化合物的方法及其应用
WO2012143526A1 (fr) * 2011-04-20 2012-10-26 Centre National De La Recherche Scientifique (C.N.R.S) 1,2,3-triazoles 1,4-disubstituées, leurs procédés de préparation et leurs utilisations diagnostiques et thérapeutiques
US8658797B2 (en) 2011-02-25 2014-02-25 Helsinn Healthcare Sa Asymmetric ureas and medical uses thereof
US10501479B2 (en) 2016-03-22 2019-12-10 Helsinn Healthcare Sa Benzenesulfonyl-asymmetric ureas and medical uses thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1706824A (zh) * 2004-06-09 2005-12-14 上海靶点药物有限公司 作为ccr5拮抗剂的化合物
CN1939916A (zh) * 2005-09-05 2007-04-04 上海靶点药物有限公司 治疗艾滋病的化合物
CN101007800A (zh) * 2006-01-26 2007-08-01 上海靶点药物有限公司 用于制备尼非韦罗的化合物及其制备方法

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1706824A (zh) * 2004-06-09 2005-12-14 上海靶点药物有限公司 作为ccr5拮抗剂的化合物
CN1939916A (zh) * 2005-09-05 2007-04-04 上海靶点药物有限公司 治疗艾滋病的化合物
CN101007800A (zh) * 2006-01-26 2007-08-01 上海靶点药物有限公司 用于制备尼非韦罗的化合物及其制备方法

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
DAWEI M.A. ET AL.: "Synthesis and Biological Evaluation of 1,3,3,4-Tetrasubstituted Pyrrolidine CCR5 Receptor Antagonists. Discovery of a Potent and Orally Bioavailable Anti-HIV Agent", CHEMMEDCHEM, 2007, pages 187 - 193 *
JEFFREY J. HALE ET AL.: "1,3,4-Trisubstituted Pyrrolidine CCR5 Receptor Antagonists. Part 1: Discovery of the Pyrrolidine Scaffold and Determination of Its Stereochemical Requirements", BIOORG. MED. CHEM., vol. 11, 2001, pages 1437 - 1440 *
LYNCH C.L. ET AL.: "1,3,4-Trisubstituted Pyrrolidine CCR5 Receptor Antagonists. Part 4: Synthesis of N-1 Acidic Functionality Affording Analogues with Enhanced Antiviral Activity Against HIV", BIOORG. MED. CHEM., vol. 12, 2002, pages 3001 - 3004 *

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101885721A (zh) * 2010-06-11 2010-11-17 银杏树药业(苏州)有限公司 作为ccr5拮抗剂的吡咯衍生物、制备该化合物的方法及其应用
US8658797B2 (en) 2011-02-25 2014-02-25 Helsinn Healthcare Sa Asymmetric ureas and medical uses thereof
US9751836B2 (en) 2011-02-25 2017-09-05 Helsinn Healthcare Sa Asymmetric ureas and medical uses thereof
US10407390B2 (en) 2011-02-25 2019-09-10 Helsinn Healthcare Sa Asymmetric ureas and medical uses thereof
WO2012143526A1 (fr) * 2011-04-20 2012-10-26 Centre National De La Recherche Scientifique (C.N.R.S) 1,2,3-triazoles 1,4-disubstituées, leurs procédés de préparation et leurs utilisations diagnostiques et thérapeutiques
FR2974365A1 (fr) * 2011-04-20 2012-10-26 Centre Nat Rech Scient 1,2,3-triazoles 1,4-disubstituees, leurs procedes de preparation et leurs utilisations diagnostiques et therapeutiques
JP2014515754A (ja) * 2011-04-20 2014-07-03 サントル ナスィオナル ド ラ ルシェルシュ スィアンティフィク(セ.エン.エル.エス.) 1,4−二置換1,2,3−トリアゾール、その調製方法、ならびにその診断的および治療的使用
US10059704B2 (en) 2011-04-20 2018-08-28 Centre National De La Recherche Scientifique (C.N.R.S.) 1,4-disubstituted 1,2,3-triazoles, methods for preparing same, and diagnostic and therapeutic uses thereof
US10501479B2 (en) 2016-03-22 2019-12-10 Helsinn Healthcare Sa Benzenesulfonyl-asymmetric ureas and medical uses thereof

Similar Documents

Publication Publication Date Title
WO2009089659A1 (fr) Composés à base de pyrollidine
EP3675848B1 (fr) Composés spirocycliques et leurs procédés de préparation et d&#39;utilisation
EP2914594B1 (fr) Imidazo[1,2-a]pyridincarboxamides amino-substitués et leur utilisation
EP1492789B1 (fr) Derives de tropane utilises comme modulateur de ccr5
ES2381686T3 (es) Derivados de alfa-aminocarboxamida cuaternarios como moduladores de canales de sodio con apertura por voltaje
JP6759110B2 (ja) 新規な化合物
US20080096861A1 (en) Chemical Compounds
JP2017504571A (ja) 置換ベンズアミド及びその使用方法
WO2002004402A1 (fr) Derives d&#39;ester
US8476290B2 (en) Compound having spiro-bonded cyclic group and use thereof
IL208215A (en) Hydroxymethyl pyrrolidines, their pharmaceutical composition and their use in the preparation of a drug
AU2005262330A1 (en) Piperidine derivatives as NK1 antagonists
JP2008510006A (ja) 化合物
RU2744784C1 (ru) Бензоксазин-оксазолидиноновое соединение, замещенное азотсодержащим гетероциклом, способ его получения и применение
EP3138841A1 (fr) Antagonistes d2, procede de synthese et procedes d&#39;utilisation
KR20180016485A (ko) 무스카린성 m2 수용체의 양성 알로스테릭 조정제
US7579471B2 (en) Tropane derivatives useful in therapy
IL310377A (en) A heterocyclic substituted aryl ether compound as a GLP1R agonist
WO2009092293A1 (fr) Composés à base de pyrollidine
EP3743419B1 (fr) Nouveaux composés pour le traitement d&#39;infections parasitaires
CA3226724A1 (fr) Cyanopyridine et cyanopyrimidine utilisees en tant qu&#39;agents de degradation de bcl6
WO2024035688A1 (fr) Agents de dégradation de bcl6 macrocycliques
TW202227404A (zh) 可用於治療與5-ht2a血清素受體相關疾病之作為5-ht2a血清素受體調節劑之異噁唑衍生物
CN116322692A (zh) 自分泌运动因子抑制剂化合物
CN101990536A (zh) 基于吡咯烷的化合物

Legal Events

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

Ref document number: 200980110077.1

Country of ref document: CN

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

Ref document number: 09703729

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09703729

Country of ref document: EP

Kind code of ref document: A1